Sie sind auf Seite 1von 23

PRIMeR

Immune-​mediated neuropathies
Bernd C. Kieseier1*, Emily K. Mathey2, Claudia Sommer3 and Hans-​Peter Hartung1
Abstract | Since the discovery of an acute monophasic paralysis, later coined Guillain–Barré
syndrome, almost 100 years ago, and the discovery of chronic, steroid-​responsive polyneuropathy
50 years ago, the spectrum of immune-​mediated polyneuropathies has broadened, with various
subtypes continuing to be identified, including chronic inflammatory demyelinating poly­
radiculoneuropathy (CIDP) and multifocal motor neuropathy (MMN). In general, these disorders
are speculated to be caused by autoimmunity to proteins located at the node of Ranvier or
components of myelin of peripheral nerves, although disease-​associated autoantibodies have not
been identified for all disorders. Owing to the numerous subtypes of the immune-​mediated
neuropathies, making the right diagnosis in daily clinical practice is complicated. Moreover,
treating these disorders, particularly their chronic variants, such as CIDP and MMN, poses a
challenge. In general, management of these disorders includes immunotherapies, such as
corticosteroids, intravenous immunoglobulin or plasma exchange. Improvements in clinical
criteria and the emergence of more disease-​specific immunotherapies should broaden the
therapeutic options for these disabling diseases.

Polyneuropathies are common neurological disorders supportive evidence, including the detection of autoanti­
that can be caused by immune responses against auto­ bodies for some disorders. Several therapies are available
antigens in the peripheral nervous system (PNS). for management of these disorders, including plasma
Immune-​mediated neuropathies can include acute dis­ exchange and intravenous immunoglobulin (IVIG),
orders, such as Guillain–Barré syndrome (GBS)1, which although the specific treatment varies between disorders3.
has several subtypes, including demyelinating (acute In this Primer, we consider the clinical features of
inflammatory demyelinating polyradiculoneuropathy acute and chronic immune-​mediated neuropathies and
(AIDP)) and axonal (acute motor axonal neuropathy discuss how these features can be used to differentiate
(AMAN)) subtypes, or chronic disorders, such as chronic these conditions from other neurological disorders. We
inflammatory demyelinating polyradiculoneuropathy also summarize the current understanding, diagnosis and
(CIDP), multifocal motor neuropathy (MMN) and neuro­ treatment recommendations for these disorders. Given
pathies associated with an immunoglobulin M (IgM) the large spectrum of clinical subtypes, we focus on GBS,
monoclonal gammopathy of undetermined signifi­ CIDP, MMN and IgM MGUS polyneuropathies on
cance (MGUS)2. Other immune-​mediated neuropathies the basis of their overall prevalence and the deeper
have been identified, many of which are considered as understanding of these disorders than with other, less
further subtypes of GBS and CIDP, although many of frequent subtypes. We do not cover other forms of neuro­
these disorders are quite rare (Box 1). In general, periph­ pathy such as vasculitic neuropathies or POEMS (poly­
eral neuropathies are characterized by progressive muscle neuropathy, organomegaly, endocrinopathy, monoclonal
1
Department of Neurology,
weakness and are often accompanied by sensory deficits. protein, skin changes) syndrome, both of which are asso­
Medical Faculty, Heinrich
Heine University, Düsseldorf,
Although immune-​mediated neuropathies are fairly ciated with a cluster of multisystem clinical features and,
Germany. rare, they confer substantial social and economic costs as such, diagnosis relies on a broader clinical spectrum
2
Neuroinflammation and affect patients' quality of life (QOL). GBS is con­ beyond the features of a neuropathy. In addition, infec­
Laboratory, Brain and Mind sidered a neurological emergency, and patients require tious neuropathies are also excluded because this Primer
Centre, University of Sydney, hospitalization and intensive care management, and are focuses on autoimmune-​driven polyneuropathies.
Sydney, Australia.
often left with long-​term disabilities, whereas chronic
3
Department of Neurology, immune-​mediated neuropathies are typically treated Epidemiology
University of Würzburg,
Würzburg, Germany.
using expensive immunotherapies over the course of the GBS
disease. Thus, making the right diagnosis is crucial to pre­ GBS has an incidence of 0.81–1.89 cases per 100,000
*e-​mail: bernd.kieseier@
uni-duesseldorf.de vent unnecessary damage to the PNS and to identify the individuals and is the most common cause of acute,
https://doi.org/10.1038/ correct treatment option. In general, diagnosis is based on flaccid paralysis worldwide4. No major geographical
s41572-018-0027-2 clinical manifestations, electrodiagnostic studies and other variation has been reported in the incidence of GBS5,

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 1

0123456789();
Primer

Box 1 | Immune-​mediated neuropathies usually an upper respiratory tract infection (~40% of


cases) or gastrointestinal infection (20% of cases)15. The
Guillain–Barré syndrome and variants most commonly associated pathogens are Campylobacter
• Acute inflammatory demyelinating polyradiculoneuropathy jejuni (particularly for AMAN), Haemophilus influenzae,
• Acute motor and sensory axonal neuropathy Mycoplasma pneumoniae16, cytomegalovirus (CMV),
• Acute motor axonal neuropathy Epstein–Barr virus and Zika virus17. The 2013–2014
• Acute sensory neuronopathy outbreak of Zika virus in Latin America and the Pacific
• Acute pandysautonomia Islands coincided with a 2–9.8-fold increase in the inci­
dence of GBS compared with the incidence before the
Regional variants:
Zika outbreak17. Although a spectrum of clinical mani­
• Miller Fisher syndrome
festations is associated with Zika-​virus-associated GBS,
• Pharyngeal–cervical–brachial variant AIDP is the most frequent18.
Overlap: An association between GBS and vaccination has
• Miller Fisher/Guillain–Barré overlap syndrome been demonstrated only with influenza vaccination (rel­
Chronic inflammatory demyelinating polyradiculoneuropathy and variantsa ative risk 1.41)19; however, the risk of GBS after influenza
infection is ~4–7-fold higher than the risk after influ­
• Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP)
-- Pure motor CIDP enza vaccination, suggesting that the risk–benefit ratio is
-- Pure sensory CIDP in favour of vaccination20. Some types of surgery, includ­
• Demyelinating neuropathies with monoclonal gammopathy of undetermined ing cardiovascular, gastrointestinal and orthopaedic sur­
significance other than IgM geries, have been associated with an increased incidence
• Chronic inflammatory axonal polyneuropathy of GBS21. In addition, immune-​modulating drugs might
precipitate GBS; for example, the triggering of GBS by
• Distal acquired demyelinating symmetric neuropathy
tumour necrosis factor (TNF) antagonists has been
• Multifocal acquired demyelinating sensory and motor neuropathy (also known as
described in isolated cases22. Despite several reports of
Lewis–Sumner syndrome)
familial GBS, no significant association between genetic
Chronic demyelinating neuropathies distinct from CIDPa alterations and GBS predisposition has been demon­
• Multifocal motor neuropathy strated. Several candidate genes that have a role in the
-- Multifocal motor neuropathy without conduction block innate immune system have been investigated, but no
• Demyelinating neuropathies associated with immunoglobulin M (IgM) monoclonal significant associations were found23. One exception is
gammopathy of undetermined significance with and without anti-​myelin-associated a case series of a GBS-​like disease in individuals with a
glycoprotein neuropathies genetic deficit in protection from complement-​mediated
• Autoimmune neuropathies associated with antibodies to paranodal antibodies injury (the p.Cys89Tyr mutation in CD59) and a clinical
(paranodopathies) response to a complement inhibitor24.
• Chronic ataxic neuropathy with ophthalmoplegia, monoclonal protein, cold
agglutinins and disialosyl antibodies Chronic inflammatory neuropathies
• Chronic ataxic neuropathy with disialosyl antibodies CIDP is the most common immune-​mediated neuro­
pathy, with a reported prevalence ranging from
a
Collectively, these disorders can be referred to as chronic inflammatory neuropathies. 0.8–8.9 cases per 100,000 individuals from data in eight
countries25. Whether the prevalence differs between
although incidence might be higher in tropical coun­ countries has not been ascertained owing to a lack of
tries6. However, substantial regional variation in the systematically collected data. The average age at onset
prevalence of the AMAN and AIDP subtypes of GBS of CIDP is 48 years of age, and CIDP predominantly
has been reported. AIDP is the main subtype of GBS in affects men more than women with a ratio of approx­
North America, Europe and Australia and accounts imately 2:1 (ref.2). Risk factors for CIDP are not known.
for 60–80% of GBS cases, whereas AMAN accounts for Antecedent infections can occur but are rarer in patients
30–65% of GBS cases in Asia and in Central and South with CIDP (4–30%) than in those with GBS. In contrast
America7,8. The causes of the regional differences are to GBS, which has a strong association with C. jejuni
unclear but could relate to geographical differences infections, no particular pathogens associated with
such as temperature and humidity, genetic susceptibil­ CIDP have been widely identified2.
ity, antecedent infection9 or underdiagnosis of AMAN10. Population-​b ased studies of MMN are rare, but
Miller Fisher syndrome (MFS), another subtype of GBS, several studies have estimated different prevalences
accounts for 5% of GBS cases in the Western hemisphere between countries, such as a prevalence of 0.29 cases per
and up to 26% in Japan11. GBS is more common in men 100,000 individuals in Japan, 0.6 cases per 100,000 indi­
than in women, with a ratio of 3:2 (ref.12), and occurs viduals in the Netherlands and 0.53 cases per 100,000
at any age but with a median age of onset of 40 years of individuals in southeast England25–27. The age of onset of
age; incidence increases to 3.3 cases per 100,000 indi­ MMN ranges from 20 to 70 years of age, with a median
viduals in those >50 years of age5,13. Overall, GBS has a onset at 40 years of age. MMN is more common in males
recurrence rate of 5%14, although younger patients have than in females, with a ratio of at least 2:1 (ref.26). The
a higher risk of recurrence than older individuals14. cause of MMN is currently unknown, and no associated
Measures to prevent recurrence are not known. antecedent infections have been reported3.
In 60–70% of cases, the first symptoms of GBS mani­ Paraproteinaemic demyelinating neuropathies are
fest between 1 and 3 weeks after an acute infection, CIDP variants that often fulfil the diagnostic criteria for

2 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

CIDP but are associated with a paraprotein, typically neuropathies, such as cellular infiltration of the periph­
IgM. More than 50% of patients with an IgM para­protein eral nerve, a monophasic clinical course with motor
have anti-​myelin-associated glycoprotein (MAG) IgM deficits and electrophysiological changes, but do not
antibodies28. Paraproteinaemic demyelinating neuro­ exactly recapitulate individual disorders. These mod­
pathies have a prevalence of 1.04–5.1 cases per 100,000 els are often used to provide proof of concepts and are
individuals25,29. Paraproteinaemias are a group of dis­ frequently developed to mimic different aspects of the
orders in which monoclonal plasma cells proliferate disease process; for example, animal models that mimic
(owing to an underlying malignancy, such as multiple the chronicity of CIDP have recently been developed34,35.
myeloma, or the myeloma precursor syndrome MGUS)
and secrete monoclonal proteins (M proteins). The GBS
prevalence of IgM MGUS increases with age, from 3% GBS subtypes are distinguished by their clinical pres­
of individuals >50 years of age to 7.5% of individuals entation, electrophysiology, pathology and a distinct
>85 years of age, with up to 30% developing a neuropathy, underlying pathophysiology. As the demyelinating
and IgM MGUS is the most common paraproteinaemic form, AIDP, and the axonal form, AMAN, are the most
demyelinating neuropathy30. commonly occurring subtypes, they will be discussed in
Notably, most epidemiological studies and clinical more detail here.
trials on chronic inflammatory neuropathies include Substantial advances have been made in identifying
only patients who meet the diagnostic criteria of the pathogenetic autoantibody responses against periph­
European Federation of Neurological Societies and eral nerve targets in some subtypes of GBS, particularly
Peripheral Nerve Society (EFNS/PNS). Owing to this AMAN. Between 30–66% of patients with AMAN had
stringency, the prevalence of these disorders might be a prior infection with C. jejuni36,37, which can result in
underestimated and might not account for patients with an aberrant antibody cross reactivity between lipo-​
atypical phenotypes of the disorder. oligosaccharides (LOSs) on the bacterium and gan­
gliosides on axons — a process known as molecular
Mechanisms/pathophysiology mimicry. However, <1 in 1,000 individuals with a symp­
Immune-​mediated neuropathies are generally con­ tomatic C. jejuni infection are estimated to subsequently
sidered to be immune-​mediated disorders in which develop AMAN38 owing to a combination of host and
humoral and cellular immune responses act synergisti­ microbial factors that determine the production of anti-​
cally to cause nerve damage and neurological dysfunc­ ganglioside antibodies (Table 1). One implicated micro­
tion. Supporting an immune-​mediated aetiology was the bial factor is the sialylation of LOSs, which is determined
identification of autoreactive antibodies and T cells in by the presence of LOS loci that harbour genes involved
sural nerve biopsy samples and in the serum of patients31. in sialylation and that are found in certain stains of
Many of these autoantibodies bind to glycolipids located C. jejuni39; however, infection with one of these strains
in Schwann cells or axonal membranes, or proteins does not always lead to GBS, therefore other factors
located at the node of Ranvier. Although pathogenetic are also involved in the development of disease after
mechanisms that are common to several immune-​ C. jejuni infection. Many studies have attempted to iden­
mediated neuropathies have been identified (Fig.  1), tify host genes that confer susceptibility to GBS after
the precise immunopathogenesis for individual neuro­ infection, although the precise genetic factors have not
pathies — such as how the disease is triggered, or the yet been determined.
identity of specific autoantigens — has yet to be fully Autoantibodies to various gangliosides found in
elucidated. Mechanisms common to several immune-​ axons are used to aid diagnosis of certain subtypes
mediated neuropathies include a peripheral immune of GBS, such as AMAN, acute motor and sensory
response, consisting of the generation of autoantibod­ axonal neuropathy (AMSAN), MFS and pharyngeal–
ies directed towards components of myelin or proteins cervical–brachial variants, and have an accepted role
located at the node of Ranvier, and the breakdown of the in causing nerve injury (Table 1; Fig. 2). In addition, the
blood–nerve barrier owing to secreted chemokines and distribution of the ganglioside in the nerve influences
proteases from activated T cells. Leukocytes and auto­ the clinical phenotype of GBS. For example, GM1 and
antibodies can penetrate the nerve compartment facilita­ GalNAc-​GD1a are expressed predominantly on the
ted by the breakdown of the blood–nerve barrier, where axolemma of motor nerves, particularly at the node of
they can lead to myelin destruction or axonal damage. Ranvier40, and anti-GM1 and anti-​GalNAc0GD1a anti­
Many of the clinical and pathological changes bodies are associated with motor phenotypes. GQ1b
observed in inflammatory neuropathies can be mod­ is enriched in cranial nerves that innervate extraocu­
elled using experimental autoimmune neuritis (EAN) lar muscles, resulting in the ophthalmoplegia seen in
in animals32. In the EAN model, genetically susceptible patients with MFS.
animals, usually inbred mice or rats, are immunized The functional effects of anti-​ganglioside antibodies
with peripheral nerve homogenate, myelin or myelin in GBS have been demonstrated in animal models
components (such as myelin protein P0 and myelin P2 and in nerve biopsy samples. Anti-​GM1 (ref.41) and anti-​
protein or peptides, or lipids such as galactocerebroside) GD1a42 antibodies bind to the nodes of Ranvier, where
or receive adoptive transfer of autoreactive, myelin P2 they disrupt the fine structural arrangements respon­
protein or myelin protein P0 peptide-​specific T cell lines sible for sodium channel clustering, leading to lack of
to induce EAN33. These experimental models resemble sodium channel clustering, slowed axonal conduction
certain aspects of the pathogenesis of inflammatory and loss of function, or they bind to the motor nerve

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 3

0123456789();
Primer

Juxtaparanode Paranode Node


BNB Systemic immune compartment

APC Activated
T cell
Ganglioside

Autoreactive Gliomedin
T cell
NrCAM
Plasma cell NF155
MAG
Chemokines, proteases
and adhesion molecules Autoantibody
Kv1.1
Kv1.2 Contactin 1 CASPR1 Nav1.6 Kv7.2
Kv1.4 NF186 Kv7.3
Macrophage
Kv1.6
Apoptosis
Peripheral nervous system

Nerve
cell
TNF, Axon
proteases, Myelin Schwann cell
ROS and microvilli
cytokines

Schwann
cell

Fig. 1 | Pathomechanisms of immune-​mediated neuropathies. Immune-​mediated neuropathies can be triggered by


certain minor infections, which can activate and induce proliferation of autoreactive T cells, owing to a similar structure of
bacterial epitopes and peripheral nerve antigens (molecular mimicry). Autoreactive T cells secrete various cytokines that
stimulate the production of autoantibodies by plasma cells, which can enter peripheral nerves following damage to the
blood–nerve barrier (BNB). Activated T cells cross the BNB through interactions between adhesion molecules on T cells
and the vascular endothelium of endoneurial blood vessels. These interactions allow the adherence of T cells to the blood
vessel wall and cross the BNB by either transcellular or intercellular diapedesis. During this process, activated T cells
secrete metalloproteinases that degrade the basement membrane, which facilitates the possibility for larger molecules as
well as immune cells to cross the BNB. In the peripheral nerve, T cells secrete pro-​inflammatory cytokines, such as tumour
necrosis factor (TNF) and IL-2 (refs248,249), that contribute to the amplification and perpetuation of inflammation within the
nerve by the recruitment of additional immune cells from the periphery as well as by clonal expansion in situ. After the BNB
has become permeable owing to the infiltration of inflammatory cells, soluble factors in serum, such as autoantibodies and
complement, can access the endoneurium of the nerve. Activated macrophages can release mediators, such as pro-​
inflammatory cytokines, reactive oxygen species (ROS) and proteases, that can propagate the inflammatory response and
directly damage Schwann cells and axons. Upon entry to the nerve, autoantibodies can bind to myelin glycoproteins or
proteins at the node of Ranvier or paranodal regions (inset). Several proteins exist at the node and paranodal regions and
have diverse functions, such as maintaining axon–Schwann cell or axon–myelin binding (such as contactin-​associated
protein 1 (CASPR1), neurofascin 155 (NF155) or contactin 1), sodium channel clustering (such as gliomedin) and governing
node organization. Autoantibody binding to these proteins can lead to complement activation, macrophage-​mediated
axonal degeneration or demyelination and disruption to nodal architecture. Long-​term disability is mainly determined
by the degree of axonal degeneration. The immune response is switched off by programmed cell death (apoptosis),
although the mechanisms underlying this process are unknown. APC, antigen-​presenting cell; MAG, myelin-​associated
glycoprotein; NrCAM, neuronal cell adhesion molecule. Inset adapted from ref.80, Springer Nature Limited.

terminals, causing degeneration of the presynaptic nerve myelination45. Autopsy studies of nerves from patients
terminal in model systems43 (Fig.  3). The effect of the with AMAN have demonstrated IgG and complement
bound antibody is largely mediated by complement deposition at the node of Ranvier in large motor fibres
fixation, resulting in formation of the membrane attack and have revealed infiltrating macrophages penetrating
complex, although activation of Fcγ receptors on infil­ the internodal periaxonal space46. Nodal lengthening
trating phagocytotic macrophages by antibody–antigen is followed by axonal degeneration, but demyelination
immune complexes has also been shown to play a role and lymphocyte infiltration are limited47. To summarize,
in macrophage activation, the release of additional toxic conceptionally, in AMAN, antibody binding to axonal
mediators and the phagocytosis of myelin and myelin target antigens located along the node of Ranvier induces
debris44. Similarly, anti-​disialosyl antibodies (which paranodal myelin detachment, with node lengthening,
bind to an epitope common to many gangliosides, sodium channel disruption and altered ion and water
including GD1b, GD3, GT1b and GQ1b) bind to the homeostasis, which ultimately, if not stopped, progresses
nodal axolemma in human induced pluripotent stem to axonal degeneration48. Clinically this pathology might
cell (iPSC)-derived myelinating neuron–Schwann cell manifest as a continuum on the basis of the relative
co-​cultures, resulting in complement-​dependent axonal presence of a nerve conduction block, which might
degeneration or complement-​independent inhibition of revert rapidly, and axonal degeneration49. Indeed, nerve

4 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

conduction block can be assessed using electrophysiol­ surface of the Schwann cell, but not on the compact
ogy as part of the diagnostic work-​up of patients with myelin, has been observed in sural nerve biopsy sam­
suspected AMAN. Most importantly, this concept of a ples from three patients with AIDP46. On the basis of
‘nodopathy’, although initially described in AMAN47, these data combined with data from the EAN model,
has also been observed in chronic immune-​mediated antibody binding in these specific regions is speculated
neuropathies, such as CIDP and MMN. to fix complement, most likely derived from serum
Mechanisms of molecular mimicry in AIDP have complement proteins or produced by infiltrating macro­
not been established despite the high incidence of ante­ phages, which then leads to vesiculation of myelin and
cedent infection in these patients. Extensive searches macrophage-mediated demyelination. Serological studies
for anti-​ganglioside antibodies have resulted in incon­ using indirect immunofluorescence to screen for anti­
sistent reports of their presence and/or relevance in body targets found that 24% of patient sera bound to
AIDP35,36,50,51. These discrepancies might be due to the proliferating, non-​myelinating Schwann cells in vitro59
incorrect diagnosis of AIDP (patients with AMAN could and 44% of sera from patients with AIDP bound to the
be misclassified as AIDP when diagnosis has been made node or paranodal region, as shown by indirect fluores­
without assessment of nerve conduction)52 or the vari­ cence on teased nerve fibres60, suggesting the presence
ous methods used to detect anti-​ganglioside antibodies. of as-​yet unidentified targets at these sites in patients
However, patients with GBS that have a demyelinating with AIDP. One target of patient sera has been identi­
phenotype when assessed using nerve conduction stud­ fied as contactin-​associated protein 1 (CASPR1), which
ies (NCS) and who harbour both anti-​ganglioside and is located at the paranodal region61 (Fig. 1). Indeed, anti-​
anti-​galactocerebroside antibodies have been reported53. CASPR1 IgG3 antibodies were detected in the serum of a
Nevertheless, the role of autoantibodies in AIDP patient with GBS, and this response was associated with
remains unclear, and no consistent autoantibody corre­ severe neuropathic pain61.
lations have been reported in patients54 despite extensive
screening of major myelin antigens such as myelin pro­ CIDP
tein P0, myelin P2 protein and peripheral myelin protein The clinical heterogeneity of CIDP suggests that dis­
22 (PMP22)55,56. Moesin, which is expressed on Schwann tinct immunopathological mechanisms, such as dif­
cell microvilli, has been suggested as a potential auto­ ferent patterns of immune responses or specificities
antigen in CMV-​related AIDP57; however, this obser­ of the antigenic targets, underlie the different subtypes of
vation was not confirmed in other studies58, although the disorder3 (Fig.  3). The pathogenesis of CIDP has
different methods of antibody detection were used. Some largely been determined through the immunopatho­
data suggest that the autoantibody response in AIDP logical study of nerve biopsy samples from patients.
is directed towards the Schwann cell surface. Indeed, These studies have demonstrated inflammatory cells
complement C3d and C5b-9 deposition on the outer such as T cells and macrophages that have infiltrated the

Table 1 | Autoantibodies with clinical implications or associations in immune-​mediated neuropathies


Disease Subgroup and/or Antigens Antibody Clinical Refs
phenotype isotype implications
Acute motor axonal Pure motor GM1, GM1b, IgG Guide to GBS 253,254

neuropathy GD1a and subtype


GalNAc-​GD1a
Acute motor and sensory Motor and sensory GM1 and GD1a IgG 255

axonal neuropathy
Miller Fisher syndrome Ataxia, ophthalmoplegia GQ1b and GT1a IgG 225,226

and areflexia
Pharyngeal–cervical– Oropharyngeal, facial and GT1a, GQ1b and IgG 256

brachial variant of GBS neck and shoulder muscle GD1a


weakness
Multifocal motor Motor GM1 and complex IgM Diagnostic guide 257

neuropathy GM1:GalC
Anti-​MAG neuropathy Sensory and ataxia MAG IgM 102

CANOMAD and CANDA Sensory and ataxia GD3, GD1b, GT1b IgM 167,258

and GQ1b
Chronic inflammatory Aggressive onset and initial Contactin 1 IgG4 Poor response to 79,259,260

demyelinating axonal involvement IVIG


polyradiculoneuropathy
Tremor, ataxia and distal NF155 IgG4 243,244,261

motor involvement
Subacute onset, sensory NF186 and NF140 IgG4 Good response to 87

ataxia and conduction block IVIG and steroids


CANDA, chronic ataxic neuropathy with disialosyl antibodies; CANOMAD, chronic ataxic neuropathy with ophthalmoplegia,
monoclonal proteins, cold agglutinins and disialosyl antibodies; GBS, Guillain–Barré syndrome; Ig, immunoglobulin; IVIG,
intravenous immunoglobulin; MAG, myelin-​associated glycoprotein; NF, neurofascin.

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 5

0123456789();
Primer

GM1 Cer GD1A Cer GT1A Cer Asialo-GM1 Cer

GM1b Cer GD1B Cer GT1b Cer LM1 Cer

GM2 Cer GD3 Cer GQ1b Cer SGPG Cer


SO4

Galactose Glucose N-acetyl galactosamine N-acetyl glucosamine Neuraminic acid Glucuronic acid Cer Ceramide

Fig. 2 | Structure of glycolipids to which antibody responses have been detected in immune-​mediated neuropathies.
Serum antibodies against GM1a, GM1b, GD1a and GalNAc-​GD1a are frequently found in patients with acute motor axonal
neuropathy250, whereas the Miller Fisher syndrome and Bickerstaff brainstem encephalitis variants are associated with
anti-​GQ1b antibodies251. SGPG, sulfated glucuronyl paragloboside.

nerve through the perivascular space or are dispersed systems might be dysfunctional or diminished in CIDP70.
throughout the perineurium and endoneurium62. These Indeed, CD4+CD25highFOXP3+ regulatory T cells from
inflammatory cells likely contribute to breakdown of the patients with CIDP have a reduced ability to suppress
blood–nerve barrier. Indeed, disruption of the blood– proliferative responses of pro-​inflammatory T cells com­
nerve barrier has been observed in CIDP and can be pared with regulatory cells from healthy controls71. CD8+
visualized using MRI as gadolinium enhancement at T cells have also been observed in nerve biopsy sam­
demyelinative foci in the nerve trunks63 and spinal nerve ples and the peripheral blood of patients with CIDP72,73.
roots and plexuses64. Most of these pro-​inflammatory Enrichment of these CD8+ T cell clones within the nerve
cells cluster around endoneurial blood vessels and are indicates that the expansion is likely to be antigen driven,
predominantly macrophages with fewer CD8+ and CD4+ although the antigen responsible is yet to be identified.
T cells62. In CIDP, resident and recruited macrophages CD8+ T cells might have a role in damage of periph­
act as antigen-​presenting cells that perpetuate the eral nerve components in patients with CIDP, such as
immune response within the nerve and act as the end-​ Schwann cells. Supporting this role of CD8+ T cells is
stage effector cells of demyelination65. Ultrastructural the upregulation of MHC class I molecules on Schwann
studies of nerve biopsy samples from patients with CIDP cells74,75. Analysis of the T cell repertoire in patients with
have demonstrated macrophages that extend their pro­ CIDP with and without IVIG treatment found a higher
cesses between the myelin lamellae, resulting in splitting number of oligoclonal expansions of CD8+ T cells than
and destruction of the myelin66. of CD4+ T cells, which was reduced after treatment with
Activated CD4+ T cells67, including pro-​inflammatory IVIG76. This broad activation of CD8+ T cells suggests
T helper 17 (TH17) and TH1 cells68, are increased in the a crucial role for this cell type in the pathogenesis of
blood of patients with CIDP. These increases point to CIDP and that multiple peptides might stimulate the
a potential pathogenetic role for these T cell subsets in antigen-​driven response.
contributing to blood–nerve barrier breakdown owing to Humoral immune mechanisms, such as antibod­
their release of pro-​inflammatory cytokines and various ies, complement or cytotoxic inflammatory molecules,
proteinases that affect blood–nerve barrier function and might also have a role in CIDP. Supporting this hypoth­
amplification of the inflammatory response via release esis is the rapid improvement of some patients after
of chemotactic molecules and inflammatory cytokines. plasma exchange and the presence of immunoglobulin
Moreover, differences in immune activation, such as and complement on myelin and Schwann cells in sural
T cell responses, could partly explain the clinical hetero­ nerve biopsy samples from patients77. In addition, the
geneity of CIDP. For example, autoreactive myelin-​ serum of patients with CIDP who were responsive to
specific T cell responses and memory T cell subsets, plasma exchange resulted in demyelination and nerve
CD4+ effector memory T cells and CD4+ central mem­ conduction block in rats, suggesting a role for autoreac­
ory T cells are increased in the blood of patients with tive antibodies in CIDP pathophysiology78. Despite these
atypical CIDP variants such as distal acquired demyelin­ findings, the role of myelin proteins as autoantigens in
ating symmetric neuropathy (DADS), multifocal acqui­ CIDP has not been confirmed. Autoantibody responses
red demyelinating sensory and motor neuropathy to myelin P2 protein, myelin protein P0, PMP22 and
(MADSAM; also called Lewis–Sumner syndrome) and connexin have been reported in some studies, but other
pure motor or sensory CIDP compared with those with studies did not detect these responses77. In addition to
typical CIDP69. In addition, cellular immunoregulatory these myelin proteins, other potential autoantigens in

6 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

Schwann cell
Paranodal detachment microvilli Demyelination

Unidentified
Myelin antigen
Macrophage
Complement
Autoantibody

Axonal damage
AMAN Glycolipid Ca2+ AIDP

CIDP MMN
CASPR1
Axonal damage
NF186 NrCAM
Contactin 1 Gliomedin Membrane IgM
attack complex
NF155 Unidentified
antigen

Paranodal detachment
T cell
Demyelination

Fig. 3 | The potential role of autoantibodies in immune-​mediated neuropathies. Autoantibodies can cause axonal
damage or demyelination via multiple pathways. In acute motor axonal neuropathy (AMAN), autoantibodies can induce
the formation of the terminal complement complex (also known as the membrane attack complex), which damages the
axon and drives macrophage activation, including macrophage penetration of the internodal periaxonal space;
in addition, paranodal detachment, nodal lengthening, sodium channel clustering and alterations in ion and water
homeostasis can be found. In acute inflammatory demyelinating polyradiculoneuropathy (AIDP) as well as chronic
inflammatory demyelinating polyradiculoneuropathy (CIDP), autoantibodies induce demyelination, which involves
complement and macrophage activation, although the target antigens are better understood in CIDP. In addition, the role
of CD8+ lymphocytes damaging the peripheral nerve has recently been described in greater detail. The in situ activation of
CD8+ lymphocytes might indirectly be co-​mediated via macrophage activation through autoantibodies. In multifocal
motor neuropathy (MMN), autoantibodies facilitate axonal damage and the disruption of sodium channel clustering252.
CASPR1, contactin-​associated protein 1; NF, neurofascin; NrCAM, neuronal cell adhesion molecule. Adapted from
refs1,80,154, Springer Nature Limited.

CIDP include proteins located in non-​compact myelin of the myelinated axon into the node, paranode and
and the axoglial junctions at the node of Ranvier, para­ internode. Compartmentalization is required for sal­
node and juxtaparanode61,77,79,80; disturbances of these pro­ tatory conduction as it maintains segregation of the
teins can result in impairment of conduction and nerve voltage-​gated sodium and potassium channels involved
conduction block81. Supporting the role of these proteins, in the transmission of action potentials. Disruption or
irregularities in the paranodal loops, including vacuola­ disorganization of these regions can result in slowed
tion at the paranodes in the Schwann cell cytoplasm and or blocked nerve conduction. In addition, the passive
abnormal expression and distribution of certain axoglial transfer of anti-​pan-neurofascin and anti-​gliomedin
proteins, such as diffuse rather than compartmental­ antibodies exacerbates EAN and causes a decrease in
ized expression of the paranodal protein CASPR1, have the clustering of nodal proteins and disorganization
been demonstrated in nerve biopsy samples from patients of the nodes of Ranvier77. Nerve biopsy samples from
with CIDP82. In addition, serum from patients with patients with anti-​NF155 or anti-​contactin 1 antibod­
CIDP binds either to the nodes of Ranvier or to the para­ ies provide evidence that this subgroup is distinct from
nodes of teased nerve fibres in 30% of cases60. Further those with typical CIDP in that the main pathological
studies identified some of the target antigens as the characteristic is paranodal detachment and loss of sep­
nodal proteins neurofascin 186 (NF186) and gliomedin, tate junctions at the paranode, without macrophage-​
and the paranodal proteins NF155 and contactin 1, mediated demyelination or onion bulbs (structures that
although the antigens are unknown in the remainder of are formed after repeated episodes of demyelination
cases77 (Table 1). In the EAN model, the nodal expres­ and remyelination, which result in concentric layers of
sion of NF186 and gliomedin is disrupted before the Schwann cells and fibroblasts around the nerve fibre)84,85.
onset of clinical signs and demyelination and coincides Serum antibodies to the peripheral nerve ganglioside
with the generation of autoantibodies against NF186 LM1 were detected in patients with CIDP and found to
and gliomedin83. These axoglial proteins at the node of be more frequently associated with ataxia, potentially
Ranvier play a pivotal role in the compartmentalization reflecting the anatomical distribution of LM1 in the

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 7

0123456789();
Primer

nervous system86. In addition, autoantibody responses IgM MGUS polyneuropathies


to the nodal proteins NF140 (refs61,87) and the para­ Approximately 50% of patients with IgM MGUS neuro­
nodal protein CASPR161 have also been demonstrated pathy have anti-​MAG autoantibodies, the first antigenic
in patients with CIDP, but only in a very small number target to be identified in immune-​mediated neuro­
of patients. Targeting CASPR1 could result in damag­ pathies102. Anti-​MAG antibodies can be detected in the
ing the axoglial junction, whereas NF140 is a neuronal serum of patients103 and can bind to myelin in nerve
neurofascin isoform that promotes the assembly of the sections 104. Nerve biopsy samples of patients with
node of Ranvier. anti-​MAG antibodies have demyelination and axonal
loss, and splitting of the outer layers of myelin can be
MMN observed in ultrastructural studies105. These areas of
The first descriptions of MMN reported the presence of damage are often colocalized with IgM and complement
anti-​GM1 IgM antibodies and responsiveness to IVIG as (C3d) depositions on the outer rim of the myelin104,106
features distinguishing the disorder from amyotrophic and on the terminal loops of myelin at the paranodes107.
lateral sclerosis (ALS)88. Anti-​GM1 IgM antibodies are Furthermore, a study of skin biopsy samples from
found in 43–85% of patients with MMN89 depending patients with anti-​MAG IgM MGUS neuropathy demon­
on the antibody detection method used. The patho­ strated anti-​MAG IgM accumulation within the nerve
genicity of these antibodies is unclear, although it is perineurium that was associated with axonal degen­
assumed that their effect is analogous to anti-​GM1 IgG eration and disruption of the axonal cytoskeleton108,
antibodies in AMAN, whereby they bind at the nodes although the precise mechanisms remain unclear. Serum
of Ranvier and activate complement, which disrupts from patients with anti-​MAG antibodies can cause
sodium channel clustering, leading to conduction block demyelination and widening of the myelin lamellae in a
in electrophysiological studies90 (Fig.  3). Serum from complement-​dependent manner when passively trans­
patients with MMN positive for anti-​GM1 IgM anti­ ferred into experimental models, in which antibodies
bodies can activate complement in vitro and in vivo91,92; in the serum bind to the outer surface of myelin109, the
anti-​GM1 IgM antibodies that have a high complement-​ nodes of Ranvier and Schmidt–Lanterman incisures,
activating capacity are associated with more severe a tube-like compartment of Schwann cell cytoplasm that
muscle weakness and axonal loss than anti-​GM1 IgM crosses the compact myelin and connects the abaxonal
antibodies with low complement-​activating capacity and adaxonal Schwann cell membranes110. Almost all
in patients with MMN93. Anti-​GM1 IgM from patients anti-MAG antibodies cross-​react with the peripheral
with MMN can also bind to motor neurons differenti­ nerve glycolipid sulfoglucuronyl glycosphingolipid
ated from human iPSCs, leading to disturbed calcium (SGPG)111. Unlike MAG, which is expressed in both the
homeostasis through both complement-​dependent and central nervous system (CNS) and PNS, SGPG is exclu­
complement-​independent mechanisms94, in addition sively expressed in peripheral nerves, making it a likely
to complement-​mediated axonal damage, which was target in peripheral neuropathy. Anti-​sulfatide, anti-​
abrogated in the presence of IVIG94. chondroitin sulfate C and anti-​ganglioside antibodies
Nerve biopsy samples from the site of conduction can also be found in patients with IgM MGUS30, which
block in patients with MMN have revealed axonal degen­ may have similar pathogenetic mechanisms.
eration and loss as well as frequent and prominent regen­
erating fibres95. Collateral reinnervation by surviving Diagnosis, screening and prevention
motor units compensates for axonal loss and preserves Patients with immune-​mediated neuropathies pres­
compound muscle action potentials (CMAPs)96. In this ent with diverse clinical manifestations. Patients with
study, axonal pathological changes predominated over GBS seek medical help because of rapidly evolving
myelin pathology; however, other studies have demon­ muscle weakness that can lead to respiratory muscle
strated demyelination and onion bulbs97. Intriguingly, fatigue and failure, whereas CIDP can have an insidious
infiltrating inflammatory cells are very rarely reported onset and patients can present late in the course of the
in biopsy samples from patients with MMN, posing the disease. The typical presentation of MMN is pure motor
question of how circulating anti-​GM1 IgM antibodies paresis of an upper limb muscle, but symptoms can start
access the endoneurium98. One possibility is that sera in the lower limbs in rare cases. The following diagnostic
from patients with MMN contains unidentified humoral procedures help to rapidly make the correct diagnosis
factors that lead to blood–nerve barrier dysfunction via and assign patients to the optimal treatment (Fig. 4).
an increase in vascular endothelial growth factor (VEGF)
secretion from endothelial cells or an unknown IgG that GBS
binds to endothelial cells99. Electrophysiological studies Clinical manifestations. GBS manifests as a rapidly
have suggested that nodal or paranodal dysfunction progressing muscle weakness and sensory disturbance
contributes to the conduction block observed in patients in arms, legs and, in some patients, facial, bulbar and
with MMN. An immune-​mediated sodium channel respiratory muscles. The typical clinical manifesta­
dysfunction has been hypothesized by some, in which tions of GBS (that is, whereby symptoms develop after
Na+/K+-ATPase pump function is potentially impeded by gastrointestinal or respiratory infections, followed by
antibody binding100,101. Owing to the implicated involve­ neuropathic back pain, distal paresthesias and progres­
ment of nodal regions in the pathophysiology of MMN, sive symmetrical flaccid paresis with a nadir within
it has been suggested that this disorder should also be 4 weeks, in the absence of bladder disturbance) are easily
classified as a nodopathy101. identifiable. Sensory deficits (such as hypesthesia and

8 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

Progressive or relapsing pareses with or without sensory deficit and reduced or no tendon reflexes

Duration <2 months Duration ≥2 months

Electrophysiological Electrophysiological and laboratory examination Other family members


examination (including CSF analysis) with similar symptoms

Electrophysiological and Electrophysiological and supportive


Electrophysiological
CSF criteria fulfilled; CSF criteria not fulfilled
criteria fulfilleda
other causes excluded

Signs of inflammation on MRI or


demyelination in biopsy samples
Consider GBS

Consider Consider Consider Consider hereditary


AMAN AIDP CIDP neuropathy

Fig. 4 | Diagnostic algorithm. A clinical duration of symptoms for ≥2 months is the clinical discriminator between Guillain–
Barré syndrome (GBS) and chronic inflammatory demyelinating polyradiculoneuropathy (CIDP). As part of the diagnostic
work-​up of patients, nerve conduction studies are carried out to examine for signs of demyelination and can be used to
identify patients with demyelinating diseases (such as CIDP or acute inflammatory demyelinating polyradiculoneuropathy
(AIDP)) or those with axonal pathologies (such as acute motor axonal neuropathy (AMAN)). Signs of demyelination in nerve
conduction studies include a partial conduction block, reduced motor nerve conduction velocity, prolonged distal latency
of the motor nerve and the absence of F-​waves or a prolonged F-​wave latency. In addition to these studies, laboratory tests
such as cell counts and the assessment of protein levels in cerebrospinal fluid (CSF) help to evaluate supportive criteria and
can rule out other causes of disease. For CIDP, pleocytosis of <10 cells μl–1 is a supporting criterion. If other causes of
neuropathy have been ruled out, and the electrophysiological and supportive criteria are fulfilled, patients can begin
term anti-​inflammatory and immunosuppressive therapies. In difficult cases, such as in patients with unfulfilled electro-
physiological and CSF criteria, MRI or nerve biopsy might be helpful to identify demyelinating lesions. aNote that
electrophysiological criteria for GBS might be negative during the first 1–2 weeks.

hypoalgesia) can vary; asymmetrical sensory deficits are and were subsequently revised115,116. The latest revision
not uncommon, and in rare cases, sensory deficits can under the Brighton criteria was developed in response to
have a pattern with a pseudo-​sensory level suggesting the 2009–2010 H1N1 swine flu vaccination campaign116;
myelopathy. Approximately 10% of patients with AMAN these criteria now enable clinical assessment to four
have preserved or even exaggerated tendon reflexes112, levels of diagnostic certainty.
which might raise doubt about the diagnosis. Patients High protein and albumin levels in the cerebrospinal
with AMAN more frequently have a preceding C. jejuni fluid (CSF) with normal cell counts (referred to as the
infection, have predominant motor neuropathy with albuminocytologic dissociation) is considered patho­
very little sensory involvement and less frequently have gnomonic for GBS in patients with the typical clinical
cranial nerve involvement compared with patients presentation. However, CSF protein and albumin levels
with AIDP. MFS, characterized by ophthalmoplegia, are normal in 50% of patients during the first week after
areflexia and ataxia, is clinically very distinctive. Other presentation16, which requires repeat lumbar puncture
subtypes of GBS include AMSAN, which manifests to ascertain elevated protein if there are doubts about
similar to AMAN but with more pronounced sensory the diagnosis. In addition, pleocytosis of 10–50 cells µl–1
involvement, in addition to a pharyngeal–cervical– is found in ~15% of patients with GBS, which may
brachial variant16,113, which manifests with rapidly pro­ confound diagnosis117.
gressive oropharyngeal and cervicobrachial weakness Clinical electrophysiology, particularly NCS, which
associated with areflexia in the upper limbs. The latter has can be used to assess for signs of demyelination or axonal
also been regarded as part of a continuum from GBS to damage, can help to support the diagnosis of GBS and
Bickerstaff encephalitis114. can identify AIDP and AMAN subtypes (Table 2). NCS
should be performed within the first week after pres­
Diagnostic work-​up. In a typical case of GBS that is entation to confirm the diagnosis; however, if nerve con­
preceded by an infection and by back pain, the diag­ duction is within normal ranges initially, NCS should
nosis is straightforward. However, several disorders be repeated >2 weeks after onset as abnormalities might
mimic the symptoms of GBS, and diagnosis can be peak at this time118. At least four motor nerves including
more difficult in atypical cases. Diagnostic criteria for F-​waves (late potentials yielding information on prox­
GBS were first developed by the US National Institute imal nerve and root segments, which may be affected
of Neurological Disorders and Stroke (NINDS) in 1978 early in this polyradiculoneuropathy) and three sensory

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 9

0123456789();
Primer

nerves should be investigated to increase the diagnostic disorders, such as rapidly progressive peripheral nerve
yield16. As AIDP is the most frequent type of GBS in vasculitis. In some patients, even in those without previ­
Europe and North America, detection of demyelination ous symptomatic infection, associated pathogens can be
is important to support the diagnosis, although to date isolated from stool or sputum samples, or a correspond­
no final consensus about which neurophysiological cri­ ing acute humoral response against these pathogens
teria are optimal for confirming the diagnosis of GBS can be detected in serum and CSF. The most common
and identifying subtypes has been reached52. The cur­ pathogens are C. jejuni (found in 20–30% of cases),
rent neurophysiological criteria for demyelination are CMV (~10% of cases)15,120, Epstein–Barr virus (2–10% of
summarized in Table  2. Nerve conduction velocities cases) and M. pneumoniae (1–5% of cases)121. However,
(NCVs) might remain normal until up to 3 weeks after the detection of these pathogens is not specific enough
onset of symptoms; thus, normal NCVs early in the dis­ to aid in the diagnosis of GBS.
ease course do not exclude a diagnosis of AIDP119. Signs Although a number of autoantibodies have been
of axonal pathology (such as reduced CMAP, amplitude detected and studied in GBS122 (Table 1), none is of direct
loss and signs of denervation on electromyography) in diagnostic value, with the exception of GQ1b antibodies
isolation occur in 3–30% of patients with GBS115 and in MFS. Antibodies against paranodal proteins are asso­
do not contradict a diagnosis of AIDP. In one study, ciated with acute-​onset CIDP79 (that is, a GBS mimic),
24% of patients with an initial diagnosis of AIDP were but their testing is rarely indicated in acute GBS. New
rediagnosed with AMAN after NCS were performed10; technologies have begun to identify antibodies that
however, distinguishing AIDP from AMAN in the early bind to multiple gangliosides or glycolipids50,51,123. These
phase of the disease is not crucial as this does not guide antibodies bind to mixtures of different ganglio­sides
therapy (see Management). arranged as they would be on cell membranes in vivo
Nerve biopsy is not recommended for the diagno­ rather than binding to individual gangliosides. Screening
sis of GBS, although it can be used to exclude other patient sera for ganglioside or glycolipid complexes

Table 2 | Characteristics of immune-​mediated neuropathies


Symptoms Reflexes Nerve conduction study Nerve biopsy sample findings Nerve imaging findings
findings
Guillain–Barré syndrome
• The first symptoms are Generalized In AIDP: • Nerve biopsy is not Nerve enlargement detected
combinations of numbness, hyporeflexia • Features of demyelinationa (only recommended, although can be using ultrasonography has
paresthesia, weakness and or areflexia assessable if the distal CMAP, used in some cases been demonstrated in early
pain in the limbs a measure of axonal integrity • Findings from sural nerve biopsy Guillain–Barré syndrome264
• Other symptoms include amplitude, is >10% LLN) sample and autopsy include
bilateral weakness of facial In AMAN and AMSAN: the infiltration of T cells and
muscles (owing to cranial • No features of demyelination macrophages in the affected
nerve involvement) and (or one demyelinating feature nerves, plexus and nerve roots;
autonomic dysfunction in one nerve if distal CMAP segmental myelin destruction
• Limb muscle weakness is amplitude is <10% LLN) with immunoglobulin and
progressive bilateral and • Distal CMAP amplitude is <80% complement deposits; and
symmetric LLN in more than two nerves prominent, macrophage
• Progression of motor • Transient motor nerve infiltration, even in an electro-
symptoms over 12 hours to conduction block might physiologically normal sural
28 days before a plateau is be present262 nerve263
reached
Chronic inflammatory demyelinating polyradiculoneuropathy
• Chronic progressive, Absent or Features of demyelinationa • Nerve biopsy is not routine • MRI can detect increased
stepwise or recurrent reduced • Findings from sural nerve signal intensity or contrast
symmetric proximal and tendon biopsy samples include: signs enhancement in the
distal muscle weakness and reflexes in all of chronic demyelination roots or plexus (>50% of
sensory dysfunction of all extremities and remyelination; length-​ patients)64,265
extremities dependent axonal damage; • Nerve ultrasonography
• Cramps and fasciculations nonspecific inflammatory may show enlarged
in the affected limb infiltrates; and pericapillary nerves266; normalizes with
accumulation of macrophages remission267
Multifocal motor neuropathy
Slowly progressive or Decreased Conduction block Increased number of thinly • T2-hyperintense or
stepwise progressive, or absent myelinated, large-​calibre fibres; gadolinium-​enhancing
asymmetric limb weakness or tendon very mild degree of sensory fibre areas in the brachial plexus
motor involvement that has a reflexes in involvement268 (one-​third of patients)
motor nerve distribution the affected • Multifocal nerve
limb enlargement on nerve
ultrasonography
AIDP, acute inflammatory demyelinating polyradiculoneuropathy; AMAN, acute motor axonal neuropathy; AMSAN, acute motor and sensory axonal neuropathy;
CMAP, compound motor action potential; LLN, lower limit of normal. aFeatures of demyelination include prolonged distal motor latency, decreased motor nerve
conduction velocity, increased F-​wave latency, conduction blocks and temporal dispersion269.

10 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

Box 2 | Predicting the clinical course and treatment response of GBS Insufficiency Score (EGRIS) predicts the development
of respiratory insufficiency within 1 week after hospital
mEGOS — modified Erasmus GBS Outcome Score admission and is based on the number of days between
Captures: the onset of muscle weakness and hospital admission,
• Age clinical assessment by the Medical Research Council
• Preceding diarrhoea (MRC) sum score indicating the extent of muscle weak­
• GBS disability score 2 weeks after hospitalization ness and the presence of facial and/or bulbar weakness,
Scores range from 1 to 7 and predict the clinical outcome after 6 months; a higher score altogether indicating severity of disease and, therefore,
corresponds to worse prognosis. the likelihood of respiratory insufficiency127. Both meas­
ures have recently been assessed in a Japanese multi­­
EGRIS — Erasmus GBS Respiratory Insufficiency Score
centre cohort of patients with GBS and could reliably
Captures at time of hospital admission: predict respiratory insufficiency128.
• Days between clinical onset of weakness
• Facial and/or bulbar weakness CIDP and CIDP subtypes
• MRC sum score Clinical manifestations. As in GBS, the clinical mani­
The scoring system ranges from 0 to 7, with corresponding chances of respiratory festations of CIDP include an acquired progressive
insufficiency within the first week after hospital admission from 1% to 91%. symmetric sensorimotor neuropathy. However, in con­
ΔIgG trast to GBS, CIDP must be progressive for ≥8 weeks
• Increase in serum IgG levels 2 weeks after treatment with intravenous immunoglobulin to fulfil the diagnostic criteria. Motor deficits dominate
in patients with CIDP, and proximal and distal muscle
Larger increase is associated with a better clinical outcome.
weakness should be present for diagnosis; distal promi­
GBS, Guillain–Barré syndrome; IgG, immunoglobulin G; MRC, Medical Research Council. nence is often present. Hyporeflexia or areflexia is pres­
ent in most patients129. Facial nerve involvement is found
increases sensitivity compared with using single mol­ in 10–15% of patients whereas 5% have oculomotor
ecules in the detection method but is not yet widely nerve involvement, although cranial nerve involvement
available clinically owing to the complexity of the assays. or autonomic involvement is rarer in patients with CIDP
Although associations between anti-​ganglioside com­ than in those with GBS. Sensory deficits occur in up to
plex antibodies and clinical phenotypes are still emerg­ 90% of patients129,130; pain can be the initial symptom
ing, they can give some insight into clinical subtypes, in rare cases131, although it is present in up to 70% of
progression and management54. patients in the long term132. About half of patients devi­
ate from this typical manifestation and have severe dis­
Differential diagnosis. Increased CSF cell counts (par­ tal sensory involvement, asymmetrical muscle weakness
ticularly >50 cells µl–1) can indicate infection with CMV or substantial cranial nerve involvement. Up to 10% of
or lyme borreliosis, or can suggest HIV radiculitis, trans­ patients have subclinical signs of CNS involvement or
verse myelitis or poliomyelitis. Blood tests can exclude central demyelination in neurophysiological studies
hypokalaemia as a rare cause of acute muscle weakness or MRI133,134.
and vitamin B6 deficiency, such as in the context of levo­ CIDP subtypes include pure motor CIDP or pure
dopa pump treatment124. If patients have asymmetric sensory CIDP, which are both believed to comprise
muscle weakness, then vasculitic neuropathy, lyme ~10% of CIDP cases; few reports broaden this spec­
borreliosis, diphtheria and poliomyelitis should be con­ trum to include axonal variants of CIDP (that is, chronic
sidered, whereas in patients with pure motor weakness, inflammatory axonal polyneuropathy (CIAP))135. Some
myasthenia gravis, polymyositis and dermatomyositis, forms of initial pure sensory CIDP can progress to
poliomyelitis, hypermagnesaemia, porphyria, botulism include the development of motor symptoms after a
and lead or organophosphate poisoning should be ruled few years. The distal subtype of CIDP, DADS, is charac­
out16. NCS are helpful to rule out disorders that are not terized by pronounced paresis of the small hand and
caused by neuropathy (such as myasthenia gravis or foot muscles and often a severe sensory afferent deficit
myositis), even if these studies do not prove demyelin­ with unsteady gait136 and is frequently associated with
ation. Spinal MRI can be used to exclude spinal cord com­ a monoclonal gammopathy. Anti-​MAG antibodies can
pression injury or transverse myelitis. Leptomeningeal be detected in approximately two-​thirds of patients
malignancy, especially in patients with lymphoma, can with DADS, which might prompt a diagnosis of IgM
also mimic GBS and is not always associated with an MGUS neuropathy. In patients with DADS, clinical
increased CSF cell count, such that spinal MRI may neurophysiology shows marked distal demyelination
be needed125. in motor nerves, indicated by greatly prolonged distal
motor latencies. The response to immunotherapy in
Prognostic tools. Various prognostic algorithms have patients with DADS and other IgM-​associated neuro­
been developed to predict the clinical course of GBS pathy is on average worse than in classical CIDP137.
(Box 2). The so-​called modified Erasmus GBS Outcome MADSAM neuropathy is a chronic progressive multi­
Score (mEGOS) captures patients' age, preceding diar­ focal asymmetric neuropathy with signs of demyelin­
rhoea and GBS disability score 2 weeks after hospital ation. MADSAM is considered a CIDP subtype by some
entry; increased age, the presence of preceding diar­ clinicians but as a separate immune-​mediated neuro­
rhoea and increased disability scores all correspond pathy by others. In this disorder, the upper extrem­
to worse prognosis126. The Erasmus GBS Respiratory ities are dominantly affected, without a generalized

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 11

0123456789();
Primer

Box 3 | Autoantibodies in CIDP: implications for diagnosis and management Nerve and root imaging are starting to be integrated into
the routine diagnostic work-​up of CIDP (Table 2). Nerve
Intense focus on anti-​paranodal antibodies in patients with chronic inflammatory biopsy (usually of the sural nerve) is not part of the rou­
demyelinating polyradiculoneuropathy (CIDP) has led to defined subgroups of patients tine work-​up in CIDP but can be carried out in patients
with common clinical phenotypes. For example, anti-​contactin 1 antibodies have with atypical clinical manifestations2 or to search for
been found in 2.4–8% of patients with CIDP55, and those positive for contactin 1
an alternative diagnosis if patients have no response to
immunoglobulin G4 (IgG4) are poor or partial responders to intravenous
immunoglobulin (IVIG) therapy and are characterized by certain clinical characteristics, immunotherapy142. Ultrasonography or MRI can guide
including rapid onset, early axonal damage and motor predominance, sensory ataxia nerve biopsy if the sural nerve is not affected and fasci­
and tremor55. Anti-neurofascin 155 antibodies have been found in 4–18% of patients cular biopsy of a mixed nerve is considered. MRI-​guided
with CIDP193,244,246, and patients with anti-​neurofascin 155 IgG4 antibodies are poor or fascicular biopsy of the sciatic nerve and of the lumbar
partial responders to IVIG and have characteristic clinical phenotypes, including a or brachial plexus has been described, has demonstrated
disabling tremor and distal motor neuropathy with ataxia. Antibodies to contactin-​ abnormalities in patients with CIDP and has few adverse
associated protein 1 have been associated with Guillain–Barré syndrome-​like and effects, although data are lacking regarding the exact
CIDP-​like disease with prominent pain61. The detection of these patient subgroups is clinical yield in comparison to other methods143. Corneal
important, as these patients might respond better to plasma exchange and anti-​B cell confocal microscopy, which is available in collaboration
therapies, such as rituximab, than to standard CIDP treatment247. Still, the overall
with ophthalmological departments, has been suggested
number of patients with paranodal antibodies is very low, making a definite statement
on incidence, clinical phenotypes and treatment responses difficult. Setting up an as an additional technical method in diagnosing and
international registry capturing this group of patients might be a helpful strategy to monitoring CIDP144.
better understand this subgroup of CIDP. The presence of several clinical or laboratory features
can be used to exclude a diagnosis of CIDP. Clinical
manifestations or other disorders that exclude CIDP
areflexia138. The distinction of MADSAM from MMN is diagnosis include retinitis pigmentosa; ichthyo­sis; evi­
therapeutically relevant as MADSAM might respond to dence of a hereditary disorder or an adequate toxin
steroids or IVIG whereas MMN is not steroid sensitive exposure; a sensory level indicating a spinal cord lesion;
(see Management). sphincter dysfunction; hypocholesterolaemia; vitamin
B12 deficiency; untreated hypothyroidism; porphyria;
Diagnostic work-​up. The diagnosis of CIDP is less electrophysiological findings of myasthenia gravis
straightforward than that of GBS137 (Table 2). No defini­ myopathy or anterior horn disease; and biopsy sample
tive test or biomarker is available for CIDP; accordingly, evidence of a storage disease, vasculitis or amyloidosis.
several diagnostic criteria have been developed, pri­
marily for use in clinical trials. These criteria are com­ Differential diagnosis. One of the clinical challenges is
posed of clinical, laboratory and electrophysiological the distinction between CIDP with acute onset and GBS.
parameters. Electrophysiological criteria are designed Acute CIDP and GBS have similar clinical presentations
to detect demyelination, although these should be during the first 4 weeks, particularly if patients with GBS
used only in conjunction with results from clinical and present with treatment-​related symptom fluctuations.
labora­tory findings owing to the presence of demyelin­ The differentiation of acute CIDP and GBS is impor­
ation in other neuropathies. Axonal pathologies, whether tant as treatment strategies and prognosis differ (see
primary or secondary to demyelination, are common Management)145. Although patients with acute CIDP
and do not exclude the diagnosis of CIDP. At least seem to exhibit a higher frequency of sensory symptoms
15 different sets of diagnostic criteria for CIDP have than those with GBS, the clinical differentiation of both
been published139, of which the criteria of the EFNS/ entities remains difficult146. The differential diagnosis of
PNS had the best sensitivity and specificity 137,139. paediatric-​onset CIDP from hereditary neuropathies is
However, the sensitivities and specificities of each cri­ challenging and genetic testing may be needed. Given
terion differ between studies, and some of the criteria the limited number of cases reported thus far, children
sets differentiate between probable, possible and defi­ diagnosed with CIDP have a more favourable long-​term
nite CIDP, which also leads to different sensitivity and course than adults with similar treatments147. Distal sym­
specificity values. metric polyneuropathy is the most common neuropathy
Elevated CSF protein in conjunction with normal or seen in patients with diabetes mellitus. However, these
only mildly elevated CSF cell count (<10 cells µl–1) is a patients can also develop CIDP, and whether diabe­
supportive criterion for CIDP. Although older studies tes mellitus predisposes to CIDP is controversial. The
reported elevated CSF protein in ~90% of patients with different treatment strategies of CIDP and distal sym­
CIDP3,140, this may not hold true if current clinical and metric polyneuropathy mandate for a thorough clinical
electrophysiological criteria are used and in chronic differentiation of diabetic and immune-​mediated neuro­
progressive forms141. In addition, >50 white cells µl–1 in pathies148. Infrequently, patients with primary axonal
the CSF rules out a diagnosis of CIDP. A clinical pic­ neuropathies, such as vasculitis149 or hereditary amyloid­
ture of CIDP, sometimes with an acute onset, may be osis150, can reveal electrophysiological abnormalities
found in patients with antibodies to the paranodal pro­ in the range of a demyelination similar to that observed in
teins NF155, contactin 1, or CASPR1 (refs61,80) (Box 3). patients with CIDP151. Late-​onset hereditary neuropa­
Neuropathies with this constellation have been named thies of the Charcot–Marie–Tooth (CMT) type often
‘paranodopathies’. The distinction is of importance fulfil the CIDP diagnostic criteria but do not respond
because symptoms may respond better to plasmaphere­ to CIDP therapies150, such that, when the diagnosis is
sis and B cell therapies than to standard CIDP treatment. in doubt, genetic testing with a panel for demyelinating

12 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

CMT may be needed. In addition, some therapies, such depending on the extent of secondary axonal degen­
as amiodarone, can cause a demyelinating neuropathy eration. In contrast to findings in patients with CIDP,
mimicking CIDP152. CSF shows moderately elevated protein in only 30%
As the clinical presentations of GBS reach their of patients, which is not diagnostically helpful160. No
nadir within 4 weeks and in >8 weeks in CIDP, a gap sensory deficits are typically observed in clinical or
evolved for those immune-​mediated neuropathies in electrophysiological testing.
which progressive motor and/or sensory deficits pre­
sented with a time to nadir between 4 and 8 weeks. Differential diagnosis. The differential diagnosis of
These neuropathies are called subacute inflammatory MMN includes early stages of motor neuron disease;
demyelinating neuropathies (SIDPs)153, bridging the however, the clinical manifestation of MMN differs from
gap between GBS and CIDP. Diagnostic criteria are motor neuron disease by slow disease progression and
similar, including elevated high spinal fluid protein the absence of upper motor neuron and bulbar signs.
level (>55 mg dl–1) and inflammatory cells in the nerve The differential diagnosis of MMN from lower motor
biopsy sample. neuron disease might be more difficult; in this case, clin­
ical follow-​up and response to immunoglobulin therapy
MMN provide the most useful information. Pure motor CIDP
Clinical manifestations. MMN is characterized by slowly and MADSAM might be considered in patients with
progressive or, less frequently, episodic asymmetric suggestive MMN. Hereditary neuropathy with pressure
paresis that follows the distribution of individual nerves palsies is usually easy to distinguish from MMN as this
and starts most often in the arms, without obvious sen­ disorder preferentially affects compression sites.
sory loss10. The ulnar, median, radial and tibial nerves
are most commonly affected. Weakness of muscles MGUS neuropathy
innervated by the same nerve can differ considerably Neuropathy with MGUS is a heterogeneous condition.
within patients, indicating an irregular pattern of nerve Because MGUS can co-incidently be present in axonal
involvement154. Proximal pareses are the main symptom neuropathies of any aetiology, and in CIDP, the clini­
in only 5% of patients and leg pareses in 10% of patients. cal manifestations are not dependent on the presence of
The early stages of MMN are characterized by paresis MGUS. There are a few typical syndromes with neuropathy
without muscle atrophy, but muscle atrophy is pro­ and specific types of MGUS that are discussed below.
nounced at later stages of disease155. Fasciculations may MGUS can be detected in 5% of the population
be observed. Muscle strength might deteriorate in cold >70 years of age; in 90%, the paraprotein is of the IgG
temperatures and after exercise. Reflexes are attenuated or IgA type161. By definition, for a diagnosis of MGUS,
within the paretic regions, although complete areflexia the paraprotein level must be <3 g dl–1 and the plasma
and hyperreflexia are rare. In addition, approximately cell composition of the bone marrow is <10%. MGUS
half of patients have muscle cramps and fasciculations, must be distinguished from other conditions that are
whereas cranial nerve involvement is observed only in associated with IgG or IgA monoclonal gammopathy,
2% of patients154. Mild sensory symptoms might develop such as POEMS syndrome, immunoglobulin light-​chain
over time in ~20% of patients156, although pain or amyloidosis, lymphoma and cryoglobulinaemia.
autonomic symptoms are not part of the syndrome. As neuropathies occur in 6.6% of the population
>60 years of age162, the co-​occurrence of monoclonal gam­
Diagnostic work-​up. Diagnostic criteria for MMN are mopathy and neuropathy is coincidental and does not
based on clinical, electrophysiological and laboratory indicate that the MGUS is causal to the neuropathy. Most
characteristics10,157. Conduction block demonstrated in experts consider a neuropathy that fulfils the diagnostic
NCS in motor nerves outside entrapment sites (such as criteria for CIDP in a patient with an IgG or IgA MGUS
the carpal or cubital tunnel) supports the diagnosis of as classical CIDP. Similarly, in a patient with a mild axonal
MMN. Although the presence of conduction block has neuropathy and concomitant IgG or IgA MGUS, the
an important role in the diagnosis of MMN, its definition co-​occurrence is usually considered coincidential and
varies between expert groups and in the sets of diagnos­ the neuropathy is diagnosed as idiopathic, which has a
tic criteria. Whether the presence of conduction block good prognosis and needs no specific therapy163. In other
is obligatory for the diagnosis of MMN is debated, and patients, a causal connection between the paraprotein and
the EFNS/PNS diagnostic criteria allow for a diagnosis the neuropathy is assumed. The best-​defined subtype of
of possible MMN in the absence of conduction block158. MGUS neuropathies is that associated with anti-​MAG
The detection rate of conduction block in patients with antibodies, which are found in up to half of the patients
MMN depends on the number of nerves investigated, with IgM MGUS. Most patients with anti-MAG anti­
and conduction blocks in proximal nerves might be par­ bodies have the DADS phenotype with paresis of the
ticularly difficult to detect. Patients with MMN with and small hand and foot muscles (see above). Enzyme-linked
without evidence of conduction block may respond to immuno­sorbent assay (ELISA) is more sensitive than
IVIG treatment159. western blot for the detection of anti-​MAG anti­bodies164.
In addition to conduction block, other signs of Their titre does not correlate with disease seve­rity.
demyelination, such as prolonged distal motor laten­ Another screening test for anti-MAG antibodies is indi­
cies and slowed NCV, are variable in patients with rect immunohistochemistry with patient serum on a nerve
MMN. Denervation and regeneration potentials can preparation. NCS show a prolonged distal latency out
be detected using electromyography in paretic muscles of proportion with conduction slowing in intermediate

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 13

0123456789();
Primer

segments of the nerves along the upper and lower no further benefit171. IVIG is usually administered over
limbs. Nerve biopsy is not mandatory in the diagnostic 2 consecutive days; this single course is usually sufficient
work-up of anti-MAG neuropathy, but data from earlier in patients with GBS and does not require consecu­
electron microscopy studies indicate that there might tive courses in classical courses of GBS. Further trials
be enlarged spaces between the myelin lamellae165. are needed to establish the optimal dose of IVIG38 for
In patients presenting with suspected MGUS neuro­ the treatment of GBS, whether additional infusions
pathy, and in whom anti-​M AG antibodies are not are beneficial in patients with severe GBS or patients
detected, screening for other antibodies against chon­ with a clinical relapse within 8 weeks after initial dis­
droitin sulfate, gangliosides, sulfatides, sulfated or ease manifestation and to determine whether IVIG is
cytoskeletal proteins and heparin disaccharide can be effective in patients with mild GBS170,172. Change in
performed using western blot or ELISA166. The presence serum IgG 2 weeks after IVIG might represent a bio­
of these antibodies increases the likelihood of a patho­ marker for treatment response173. The clinical use of
genetic link between the paraprotein and neuropathy; IVIG is mostly safe; however, adverse effects such as
however, their diagnostic importance is uncertain. Other hypersensitivity reactions, haemolytic anaemia, asep­
IgM antibodies have been associated with MGUS neuro­ tic meningitis and thromboembolism may occur in
pathy, including anti-​G D1b, anti-​G D2, anti-​G D3, rare instances, whereas with plasma exchange adverse
anti-​GT1a, anti-​GT1b, anti-​GQ1b, anti-​GM2 or anti-​ events related to the use of citrate (hypocalcaemia and
GalNac-GD1a antibodies. IgM antibodies that bind to metabolic alkalosis) and haemodynamic changes are
the disialosyl epitope of several gangliosides such as not uncommon.
GD1b, GD2, GD3, GT1a, GT1b and GQ1b are associated Corticosteroids are not effective for the treatment
with chronic ataxic neuropathy. A chronic ataxic neuro­ of GBS regardless of whether they are used alone or in
pathy with ophthalmoplegia and bulbar involvement, combination with other therapies174. Owing to the pro­
also named chronic ataxic neuropathy with ophthal­ posed role of complement in GBS, the anti-​C5 human­
moplegia, M proteins, cold agglutinins and disialosyl ized monoclonal antibody eculizumab has been trialled
antibodies (CANOMAD), is observed in connection in patients with severe GBS175,176. However, the two trials
with asialoganglioside antibodies167. Anti-​trisulfated evaluating this drug included a small cohort, which pre­
heparin disaccharide antibodies cause a painful sensory cludes statistically valid assessment of efficacy. However,
axonal neuropathy or motor neuropathy168. these trials do indicate that eculizumab is well tolerated
in patients with GBS and that larger, multicentre, inter­
Management national trials are warranted. Mechanical ventilation,
The mainstay of treatment for the immune-​mediated often for months at a time, is required in the most severe
neuropathies is immunotherapy — particularly for acute cases of GBS in which patients develop tetraparalysis and
disorders but also for chronic variants. The low preva­ respiratory failure. At present, the treatment strategies
lence of these diseases makes it difficult to accumulate for AMAN and AIDP do not differ in clinical practice.
sufficient clinical trial data to qualify as class I evidence.
As such, recommendations for many therapies are based CIDP and CIDP subtypes
IVIG, plasma exchange and corticosteroids. Randomized
on case series or smaller open-​label studies. In addition
to immunotherapy, physiotherapies can improve clinical controlled studies (reviewed in ref.177) demonstrated
disability in patients. that plasma exchange and IVIG are of clinical benefit
in patients with CIDP, and other evidence from clinical
GBS trials, case series and anecdotes from clinical practice
The first-​line treatments for GBS are plasma exchange suggests that corticosteroids are also effective in these
and IVIG, both of which have proven clinical effi­ patients178–180. Indeed, IVIG, plasma exchange and corti­
cacy as shown in numerous double-​blinded, placebo-​ costeroids represent the mainstay of first-​line treatment
controlled clinical trials (reviewed in ref.169). These for CIDP and have a similar level of clinical efficacy.
therapies improve motor deficits in patients with GBS, IVIG treatment resulted in a significant clinical
in addition to cranial nerve involvement and sensory improvement of motor symptoms in patients with
deficits at a similar rate. Plasma exchange is a process CIDP compared with placebo181. However, the limitation
whereby the patient’s plasma is removed and discarded of earlier studies was a short observation period. The
and is replaced by donor plasma or a plasma substitute, ICE (IVIG-​C CIDP efficacy) study182 addressed some
whereas IVIG involves the intravenous delivery of puri­ of these limitations and evaluated the use of IVIG in a
fied, donor immunoglobulins into the circulation. No randomized, double-​blind, placebo-​controlled trial for
difference in the efficacy of plasma exchange or IVIG in 24 weeks, followed by a secondary round of randomi­
patients with GBS has been reported170. In patients with zation and an extension of 24 weeks. This study was the
mild GBS, two plasma exchanges administered 2 days first to demonstrate a significant clinical improvement
apart shortened the time until onset of motor recovery with long-​term IVIG therapy compared with placebo.
compared with patients not receiving plasma exchange, However, questions still remain on the optimal dose for
whereas administering four plasma exchanges every patients as well as timing and duration. Individual clini­
other day was superior to administering two plasma cal patient responses differ and require adequate clinical
exchanges in terms of the time to walk with assistance monitoring as well as dose adaptation. Similarly, the
and muscle strength recovery in patients with mod­ clinical effect of plasma exchange usually lasts only for
erate to severe GBS; additional plasma exchanges had weeks, sometimes months, and therefore may require

14 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

continuous repetition if other treatment modalities, drugs have been tested in controlled clinical trials, and
which are easier to apply, fail in patients. the use of these drugs is largely based on anecdotal evi­
In a retrospective analysis, intermittent high-​dose dence, which underlines the pressing need for more
intravenous methylprednisolone (IVMP) improved rigorous clinical evaluation.
muscle strength in patients with CIDP, similar to the Only a few studies have evaluated the use of metho­
effect observed with IVIG or oral prednisolone in these trexate monotherapy in patients with CIDP193. In a series
patients183. In addition, in another study, patients who of ten patients, seven increased muscle strength by
responded to IVMP had longer-​term remission than ≥2 points on the MRC sum score with weekly low-​dose
those on IVIG184. However, the discontinuation of treat­ oral methotrexate, although three patients deteriorated.
ment is higher with IVMP than IVIG owing to ineffi­ In this study, only two patients had an improvement in
cacy or an increased frequency of adverse effects185. disability, although these patients were also receiving
Prospective clinical data regarding the benefit of IVMP corticosteroid treatment193. In a randomized controlled
should be obtained before this drug is recommended trial of 59 patients with CIDP, no benefit in methotrexate
as a standard therapy for CIDP. CIAP might res­pond was demonstrated compared with placebo188. Thus, at
to immunomodulatory treatment in a similar way present, data are insufficient to recommend low-​dose
as CIDP186. methotrexate as a standard immunosuppressive therapy
in patients with CIDP.
Subcutaneous immunoglobulins. The increasing use of Cyclosporine should be considered only in patients
immunoglobulins for the treatment of chronic immune-​ resistant to more conventional therapies given the
mediated neuropathies, particularly in patients who severe adverse effects (nephrotoxicity, neurotoxicity,
require long-​term therapy, has led to interest in admin­ hypertension, hyperlipidaemia and diabetes mellitus)
istration of subcutaneous immunoglobulin (SCIG) as associated with this drug. In one study, three out of
the long-​term application at home might be preferred eight patients with CIDP had a clinical improvement, as
by some patients. To date, only two placebo-​controlled, demonstrated by assessing muscle strength and walk­
double-​blind clinical trials and one observational study ing speed with cyclosporine A treatment, and no seri­
have reported on the efficacy and tolerability of SCIG ous adverse effects were reported194. In another study,
in patients with CIDP187. The first study demonstrated modified Rankin and Inflammatory Neuropathy Cause
the superiority of SCIG compared with placebo in and Treatment Group (INCAT) disability scores were
patients with CIDP who previously responded to IVIG significantly reduced and grip strength was significantly
as assessed by muscle strength and functional abil­ enhanced in patients with treatment-​resistant CIDP
ity169,188. The second randomized, double-​blind, placebo-​ who received a microemulsion of cyclosporine A, and
controlled trial demonstrated a significant reduction in these effects persisted for the observation period of up
the percentage of patients with CIDP relapse or treat­ to 1.5 years195.
ment discontinuation during 24 weeks of treatment with Mycophenolate mofetil add-​on therapy stabilized
SCIG compared with placebo189. In a prospective obser­ the clinical condition and permitted a reduction in the
vational study, an equivalent efficacy of SCIG and IVIG use of previous medications, such as steroids or IVIG
was demonstrated in these patients who switched from in three patients with CIDP and one patient with IgM
IVIG to SCIG190. In addition, several case series and case neuropathy in one study196. However, median muscle
reports have corroborated the utility of SCIG in patients strength, sensory or the modified Rankin score, a com­
with CIDP (reviewed in ref.191). As such, in countries monly used clinical outcome measure in neurology, did
where reimbursed, SCIG is used as a treatment option not change after treatment. Despite the small number of
for patients with CIDP. The spectrum of adverse effects patients included in these trials, these data do support
is similar to IVIG, such as tolerability, headache, nausea the use of mycophenolate mofetil as an add-​on therapy
and local skin reactions. in immune-​mediated polyneuropathies197.
In addition, high-​dose cyclophosphamide improved
IFNβ. IFNβ has numerous immunomodulatory effects motor strength and QOL (demonstrated using the
and is used for the treatment of multiple sclerosis, provid­ 36-Item Short Form Survey (SF-36) questionnaire) in
ing the rationale for the use of this drug in inflammatory a small study of patients with a subset of severe, refrac­
neuropathies. tory CIDP198. In another series, 11 out of 15 patients
Anecdotal observations suggest that IFNβ may be with CIDP had clinical remission for up to 9 years after
a useful adjunctive therapeutic option in patients with monthly pulse intravenous cyclophosphamide for up to
CIDP. However, in a controlled clinical trial, intramus­ 6 months199. However, patients and clinicians are hesitant
cular IFNβ1a did not exhibit any clinical superiority to use or prescribe cyclophosphamide for CIDP until
compared with placebo in patients with CIDP192. further controlled clinical trials provide sufficient evi­
dence for its efficacy owing to the severe adverse effects
Classical immunosuppressive drugs. The use of immuno­ of this drug, such as haemorrhagic cystitis, infertility
suppressive drugs, such as methotrexate, cyclosporine, and increased risk of infections and cancer. Combining
mycophenolate mofetil and cyclophosphamide, in cyclophosphamide with fludarabine might represent
inflammatory neuropathy is based on the assumption a potent treatment approach in otherwise treatment-​
that immunomodulatory agents that are effective in other resistant patients with CIDP, as demonstrated in a small
immune-​mediated diseases would also be of benefit in case series reporting improvements in motor strength200.
these neuropathies. However, few immunosuppressive Limited studies have been carried out to evaluate the use

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 15

0123456789();
Primer

of azathioprine in CIDP; accordingly, insufficient evi­ improvements in functional self-assessment, standard­


dence is available to determine the efficacy of this drug ized neurological examination and formal strength
in patients with this disorder197,201. testing and NCS with IVIG treatment213.
SCIG could represent an interesting alternative
Monoclonal antibodies. Monoclonal antibodies can tar­ to IVIG in selected patients with MMN. Indeed, one
get specific immunopathogenetic mechanisms that are single-​blind crossover study demonstrated the therapeu­
common to a number of immune-​mediated diseases tic equivalence of SCIG and IVIG at improving motor
and therefore have the potential to be similarly effective strength in patients with MMN214, although the cohort
across a range of disorders. However, unexpected and size was small (nine patients). Observation of five of
severe adverse effects, such as infections or secondary these patients and one additional patient for 24 months
autoimmune diseases, can arise even years after therapy confirmed sustained efficacy and tolerability215. In addi­
has been initiated202, therefore, long-​term monitoring tion, one other study demonstrated an equivalent clinical
is warranted. efficacy in terms of improving motor strength of SCIG
Alemtuzumab is a humanized monoclonal antibody treatments in patients previously treated with IVIG216.
that targets CD52, which is expressed by most lympho­ Additional smaller case series or reports have empha­
cytes and monocytes. In a single case report, clinical sized the potential efficacy of SCIG in patients with
improvement of motor deficits of a patient with CIDP MMN191. However, data for the long-​term use of SCIG
with alemtuzumab was noted203. A retrospective study in the treatment of MMN are not available.
of seven patients with CIDP who were heavily depend­
ent on IVIG therapy demonstrated a reduction in the Monoclonal antibodies. Among the different monoclo­
required dose and frequency of IVIG treatment with nal antibodies, evidence for clinical efficacy has been
alemtuzumab. In addition, two of the seven patients had gathered in MMN only for rituximab. Rituximab is
prolonged remission, and two demonstrated a partial a mouse–human chimeric antibody that binds to the
response, although no clinical benefit was observed in CD20 antigen expressed on the surface of pre-​B cells,
the remaining three patients204. Three patients also devel­ mature B cells and memory B cells. Rituximab was orig­
oped secondary autoimmune diseases204. Accordingly, inally used for the treatment of non-​Hodgkin lymphoma
further studies, including studies with larger num­ and other neoplastic B cell-​mediated diseases owing to
bers of participants, are needed to evaluate the thera­ the function of this drug in eliminating B cell precursors
peutic potential of alemtuzumab in the treatment of and B cells from the circulation via antibody-​dependent
immune-​mediated neuropathies. cytotoxicity and complement-​dependent cytolysis. In an
Natalizumab is a monoclonal antibody directed open-​label study, six patients with MMN receiving IVIG
towards the α4 integrin of the VLA4 antigen that is therapy were also given rituximab, although rituximab
expressed on the surface of T cells and blocks the inter­ add-​on therapy did not reduce the required dose of IVIG
action between VLA4 and VCAM1 on endothelial or convey any clinical improvement. However, other
blood vessels, thereby inhibiting the extravasation of studies reported improved motor strength after switch­
immune cells along the blood–brain barrier as well as ing from IVIG to rituximab217. Thus, controlled studies
their migration through the extracellular matrix205. In are warranted to better understand the clinical value of
one case series, natalizumab therapy demonstrated a rituximab for the treatment of MMN.
variable efficacy in three patients with CIDP who were
unresponsive to first-​line therapies and presented with MGUS neuropathy
severe and frequent relapses over a number of years206. IVIG, PE and corticosteroids. IVIG is not the first-​line
Some patients demonstrated long-​term improvement, therapy for MGUS neuropathy with anti-​MAG IgM anti­
dramatic improvement over a substantial time frame bodies owing to a lack of compelling evidence of clinical
or stabilization on clinical measures assessing motor efficacy. By contrast, smaller studies have suggested a
strength206, whereas patients in another case report did stronger clinical improvement in muscle strength with
not have a clinical benefit with natalizumab use207. These plasma exchange. The evidence for a clinical beneficial
discrepancies reflect the underlying immunopathologi­ effect for corticosteroids in MGUS neuropathies is rather
cal heterogeneity in CIDP; further evaluation of natali­ poor. In a combination study with cyclophosphamide
zumab will require controlled clinical trials, perhaps in for 6 months, no difference was observed in functional
different subgroups of patients. scales compared with placebo218. Patients with MGUS
neuropathy with IgG or IgA antibodies should be con­
MMN sidered indistinguishable from patients with idiopathic
Immunoglobulin therapy. IVIG is the first-​line treatment CIDP and should therefore be treated with IVIG, plasma
for MMN and, in contrast to CIDP, plasma exchange exchange or corticosteroids.
and corticosteroids are ineffective in these patients208.
The response to treatment with IVIG in patients with Monoclonal antibodies. Rituximab has been shown to
MMN can be maintained in the long term by increas­ be of benefit to patients with polyneuropathies associ­
ing the induction dose and frequency of administra­ ated with anti-​MAG antibodies219: six patients out of
tion209,210. The efficacy of IVIG in patients with MMN nine demonstrated clinical improvement assessed using
has been demonstrated in randomized, double-​blind, the Neurological Disability Score, two remained stable
placebo-​controlled studies and a few case reports211,212. and one deteriorated219. B cells were depleted in all nine
In one study, 16 patients with MMN demonstrated patients, with a concomitant reduction of anti-​MAG

16 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

antibody titres by >52% in eight patients. A follow-​up assumption on the underlying immunopathogenesis. In
study demonstrated the tolerability of a higher dose of addition, these findings emphasize the complex nature
rituximab and a clinical improvement was demonstrated of the immune responses in peripheral nerve disorders
in four out of eight patients and coincided with improve­ and teach us more about the underlying immunopatho­
ment in motor NCVs and a decrease of anti-​MAG anti­ genesis. With the use of immunomodulatory therapies,
body titres220. In a case series, rituximab stabilized 29% of clinicians should be highly vigilant in monitoring for
patients with anti-​MAG neuropathy and improved motor unexpected complications, even when the drug in ques­
as well as sensory symptoms in 31.5% of patients within tion has proved to be beneficial in clinical trials. Indeed,
the 7–12-month post-​treatment follow-​up period221. the use of these drugs in other immune-​mediated dis­
However, 11% of patients had a transient and revers­ orders, such as multiple sclerosis or rheumatoid arthri­
ible clinical deterioration during or immediately after tis, has shown that severe complications can arise years
treatment, possibly owing to the phenomenon of IgM after therapy.
flare or an increase in pro-​inflammatory cytokines221. In
addition, rituximab was effective in a patient with CIDP Quality of life
and high titres of anti-​sulfated glucuronyl paragloboside QOL is adversely affected in many chronic diseases, but
IgM antibody without M protein222. objectively measuring a patient’s physical, emotional and
Overall, the current assumption based on the avail­ social well-​being is often difficult. Most clinical trials
able data is that one-​third of patients with anti-​MAG IgM involving patients with immune-​mediated neuropathies
MGUS neuropathy will respond well to rituximab, one-​ include outcomes assessing strength and sensory deficits,
third will exhibit a marginal response and one-​third may which are present in 40–50% of patients. Residual dis­
have no response28. To what extent humanized versions ability can persist for many years after recovery from GBS
of rituximab, ocrelizumab or ofatumumab might exhibit or CIDP and can interfere with daily functioning and
clinical efficacy in immune-​mediated neuropathies social activities and, therefore, contribute to a reduction
needs to be explored. in QOL229. Indeed, even 10 years after disease onset, 14%
of patients with GBS have moderate to severe disability,
Physiotherapy whereas a further 50% of patients have minor sensori­
Physiotherapy might be beneficial in improving func­ motor symptoms, indicating that this residual disability
tional limitations and reducing chronic disability in might be permanent230. In addition, one-​third of patients
patients with immune-​mediated peripheral neuropa­ with GBS have long-​term activity limitations231.
thies; however, evidence supporting this therapy is Other factors affecting QOL include fatigue, pain, anxi­
limited. One study demonstrated the feasibility and ety and depression, although these factors are difficult to
acceptability of a 12-week unsupervised, community-​ assess quantitatively232. Fatigue is reported by 40–80% of
based strengthening, aerobic and functional exercise patients with immune-​mediated neuropathies233,234 and
programme in patients with stable inflammatory motor is associated with reduced QOL233 and greater activity
neuropathy223. Significant improvements were seen in all restriction230. In a study by the INCAT group, the average
measures of activity limitation in participating patients Fatigue Severity Scale scores were significantly higher in
and in other factors, such as anxiety, depression and patients with previous GBS, stable CIDP or MGUS poly­
fatigue. The chosen modality of physiotherapy might neuropathy than in healthy controls, and 80% of patients
have a critical role in the functional benefit exerted by scored >5, which is indicative of severe fatigue233. Severe
this therapy. In another study, a high-​intensity physio­ fatigue was experienced by 81–86% of patients with nor­
therapy programme was associated with a significant mal strength or sensation and was not associated with the
improvement in the function of relevant domains of level of disability as ambulatory and wheelchair-​bound
the Functional Independence Measure, such as mobil­ patients were similarly affected. Although many chronic
ity, transfers, sphincter control and locomotion, com­ conditions have a negative effect on mental health235,
pared with a lower-​intensity programme in patients few studies have specifically examined mental health
with chronic GBS224. Other studies support these obser­ in patients with immune-​mediated neuropathies. One
vations225,226. Tailoring the physiotherapeutic interven­ study by 323 members of the Dutch Society of Neuro­
tion, as well as considering patient preference for type muscular Disorders reported depression in 6% of
and location of the strengthening exercises, appears to patients with GBS and in 9% of patients with CIDP236.
be important. Further research is required to understand how fatigue,
pain, anxiety and depression contribute to prognosis in
Failed translation patients with immune-​mediated neuropathies and how
Despite a valid immunological rationale supported by these factors can be effectively assessed in the clinic. An
convincing data from preclinical models, some thera­ inflammatory-​neuropathy-specific survey, the IN-​QOL,
peutic strategies did not demonstrate a clinical benefit or has been developed to assess QOL in patients with GBS,
yielded unexpected adverse effects when applied in the MMN, CIDP and MGUS polyneuropathy237. This instru­
clinic. Examples of these therapeutic strategies include ment combines six commonly used QOL questionnaires
infliximab (a monoclonal antibody targeting TNF)227, and tailors them towards outcomes most relevant to
tacrolimus (FK506; an immunosuppressant)228 or fingo­ inflammatory neuropathies.
limod32. These failures could be driven by various factors, Long-​term disability, fatigue or depression can affect
including less meaningful end points, insufficient sam­ all aspects of patients' lives, including family life, social
ple sizes for detecting the expected result or an incorrect activities or working life132. Up to 30% of patients with

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 17

0123456789();
Primer

CIDP retire early owing to their disability, and patients have failed. As such, specifically for a better understand­
with MMN need frequent leave from work owing to ing of chronic immune-​mediated neuropathies, better
hospital admission for IVIG administration238. Many model systems are needed.
patients require long-​term care outside of the clinical
setting as well as additional support from health and Nodopathies
social services239, which can be financially costly over the Although the role of nodal dysfunction in AMAN
course of the disease. Economical self-​management pro­ has been recognized for some time47, the discovery
grammes designed to improve disability, mental health, of autoantibodies to nodal and paranodal proteins in
social participation and QOL are urgently needed, par­ CIDP has helped focus attention on nodal dysfunction
ticularly for the chronic immune-​mediated neuropathies and pathology across several clinical neuropathies.
in which health-​related QOL among patients with CIDP, Immune-​mediated neuropathies have traditionally been
MMN or MGUS polyneuropathies is similarly affected238. classified as either axonal or demyelinating according
Numerous studies have reported the effect of long-​ to certain pathological and electrophysiological charac­
term immunotherapy on health-​related QOL. Indeed, teristics. However, the concept of a nodopathy allows
long-​term immunotherapy, by improving strength, novel pathogenetic mechanisms to be postulated (for
sensation and some neurophysiological parameters, example, the reversible loss of sodium channel func­
increases patient activity and participation, thereby tion due to antibody binding at the node in GBS and
improving health-​related QOL229,240. CIDP). Classifying immune-​mediated neuropathies
as nodo­pathies paves the way for individualized treat­
Outlook ments across the spectrum of immune-​mediated neuro­
Although much progress has been achieved in the pathies incorporating the use of immune biomarkers
field of immune-​mediated neuropathies, many ques­ and electro­physiology. For example, antibody responses
tions remain that need to be addressed to improve the might be useful for guiding treatment options in this
treatment of patients with these disabling diseases. subgroup of CIDP patients. Indeed, resistance to IVIG
treatment is in part due to the IgG isotype subclass of
Animal models the anti-​NF155–contactin 1 response and its ability to
Considerable advances have broadened our understand­ activate complement. In patients with IgG1–3 subclasses
ing of the pathogenesis of immune-​mediated disorders (which fixes and activates complement), administration
of the peripheral nerve, and many of these advances have of IVIG could inhibit complement deposition, providing
been translated into promising therapeutic approaches some therapeutic effect241. However, patients with IgG4
in the EAN model. However, this classical model mimics antibody responses that do not fix complement are resist­
only certain parts of an acute demyelinating immune ant to treatment with IVIG but do seem to respond to
neuropathy and is induced in rats. Genetically modified treatment with rituximab242.
mice would be a more suitable tool to decipher critical
pathogenetic pathways of immune-​mediated damage Biomarkers and diagnostic criteria
to the PNS. Limited protocols are available that allow Important advances have also been made in studying
the induction of EAN in mice, providing the opportu­ surrogate markers in patients. With the introduction of
nity to study various pathogenetic aspects in transgenic advanced biomarker research tools, the understanding
models32. Mouse or rat models of AMAN would be an of the potential pathogenetic role of autoantibodies tar­
interesting and useful expansion of the present experi­ geting peripheral nerve antigens is broadening. As pre­
mental repertoire. Rabbit models of AMAN have been viously mentioned, autoantibodies in GBS that target
generated by immunizing these animals with GD1b gangliosides are predominantly found in axonal variants,
and GM1, although better models may help to further which are less prevalent in the Western hemisphere. The
dissect disease mechanisms of acute AMAN. discovery of autoantibodies that are directed towards
Chronic immune-​m ediated neuropathies, par­ the nodal and paranodal regions of myelinated fibres
ticularly CIDP, are more difficult to mimic in animal and the correlation with certain clinical phenotypes or
models because EAN represents an acute monophasic response to treatment in CIDP80,242–244 provide a conceiv­
model of disease. Repeated immunization protocols in able pattern for future therapeutic strategies in CIDP
rabbits or repeated adoptive transfer of T cells in Lewis research. It seems likely that CIDP, similar to GBS, will
rats prompted a relapsing but not robust clinical course eventually be divided into subtypes on the basis of clini­
(reviewed in ref.32). An alternative model was generated cal features and autoantibody response. However, larger
by cross-​breeding mice deficient in the co-​stimulatory studies are warranted to establish robust differentiation
molecule T lymphocyte activation antigen CD86 (also pathways to be implemented in clinical practice.
known as B7-2) with the immune-​mediated diabetes-​ Beyond the use of clinical manifestations, better clin­
prone non-​obese diabetic (NOD) mouse strain34. Use ical and paraclinical criteria are needed to help predict
of these models might clarify the differences between the individual clinical course and treatment response of
self-​limiting immune responses in GBS and progressive patients. Anticipating response to therapy in patients
or recurring inflammation in CIDP and other chronic with immune-​mediated neuropathies is difficult. Even
immune-​mediated neuropathies. However, these models the INCAT electrophysiological criteria, which are well
reflect only parts of complex immune-​driven disorders elaborated for demyelinating neuropathy, cannot pre­
and might explain why translation of most of the innova­ dict a higher rate of response to immunotherapy245. The
tive treatment strategies described in preclinical models lack of reliable clinical measures underlines the need

18 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

for new ways to conduct clinical trials in this group of identification might extend the clinical spectrum of
heterogeneous disorders. Innovative trial design and the immune-​mediated neuropathies. On the other hand,
use of new end points could enable us to conduct more markers for sensitivity, efficacy, safety and resistance for
treatment trials, perhaps in more clearly predefined on-​market drugs might make it possible to stratify the
subgroups of patients, which might broaden our rather use of the right drug for the right patient at the right time.
limited therapeutic options in these disabling diseases. Additionally, personalized medicine could also drive
innovation for focused treatment approaches tailored to
Management the needs of specific subgroups of patients. The appar­
Personalized medicine in immune-​mediated neuro­ ent imminence of such promising developments offers
pathies, as the ultimate step in this development, would hope that progress in the treatment of these disabling
probably require a combination of new models, improved neurological diseases will accelerate in the near future.
tools to guide treatment responses and new biomarkers.
Advanced diagnostic techniques for screening and risk Published online xx xx xxxx

1. Goodfellow, J. A. & Willison, H. J. Guillain-​Barré 19. Martin Arias, L. H., Sanz, R., Sainz, M., Treceno, C. and anti-​glycolipid antibodies. Brain 118, 597–605
syndrome: a century of progress. Nat. Rev. Neurol. & Carvajal, A. Guillain-​Barré syndrome and influenza (1995).
12, 723–731 (2016). vaccines: a meta-​analysis. Vaccine 33, 3773–3778 38. Hughes, R. A. & Cornblath, D. R. Guillain-​Barré
This article provides an excellent review on the (2015). syndrome. Lancet 366, 1653–1666 (2005).
clinical and scientific developments in a dynamic 20. Poland, G. A. & Jacobsen, S. J. Influenza vaccine, 39. Godschalk, P. C. et al. The crucial role of Campylobacter
field in neuroimmunology. Guillain-​Barré syndrome, and chasing zero. Vaccine jejuni genes in anti-​ganglioside antibody induction in
2. Vallat, J. M., Sommer, C. & Magy, L. Chronic 30, 5801–5803 (2012). Guillain-​Barré syndrome. J. Clin. Invest. 114,
inflammatory demyelinating polyradiculoneuropathy: 21. Hocker, S., Nagarajan, E., Rubin, M. & Wijdicks, E. F. M. 1659–1665 (2004).
diagnostic and therapeutic challenges for a treatable Clinical factors associated with Guillain-​Barré syndrome 40. Sheikh, K. A., Deerinck, T. J., Ellisman, M. H. &
condition. Lancet Neurol. 9, 402–412 (2010). following surgery. Neurol. Clin. Pract. 8, 201–206 Griffin, J. W. The distribution of ganglioside-​like moieties
This comprehensive overview of CIDP discusses (2018). in peripheral nerves. Brain 122, 449–460 (1999).
how this disorder can be diagnosed and treated. 22. Stübgen, J. P. Tumor necrosis factor-​alpha antagonists 41. Yuki, N. et al. Carbohydrate mimicry between human
3. Latov, N. Diagnosis and treatment of chronic acquired and neuropathy. Muscle Nerve 37, 281–292 (2008). ganglioside GM1 and Campylobacter jejuni
demyelinating polyneuropathies. Nat. Rev. Neurol. 10, 23. Blum, S. & McCombe, P. A. Genetics of Guillain-Barré lipooligosaccharide causes Guillain-​Barré syndrome.
435–446 (2014). syndrome (GBS) and chronic inflammatory Proc. Natl Acad. Sci. USA 101, 11404–11409 (2004).
This extensive summary of the current knowledge demyelinating polyradiculoneuropathy (CIDP): 42. McGonigal, R. et al. Anti-​GD1a antibodies activate
on immune-​mediated neuropathies describes how current knowledge and future directions. J. Peripher. complement and calpain to injure distal motor nodes
to approach these heterogeneous disorders in Nerv. Syst. 19, 88–103 (2014). of Ranvier in mice. Brain 133, 1944–1960 (2010).
clinical practice. 24. Mevorach, D. et al. Therapy with eculizumab for patients 43. Rupp, A. et al. Motor nerve terminal destruction and
4. Sejvar, J. J., Baughman, A. L., Wise, M. & with CD59 p.Cys89Tyr mutation. Ann. Neurol. 80, regeneration following anti-​ganglioside antibody
Morgan, O. W. Population incidence of Guillain-Barré 708–717 (2016). and complement-​mediated injury: an in and ex vivo
syndrome: a systematic review and meta-​analysis. 25. Mahdi-​Rogers, M. & Hughes, R. A. Epidemiology of imaging study in the mouse. Exp. Neurol. 233,
Neuroepidemiology 36, 123–133 (2011). chronic inflammatory neuropathies in southeast 836–848 (2012).
5. McGrogan, A., Madle, G. C., Seaman, H. E. & England. Eur. J. Neurol. 21, 28–33 (2014). 44. He, L., Zhang, G., Liu, W., Gao, T. & Sheikh, K. A.
de Vries, C. S. The epidemiology of Guillain-​Barré 26. Cats, E. A. et al. Correlates of outcome and response Anti-​ganglioside antibodies induce nodal and axonal
syndrome worldwide — a systematic literature review. to IVIg in 88 patients with multifocal motor injury via Fcgamma receptor-​mediated inflammation.
Neuroepidemiology 32, 150–163 (2009). neuropathy. Neurology 75, 818–825 (2010). J. Neurosci. 35, 6770–6785 (2015).
6. Suryapranata, F. S. et al. Epidemiology of Guillain-​ 27. Miyashiro, A. et al. Are multifocal motor neuropathy 45. Clark, A. J. et al. Co-​cultures with stem cell-​derived
Barré syndrome in Aruba. Am. J. Trop. Med. Hyg. 94, patients underdiagnosed? An epidemiological survey human sensory neurons reveal regulators of
1380–1384 (2016). in Japan. Muscle Nerve 49, 357–361 (2014). peripheral myelination. Brain 140, 898–913 (2017).
7. Blum, S., Reddel, S., Spies, J. & McCombe, P. 28. Nobile-​Orazio, E., Bianco, M. & Nozza, A. Advances 46. Hafer-​Macko, C. E. et al. Immune attack on the Schwann
Clinical features of patients with Guillain-​Barré in the treatment of paraproteinemic neuropathy. cell surface in acute inflammatory demyelinating
syndrome at seven hospitals on the east coast of Curr. Treat Options Neurol. 19, 43 (2017). polyneuropathy. Ann. Neurol. 39, 625–635 (1996).
Australia. J. Peripher. Nerv. Syst. 18, 316–320 (2013). 29. Mygland, A. & Monstad, P. Chronic polyneuropathies 47. Griffin, J. W. et al. Early nodal changes in the acute
8. Nachamkin, I. et al. Patterns of Guillain-​Barré in Vest-​Agder, Norway. Eur. J. Neurol. 8, 157–165 motor axonal neuropathy pattern of the Guillain-​Barré
syndrome in children: results from a Mexican (2001). syndrome. J. Neurocytol. 25, 33–51 (1996).
population. Neurology 69, 1665–1671 (2007). 30. Raheja, D., Specht, C. & Simmons, Z. Paraproteinemic 48. Vural, A., Doppler, K. & Meinl, E. Autoantibodies
9. Bae, J. S. et al. Guillain-​Barré syndrome in Asia. neuropathies. Muscle Nerve 51, 1–13 (2015). against the node of Ranvier in seropositive chronic
J. Neurol. Neurosurg. Psychiatry 85, 907–913 (2014). 31. Mathey, E. K. et al. Chronic inflammatory demyelinating inflammatory demyelinating polyneuropathy:
10. Uncini, A., Manzoli, C., Notturno, F. & Capasso, M. polyradiculoneuropathy: from pathology to phenotype. diagnostic, pathogenic, and therapeutic relevance.
Pitfalls in electrodiagnosis of Guillain-​Barré syndrome J. Neurol. Neurosurg. Psychiatry 86, 973–985 (2015). Front. Immunol. 9, 1029 (2018).
subtypes. J. Neurol. Neurosurg. Psychiatry 81, This excellent review on the immunopathogenesis of 49. Uncini, A. & Kuwabara, S. Nodopathies of the
1157–1163 (2010). immune-​mediated neuropathies emphasizes the role peripheral nerve: an emerging concept. J. Neurol.
11. Mitsui, Y. et al. A multicentre prospective study of of humoral immune responses in these disorders. Neurosurg. Psychiatry 86, 1186–1195 (2015).
Guillain-​Barré syndrome in Japan: a focus on the 32. Meyer zu Horste, G., Hartung, H. P. & Kieseier, B. C. This article provides an excellent review on the
incidence of subtypes. J. Neurol. Neurosurg. From bench to bedside — experimental rationale for emerging concept of various clinical subclasses
Psychiatry 86, 110–114 (2015). immune-​specific therapies in the inflamed peripheral of chronic neuropathies defined by the presence of
12. Hauck, L. J. et al. Incidence of Guillain-​Barré syndrome nerve. Nat. Clin. Pract. Neurol. 3, 198–211 (2007). different autoantibodies.
in Alberta, Canada: an administrative data study. This comprehensive overview on existing animal 50. Shahrizaila, N. et al. Antibodies to single glycolipids
J. Neurol. Neurosurg. Psychiatry 79, 318–320 (2008). models for studying immune-​mediated and glycolipid complexes in Guillain-​Barré syndrome
13. Yuki, N. & Hartung, H. P. Guillain-​Barré syndrome. neuropathies discusses how insights from these subtypes. Neurology 83, 118–124 (2014).
N. Engl. J. Med. 366, 2294–2304 (2012). model systems help our understanding for 51. Rinaldi, S. et al. Antibodies to heteromeric glycolipid
14. Kuitwaard, K., van Koningsveld, R., Ruts, L., translational medicine in this disease entity. complexes in Guillain-​Barré syndrome. PLOS ONE 8,
Jacobs, B. C. & van Doorn, P. A. Recurrent Guillain-​ 33. Mäurer, M. & Gold, R. Animal models of immune- e82337 (2013).
Barré syndrome. J. Neurol. Neurosurg. Psychiatry 80, mediated neuropathies. Curr. Opin. Neurol. 15, This excellent summary of a complex topic
56–59 (2009). 617–622 (2002). deciphers the broad spectrum of potential
15. Jacobs, B. C. et al. The spectrum of antecedent 34. Meyer zu Horste, G. et al. Thymic epithelium determines autoantibodies in GBS.
infections in Guillain-​Barré syndrome: a case-​control a spontaneous chronic neuritis in Icam1(tm1Jcgr)NOD 52. Uncini, A. & Kuwabara, S. Electrodiagnostic criteria
study. Neurology 51, 1110–1115 (1998). mice. J. Immunol. 193, 2678–2690 (2014). for Guillain-​Barré syndrome: a critical revision and the
16. van den Berg, B. et al. Guillain-​Barré syndrome: 35. de Seze, J. et al. Chronic inflammatory demyelinating need for an update. Clin. Neurophysiol. 123,
pathogenesis, diagnosis, treatment and prognosis. polyradiculoneuropathy: a new animal model for new 1487–1495 (2012).
Nat. Rev. Neurol. 10, 469–482 (2014). therapeutic targets. Rev. Neurol. 172, 767–769 53. Samukawa, M. et al. Electrophysiological assessment
17. Cao-​Lormeau, V. M. et al. Guillain-​Barré Syndrome (2016). of Guillain-​Barré syndrome with both Gal-​C and
outbreak associated with Zika virus infection in French 36. Zautner, A. E. et al. Seroprevalence of ganglioside antibodies; tendency for demyelinating
Polynesia: a case-​control study. Lancet 387, campylobacteriosis and relevant post-​infectious type. J. Neuroimmunol. 301, 61–64 (2016).
1531–1539 (2016). sequelae. Eur. J. Clin. Microbiol. Infect. Dis. 33, 54. Goodfellow, J. A. & Willison, H. J. Antiganglioside,
18. Nascimento, O. J. M. & da Silva, I. R. F. Guillain-​Barré 1019–1027 (2014). antiganglioside-​complex, and antiglycolipid-​complex
syndrome and Zika virus outbreaks. Curr. Opin. 37. Ho, T. W. et al. Guillain-​Barré syndrome in northern antibodies in immune-​mediated neuropathies.
Neurol. 30, 500–507 (2017). China. Relationship to Campylobacter jejuni infection Curr. Opin. Neurol. 29, 572–580 (2016).

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 19

0123456789();
Primer

55. Makowska, A. et al. Immune responses to myelin chronic inflammatory demyelinating polyneuropathies. 102. Latov, N. et al. Plasma-​cell dyscrasia and peripheral
proteins in Guillain-​Barré syndrome. J. Neurol. Autoimmun. Rev. 13, 1070–1078 (2014). neuropathy with a monoclonal antibody to
Neurosurg. Psychiatry 79, 664–671 (2008). 78. Yan, W. X., Taylor, J., Andrias-​Kauba, S. & Pollard, J. D. peripheral-​nerve myelin. N. Engl. J. Med. 303,
56. Kwa, M. S., van Schaik, I. N., Brand, A., Baas, F. & Passive transfer of demyelination by serum or IgG from 618–621 (1980).
Vermeulen, M. Investigation of serum response to chronic inflammatory demyelinating polyneuropathy 103. Nobile-​Orazio, E. et al. Frequency and clinical
PMP22, connexin 32 and P(0) in inflammatory patients. Ann. Neurol. 47, 765–775 (2000). correlates of anti-​neural IgM antibodies in neuropathy
neuropathies. J. Neuroimmunol. 116, 220–225 79. Doppler, K. et al. Destruction of paranodal associated with IgM monoclonal gammopathy.
(2001). architecture in inflammatory neuropathy with anti-​ Ann. Neurol. 36, 416–424 (1994).
57. Sawai, S. et al. Moesin is a possible target molecule contactin-1 autoantibodies. J. Neurol. Neurosurg. 104. Takatsu, M. et al. Immunofluorescence study of patients
for cytomegalovirus-​related Guillain-​Barré syndrome. Psychiatry 86, 720–728 (2015). with neuropathy and IgM M proteins. Ann. Neurol. 18,
Neurology 83, 113–117 (2014). 80. Querol, L., Devaux, J., Rojas-​Garcia, R. & Illa, I. 173–181 (1985).
58. Miyaji, K., Devaux, J. & Yuki, N. Moesin is a possible Autoantibodies in chronic inflammatory neuropathies: 105. Vital, C. et al. Myelin modifications in 8 cases of
target molecule for cytomegalovirus-​related Guillain-​ diagnostic and therapeutic implications. Nat. Rev. peripheral neuropathy with Waldenstrom’s
Barré syndrome. Neurology 83, 2314 (2014). Neurol. 13, 533–547 (2017). macroglobulinemia and anti-​MAG activity.
59. Kwa, M. S. et al. Autoimmunoreactivity to Schwann This extensive summary on the role of Ultrastruct. Pathol. 21, 509–516 (1997).
cells in patients with inflammatory neuropathies. Brain autoantibodies in immune-​mediated neuropathies 106. Hays, A. P., Lee, S. S. & Latov, N. Immune reactive
126, 361–375 (2003). describes how to utilize this information in clinical C3d on the surface of myelin sheaths in neuropathy.
60. Devaux, J. J., Odaka, M. & Yuki, N. Nodal proteins practice. J. Neuroimmunol. 18, 231–244 (1988).
are target antigens in Guillain-​Barré syndrome. 81. Uncini, A., Zappasodi, F. & Notturno, F. 107. Jacobs, J. M. Morphological changes at paranodes in
J. Peripher. Nerv. Syst. 17, 62–71 (2012). Electrodiagnosis of GBS subtypes by a single study: IgM paraproteinaemic neuropathy. Microsc. Res. Tech.
61. Doppler, K. et al. Auto-​antibodies to contactin-​ not yet the squaring of the circle. J. Neurol. 34, 544–553 (1996).
associated protein 1 (Caspr) in two patients with Neurosurg. Psychiatry 86, 5–8 (2015). 108. Stalder, A. K. et al. Immunoglobulin M deposition
painful inflammatory neuropathy. Brain 139, 82. Cifuentes-​Diaz, C. et al. Nodes of Ranvier and paranodes in cutaneous nerves of anti-​myelin-associated
2617–2630 (2016). in chronic acquired neuropathies. PLOS ONE 6, e14533 glycoprotein polyneuropathy patients correlates with
62. Kiefer, R., Kieseier, B. C., Bruck, W., Hartung, H. P. (2011). axonal degeneration. J. Neuropathol. Exp. Neurol. 68,
& Toyka, K. V. Macrophage differentiation antigens in 83. Lonigro, A. & Devaux, J. J. Disruption of neurofascin 148–158 (2009).
acute and chronic autoimmune polyneuropathies. and gliomedin at nodes of Ranvier precedes 109. Hays, A. P., Latov, N., Takatsu, M. & Sherman, W. H.
Brain 121, 469–479 (1998). demyelination in experimental allergic neuritis. Brain Experimental demyelination of nerve induced by
63. Kuwabara, S., Nakajima, M., Matsuda, S. & Hattori, T. 132, 260–273 (2009). serum of patients with neuropathy and an anti-​MAG
Magnetic resonance imaging at the demyelinative 84. Koike, H. et al. Paranodal dissection in chronic IgM M-​protein. Neurology 37, 242–256 (1987).
foci in chronic inflammatory demyelinating inflammatory demyelinating polyneuropathy with 110. Tatum, A. H. Experimental paraprotein neuropathy,
polyneuropathy. Neurology 48, 874–877 (1997). anti-​neurofascin-155 and anti-​contactin-1 antibodies. demyelination by passive transfer of human IgM
64. Duggins, A. J. et al. Spinal root and plexus J. Neurol. Neurosurg. Psychiatry 88, 465–473 (2017). anti-​myelin-associated glycoprotein. Ann. Neurol. 33,
hypertrophy in chronic inflammatory demyelinating 85. Vallat, J. M. et al. Paranodal lesions in chronic 502–506 (1993).
polyneuropathy. Brain 122, 1383–1390 (1999). inflammatory demyelinating polyneuropathy 111. Ilyas, A. A., Quarles, R. H., Dalakas, M. C. & Brady, R. O.
65. Kiefer, R., Kieseier, B. C., Stoll, G. & Hartung, H. P. associated with anti-​neurofascin 155 antibodies. Polyneuropathy with monoclonal gammopathy:
The role of macrophages in immune-​mediated damage Neuromuscul. Disord. 27, 290–293 (2017). glycolipids are frequently antigens for IgM paraproteins.
to the peripheral nervous system. Prog. Neurobiol. 86. Kuwahara, M. et al. Clinical features of CIDP with Proc. Natl Acad. Sci. USA 82, 6697–6700 (1985).
64, 109–127 (2001). LM1-associated antibodies. J. Neurol. Neurosurg. 112. Kuwabara, S. et al. Hyperreflexia in axonal
66. Vital, A. et al. Chronic inflammatory demyelinating Psychiatry 84, 573–575 (2013). Guillain-Barré syndrome subsequent to Campylobacter
polyneuropathy associated with dysglobulinemia: 87. Delmont, E. et al. Autoantibodies to nodal isoforms of jejuni enteritis. J. Neurol. Sci. 199, 89–92 (2002).
a peripheral nerve biopsy study in 18 cases. neurofascin in chronic inflammatory demyelinating 113. Wakerley, B. R. & Yuki, N. Pharyngeal-​cervical-brachial
Acta Neuropathol. 100, 63–68 (2000). polyneuropathy. Brain 140, 1851–1858 (2017). variant of Guillain-​Barré syndrome. J. Neurol.
67. Van den Berg, L. H., Mollee, I., Wokke, J. H. & 88. Pestronk, A. et al. A treatable multifocal motor Neurosurg. Psychiatry 85, 339–344 (2014).
Logtenberg, T. Increased frequencies of HPRT mutant neuropathy with antibodies to GM1 ganglioside. 114. Wakerley, B. R. & Yuki, N. Mimics and chameleons
T lymphocytes in patients with Guillain-​Barré Ann. Neurol. 24, 73–78 (1988). in Guillain-​Barré and Miller Fisher syndromes.
syndrome and chronic inflammatory demyelinating 89. Cats, E. A. et al. Multifocal motor neuropathy: Pract. Neurol. 15, 90–99 (2014).
polyneuropathy: further evidence for a role of T cells in association of anti-​GM1 IgM antibodies with clinical 115. Asbury, A. K. & Cornblath, D. R. Assessment of
the etiopathogenesis of peripheral demyelinating features. Neurology 75, 1961–1967 (2010). current diagnostic criteria for Guillain-​Barré syndrome.
diseases. J. Neuroimmunol. 58, 37–42 (1995). 90. Susuki, K. et al. Anti-​GM1 antibodies cause Ann. Neurol. 27, S21–S24 (1990).
68. Chi, L. J. et al. Distribution of Th17 cells and Th1 cells complement-​mediated disruption of sodium channel 116. Sejvar, J. J. et al. Guillain-​Barré syndrome and Fisher
in peripheral blood and cerebrospinal fluid in chronic clusters in peripheral motor nerve fibers. J. Neurosci. syndrome: case definitions and guidelines for
inflammatory demyelinating polyradiculoneuropathy. 27, 3956–3967 (2007). collection, analysis, and presentation of immunization
J. Peripher. Nerv. Syst. 15, 345–356 (2010). 91. Piepers, S. et al. IVIg inhibits classical pathway activity safety data. Vaccine 29, 599–612 (2011).
69. Staudt, M., Diederich, J. M., Meisel, C., Meisel, A. & and anti-​GM1 IgM-​mediated complement deposition 117. Fokke, C. et al. Diagnosis of Guillain-​Barré syndrome
Klehmet, J. Differences in peripheral myelin antigen-​ in MMN. J. Neuroimmunol. 229, 256–262 (2010). and validation of Brighton criteria. Brain 137, 33–43
specific T cell responses and T memory subsets in 92. Yuki, N., Watanabe, H., Nakajima, T. & Spath, P. J. (2014).
atypical versus typical CIDP. BMC Neurol. 17, 81 IVIG blocks complement deposition mediated by anti-​ 118. Hadden, R. D. et al. Electrophysiological classification
(2017). GM1 antibodies in multifocal motor neuropathy. of Guillain-​Barré syndrome: clinical associations and
70. Sanvito, L., Makowska, A., Gregson, N., Nemni, R. & J. Neurol. Neurosurg. Psychiatry 82, 87–91 (2011). outcome. Plasma Exchange/Sandoglobulin Guillain-​
Hughes, R. A. Circulating subsets and CD4(+)CD25(+) 93. Vlam, L. et al. Complement activity is associated with Barré Syndrome Trial Group. Ann. Neurol. 44,
regulatory T cell function in chronic inflammatory disease severity in multifocal motor neuropathy. 780–788 (1998).
demyelinating polyradiculoneuropathy. Autoimmunity Neurol. Neuroimmunol. Neuroinflamm. 2, e119 (2015). 119. Albers, J. W., Donofrio, P. D. & McGonagle, T. K.
42, 667–677 (2009). 94. Harschnitz, O. et al. Autoantibody pathogenicity in Sequential electrodiagnostic abnormalities in acute
71. Sanvito, L. et al. Humoral and cellular immune a multifocal motor neuropathy induced pluripotent inflammatory demyelinating polyradiculoneuropathy.
responses to myelin protein peptides in chronic stem cell-​derived model. Ann. Neurol. 80, 71–88 Muscle Nerve 8, 528–539 (1985).
inflammatory demyelinating polyradiculoneuropathy. (2016). 120. Orlikowski, D. et al. Guillain-​Barré syndrome following
J. Neurol. Neurosurg. Psychiatry 80, 333–338 95. Taylor, B. V. et al. Multifocal motor neuropathy: primary cytomegalovirus infection: a prospective
(2009). pathologic alterations at the site of conduction block. cohort study. Clin. Infect. Dis. 52, 837–844 (2011).
72. Schneider-​Hohendorf, T. et al. CD8+ T cell J. Neuropathol. Exp. Neurol. 63, 129–137 (2004). 121. Hadden, R. D. et al. Preceding infections, immune
immunity in chronic inflammatory demyelinating 96. Garg, N. et al. Motor unit remodelling in multifocal factors, and outcome in Guillain-​Barré syndrome.
polyradiculoneuropathy. Neurology 78, 402–408 motor neuropathy: the importance of axonal loss. Neurology 56, 758–765 (2001).
(2012). Clin. Neurophysiol. 128, 2022–2028 (2017). 122. Willison, H. J., Jacobs, B. C. & van Doorn, P. A.
73. Klehmet, J. et al. Circulating lymphocyte and T 97. Kaji, R. et al. Pathological findings at the site of Guillain-​Barré syndrome. Lancet 388, 717–727
memory subsets in glucocorticosteroid versus IVIG conduction block in multifocal motor neuropathy. (2016).
treated patients with CIDP. J. Neuroimmunol. 283, Ann. Neurol. 33, 152–158 (1993). 123. Kaida, K. et al. Ganglioside complexes as new target
17–22 (2015). 98. Oh, S. J., Claussen, G. C., Odabasi, Z. & Palmer, C. P. antigens in Guillain-​Barré syndrome. Ann. Neurol. 56,
74. Pollard, J. D., McCombe, P. A., Baverstock, J., Multifocal demyelinating motor neuropathy: 567–571 (2004).
Gatenby, P. A. & McLeod, J. G. Class II antigen pathologic evidence of ‘inflammatory demyelinating 124. Galazky, I. et al. Guillain-​Barré/CIDP-​like neuropathy
expression and T lymphocyte subsets in chronic polyradiculoneuropathy’. Neurology 45, 1828–1832 in two parkinsonian patients following intestinal
inflammatory demyelinating polyneuropathy. (1995). levodopa/carbidopa treatment. Parkinsonism Relat.
J. Neuroimmunol. 13, 123–134 (1986). 99. Shimizu, F. et al. Sera from patients with multifocal Disord. 20, 125–127 (2014).
75. Meyer zu Horste, G. et al. Mouse Schwann cells motor neuropathy disrupt the blood-​nerve barrier. 125. Prommel, P., Pilgram-​Pastor, S., Sitter, H., Buhk, J. H.
activate MHC class I and II restricted T cell responses, J. Neurol. Neurosurg. Psychiatry 85, 526–537 (2014). & Strik, H. Neoplastic meningitis: how MRI and CSF
but require external peptide processing for MHC 100. Priori, A. et al. Distinctive abnormalities of motor cytology are influenced by CSF cell count and tumor
class II presentation. Neurobiol. Dis. 37, 483–490 axonal strength-​duration properties in multifocal type. ScientificWorldJournal 2013, 248072 (2013).
(2010). motor neuropathy and in motor neurone disease. 126. van Koningsveld, R. et al. A clinical prognostic scoring
76. Mausberg, A. K. et al. Recovery of the T cell repertoire Brain 125, 2481–2490 (2002). system for Guillain-​Barré syndrome. Lancet Neurol. 6,
in CIDP by IV immunoglobulins. Neurology 80, 101. Kiernan, M. C., Guglielmi, J. M., Kaji, R., Murray, N. M. 589–594 (2007).
296–303 (2013). & Bostock, H. Evidence for axonal membrane This study is one of the first promising attempts to
77. Lim, J. P., Devaux, J. & Yuki, N. Peripheral nerve hyperpolarization in multifocal motor neuropathy with introduce clinical predictive markers in a potentially
proteins as potential autoantigens in acute and conduction block. Brain 125, 664–675 (2002). life-​threatening disease.

20 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

127. Walgaard, C. et al. Prediction of respiratory 150. Mathis, S., Magy, L., Diallo, L., Boukhris, S. & 176. Misawa, S. et al. Safety and efficacy of eculizumab in
insufficiency in Guillain-​Barré syndrome. Ann. Neurol. Vallat, J. M. Amyloid neuropathy mimicking chronic Guillain-​Barré syndrome: a multicentre, double-​blind,
67, 781–787 (2010). inflammatory demyelinating polyneuropathy. randomised phase 2 trial. Lancet Neurol. 17,
128. Yamagishi, Y. et al. Markers for Guillain-​Barré Muscle Nerve 45, 26–31 (2012). 519–529 (2018).
syndrome with poor prognosis: a multi-​center study. 151. Cortese, A. et al. Misdiagnoses of transthyretin 177. Koller, H., Kieseier, B. C., Jander, S. & Hartung, H. P.
J. Peripher. Nerv. Syst. 22, 433–439 (2017). amyloidosis: a clinical and electrodiagnostic study. Chronic inflammatory demyelinating polyneuropathy.
129. Koski, C. L. et al. Derivation and validation of Orphanet J. Rare Dis. 10, O13 (2015). N. Engl. J. Med. 352, 1343–1356 (2005).
diagnostic criteria for chronic inflammatory 152. Manji, H. Drug-​induced neuropathies. Handb. Clin. 178. Dyck, P. J. et al. Chronic inflammatory
demyelinating polyneuropathy. J. Neurol. Sci. 277, Neurol. 115, 729–742 (2013). polyradiculoneuropathy. Mayo Clin. Proc. 50,
1–8 (2009). 153. Oh, S. J., Kurokawa, K., de Almeida, D. F., Ryan, H. F. Jr 621–637 (1975).
130. Barohn, R. J., Kissel, J. T., Warmolts, J. R. & & Claussen, G. C. Subacute inflammatory demyelinating 179. Dyck, P. J. et al. Prednisone improves chronic
Mendell, J. R. Chronic inflammatory demyelinating polyneuropathy. Neurology 61, 1507–1512 (2003). inflammatory demyelinating polyradiculoneuropathy
polyradiculoneuropathy — clinical characteristics, 154. Vlam, L. et al. Multifocal motor neuropathy: diagnosis, more than no treatment. Ann. Neurol. 11, 136–141
course, and recommendations for diagnostic criteria. pathogenesis and treatment strategies. Nat. Rev. (1982).
Arch. Neurol. 46, 878–884 (1989). Neurol. 8, 48–58 (2012). 180. Hughes, R. A., Mehndiratta, M. M. & Rajabally, Y. A.
131. Boukhris, S., Magy, L., Khalil, M., Sindou, P. & 155. Stangel, M. et al. Treatment of patients with multifocal Corticosteroids for chronic inflammatory
Vallat, J. M. Pain as the presenting symptom of chronic motor neuropathy with immunoglobulins in clinical demyelinating polyradiculoneuropathy. Cochrane
inflammatory demyelinating polyradiculoneuropathy practice: the SIGNS registry. Ther. Adv. Neurol. Disord. Database Syst. Rev. 11, CD002062 (2017).
(CIDP). J. Neurol. Sci. 254, 33–38 (2007). 9, 165–179 (2016). 181. Gold, R. & Kieseier, B. C. Therapy of immune
132. Kuitwaard, K., Bos-​Eyssen, M. E., 156. Lambrecq, V., Krim, E., Rouanet-​Larriviere, M. neuropathies with intravenous immunoglobulins.
Blomkwist-Markens, P. H. & van Doorn, P. A. & Lagueny, A. Sensory loss in multifocal motor J. Neurol. 253(Suppl. 5), V59–V63 (2006).
Recurrences, vaccinations and long-​term symptoms neuropathy: a clinical and electrophysiological study. 182. Hughes, R. A. et al. Intravenous immune globulin
in GBS and CIDP. J. Peripher. Nerv. Syst. 14, Muscle Nerve 39, 131–136 (2009). (10% caprylate-​chromatography purified) for the
310–315 (2009). 157. Van den Berg-​Vos, R. M. et al. Multifocal inflammatory treatment of chronic inflammatory demyelinating
133. Mendell, J. R. et al. Evidence for central nervous demyelinating neuropathy: a distinct clinical entity? polyradiculoneuropathy (ICE study): a randomised
system demyelination in chronic inflammatory Neurology 54, 26–32 (2000). placebo-​controlled trial. Lancet Neurol. 7, 136–144
demyelinating polyradiculoneuropathy. Neurology 37, 158. Joint Task Force of the EFNS and the PNS. European (2008).
1291–1294 (1987). Federation of Neurological Societies/Peripheral Nerve This article presents the first and largest
134. Pineda, A. A. et al. A distinct subgroup of chronic Society guideline on management of multifocal motor conducted trial in CIDP demonstrating long-​term
inflammatory demyelinating polyneuropathy with CNS neuropathy. Report of a joint task force of the effects of IVIG in patients with this disorder.
demyelination and a favorable response to European Federation of Neurological Societies and the 183. Lopate, G., Pestronk, A. & Al-​Lozi, M. Treatment of
immunotherapy. J. Neurol. Sci. 255, 1–6 (2007). Peripheral Nerve Society — first revision. J. Peripher. chronic inflammatory demyelinating polyneuropathy
135. Rotta, F. T. et al. The spectrum of chronic inflammatory Nerv. Syst. 15, 295–301 (2010). with high-​dose intermittent intravenous
demyelinating polyneuropathy. J. Neurol. Sci. 173, 159. Delmont, E. et al. Multifocal motor neuropathy with methylprednisolone. Arch. Neurol. 62, 249–254
129–139 (2000). and without conduction block: a single entity? (2005).
136. Saperstein, D. S., Katz, J. S., Amato, A. A. & Neurology 67, 592–596 (2006). 184. Nobile-​Orazio, E. et al. Frequency and time to relapse
Barohn, R. J. Clinical spectrum of chronic acquired 160. Meuth, S. G. & Kleinschnitz, C. Multifocal motor after discontinuing 6-month therapy with IVIg or
demyelinating polyneuropathies. Muscle Nerve 24, neuropathy: update on clinical characteristics, pulsed methylprednisolone in CIDP. J. Neurol.
311–324 (2001). pathophysiological concepts and therapeutic options. Neurosurg. Psychiatry 86, 729–734 (2015).
137. Joint Task Force of the EFNS and the PNS. European Eur. Neurol. 63, 193–204 (2010). 185. Nobile-​Orazio, E. et al. Intravenous immunoglobulin
Federation of Neurological Societies/Peripheral Nerve 161. Kyle, R. A. et al. Long-​term follow-​up of monoclonal versus intravenous methylprednisolone for chronic
Society Guideline on management of chronic gammopathy of undetermined significance. N. Engl. inflammatory demyelinating polyradiculoneuropathy:
inflammatory demyelinating polyradiculoneuropathy: J. Med. 378, 241–249 (2018). a randomised controlled trial. Lancet Neurol. 11,
report of a joint task force of the European Federation 162. Hoffman, E. M. et al. Impairments and comorbidities 493–502 (2012).
of Neurological Societies and the Peripheral Nerve of polyneuropathy revealed by population-​based 186. Vrancken, A. F., Notermans, N. C., Jansen, G. H.,
Society — first revision. J. Peripher. Nerv. Syst. 15, analyses. Neurology 84, 1644–1651 (2015). Wokke, J. H. & Said, G. Progressive idiopathic
1–9 (2010). 163. Zis, P., Sarrigiannis, P. G., Rao, D. G., Hewamadduma, C. axonal neuropathy—a comparative clinical and
138. Lewis, R. A., Sumner, A. J., Brown, M. J. & Asbury, A. K. & Hadjivassiliou, M. Chronic idiopathic axonal histopathological study with vasculitic neuropathy.
Multifocal demyelinating neuropathy with persistent polyneuropathy: a systematic review. J. Neurol. 263, J. Neurol. 251, 269–278 (2004).
conduction block. Neurology 32, 958–964 (1982). 1903–1910 (2016). 187. Markvardsen, L. H. et al. Subcutaneous
139. Breiner, A. & Brannagan, T. H. 3rd. Comparison of 164. Kuijf, M. L. et al. Detection of anti-​MAG antibodies in immunoglobulin in responders to intravenous
sensitivity and specificity among 15 criteria for polyneuropathy associated with IgM monoclonal therapy with chronic inflammatory demyelinating
chronic inflammatory demyelinating polyneuropathy. gammopathy. Neurology 73, 688–695 (2009). polyradiculoneuropathy. Eur. J. Neurol. 20, 836–842
Muscle Nerve 50, 40–46 (2014). 165. Trojaborg, W., Hays, A. P., van den Berg, L., (2013).
140. Latov, N. et al. Timing and course of clinical response Younger, D. S. & Latov, N. Motor conduction parameters 188. RMC Trial Group. Randomised controlled trial of
to intravenous immunoglobulin in chronic in neuropathies associated with anti-​MAG antibodies methotrexate for chronic inflammatory demyelinating
inflammatory demyelinating polyradiculoneuropathy. and other types of demyelinating and axonal polyradiculoneuropathy (RMC trial): a pilot,
Arch. Neurol. 67, 802–807 (2010). neuropathies. Muscle Nerve 18, 730–735 (1995). multicentre study. Lancet Neurol. 8, 158–164
141. Tackenberg, B. et al. Classifications and treatment 166. Nobile-​Orazio, E. IgM paraproteinaemic neuropathies. (2009).
responses in chronic immune-​mediated demyelinating Curr. Opin. Neurol. 17, 599–605 (2004). 189. van Schaik, I. N. et al. Subcutaneous immunoglobulin
polyneuropathy. Neurology 68, 1622–1629 (2007). 167. Willison, H. J. et al. The clinical and laboratory features for maintenance treatment in chronic inflammatory
142. French CIDP Study Group. Recommendations on of chronic sensory ataxic neuropathy with anti-​ demyelinating polyneuropathy (PATH): a randomised,
diagnostic strategies for chronic inflammatory disialosyl IgM antibodies. Brain 124, 1968–1977 double-​blind, placebo-​controlled, phase 3 trial.
demyelinating polyradiculoneuropathy. J. Neurol. (2001). Lancet Neurol. 17, 35–46 (2018).
Neurosurg. Psychiatry 79, 115–118 (2008). 168. Steck, A., Yuki, N. & Graus, F. Antibody testing in 190. Cocito, D. et al. Subcutaneous immunoglobulin in
143. Capek, S., Amrami, K. K., Dyck, P. J. & Spinner, R. J. peripheral nerve disorders. Handb. Clin. Neurol. 115, CIDP and MMN: a different long-​term clinical
Targeted fascicular biopsy of the sciatic nerve and its 189–212 (2013). response? J. Neurol. Neurosurg. Psychiatry 87,
major branches: rationale and operative technique. 169. Kieseier, B. C. et al. Advances in understanding and 791–793 (2015).
Neurosurg. Focus 39, E12 (2015). treatment of immune-​mediated disorders of the 191. Leussink, V. I., Hartung, H. P., Kieseier, B. C. &
144. Stettner, M. et al. Corneal confocal microscopy in peripheral nervous system. Muscle Nerve 30, Stettner, M. Subcutaneous immunoglobulins in
chronic inflammatory demyelinating polyneuropathy. 131–156 (2004). the treatment of chronic immune-​mediated
Ann. Clin. Transl Neurol. 3, 88–100 (2016). 170. Hughes, R. A., Swan, A. V. & van Doorn, P. A. neuropathies. Ther. Adv. Neurol. Disord. 9, 336–343
145. Ruts, L., Drenthen, J., Jacobs, B. C. & van Doorn, P. A. Intravenous immunoglobulin for Guillain-​Barré (2016).
Distinguishing acute-​onset CIDP from fluctuating syndrome. Cochrane Database Syst. Rev. 9, 192. Hughes, R. A. et al. Intramuscular interferon
Guillain-​Barré syndrome: a prospective study. CD002063 (2004). beta-1a in chronic inflammatory demyelinating
Neurology 74, 1680–1686 (2010). 171. The French Cooperative Group on Plasma Exchange polyradiculoneuropathy. Neurology 74, 651–657
146. Alessandro, L. et al. Differences between acute-​onset in Guillain-​Barré Syndrome. Appropriate number of (2010).
chronic inflammatory demyelinating polyneuropathy plasma exchanges in Guillain-​Barre syndrome. 193. Fialho, D., Chan, Y. C., Allen, D. C., Reilly, M. M. &
(A-​CIDP) and acute inflammatory demyelinating Ann. Neurol. 41, 298–306 (1997). Hughes, R. A. Treatment of chronic inflammatory
polyneuropathy (AIDP) in adult patients. J. Peripher. 172. Hughes, R. A. Treatment of peripheral nerve disorders. demyelinating polyradiculoneuropathy with
Nerv. Syst. 23, 154–158 (2018). Curr. Opin. Neurol. 18, 554–556 (2005). methotrexate. J. Neurol. Neurosurg. Psychiatry 77,
147. Markowitz, J. A., Jeste, S. S. & Kang, P. B. 173. van Doorn, P. A., Kuitwaard, K. & Jacobs, B. C. Serum 544–547 (2006).
Child neurology: chronic inflammatory demyelinating IgG levels as biomarkers for optimizing IVIg therapy 194. Mahattanakul, W., Crawford, T. O., Griffin, J. W.,
polyradiculoneuropathy in children. Neurology 71, in CIDP. J. Peripher. Nerv. Syst. 16 (Suppl. 1), 38–40 Goldstein, J. M. & Cornblath, D. R. Treatment of
e74–e78 (2008). (2011). chronic inflammatory demyelinating polyneuropathy
148. Rajabally, Y. A., Stettner, M., Kieseier, B. C., 174. Hughes, R. A., Brassington, R., Gunn, A. A. & with cyclosporin-​A. J. Neurol. Neurosurg. Psychiatry
Hartung, H. P. & Malik, R. A. CIDP and other van Doorn, P. A. Corticosteroids for Guillain-​Barré 60, 185–187 (1996).
inflammatory neuropathies in diabetes — diagnosis and syndrome. Cochrane Database Syst. Rev. 10, 195. Matsuda, M., Hoshi, K., Gono, T., Morita, H. &
management. Nat. Rev. Neurol. 13, 599–611 (2017). CD001446 (2016). Ikeda, S. Cyclosporin A in treatment of refractory
149. Mohamed, A., Davies, L. & Pollard, J. D. Conduction 175. Davidson, A. I. et al. Inhibition of complement in patients with chronic inflammatory demyelinating
block in vasculitic neuropathy. Muscle Nerve 21, Guillain-​Barré syndrome: the ICA-​GBS study. polyradiculoneuropathy. J. Neurol. Sci. 224, 29–35
1084–1088 (1998). J. Peripher. Nerv. Syst. 22, 4–12 (2017). (2004).

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 21

0123456789();
Primer

196. Gorson, K. C., Amato, A. A. & Ropper, A. H. Efficacy of 219. Renaud, S. et al. Rituximab in the treatment of binding of anti-​contactin-1 auto-​antibodies is modified
mycophenolate mofetil in patients with chronic polyneuropathy associated with anti-​MAG antibodies. by immunoglobulins. Exp. Neurol. 287, 84–90
immune demyelinating polyneuropathy. Neurology Muscle Nerve 27, 611–615 (2003). (2017).
63, 715–717 (2004). 220. Renaud, S. et al. High-​dose rituximab and anti-​MAG- 242. Querol, L. et al. Rituximab in treatment-​resistant
197. Mahdi-​Rogers, M., Brassington, R., Gunn, A. A., associated polyneuropathy. Neurology 66, 742–744 CIDP with antibodies against paranodal proteins.
van Doorn, P. A. & Hughes, R. A. C. Immunomodulatory (2006). Neurol. Neuroimmunol. Neuroinflamm. 2, e149
treatment other than corticosteroids, immunoglobulin 221. Svahn, J. et al. Anti-​MAG antibodies in 202 patients: (2015).
and plasma exchange for chronic inflammatory clinicopathological and therapeutic features. J. Neurol. 243. Querol, L. et al. Neurofascin IgG4 antibodies in CIDP
demyelinating polyradiculoneuropathy. Cochrane Neurosurg. Psychiatry 89, 499–505 (2017). associate with disabling tremor and poor response to
Database Syst. Rev. 5, CD003280 (2017). 222. Gono, T. et al. Rituximab therapy in chronic IVIg. Neurology 82, 879–886 (2014).
198. Gladstone, D. E., Prestrud, A. A. & Brannagan, T. H. inflammatory demyelinating polyradiculoneuropathy 244. Ogata, H. et al. Characterization of IgG4 anti-​
3rd. High-​dose cyclophosphamide results in long-​term with anti-​SGPG IgM antibody. J. Clin. Neurosci. 13, neurofascin 155 antibody-​positive polyneuropathy.
disease remission with restoration of a normal quality 683–687 (2006). Ann. Clin. Transl Neurol. 2, 960–971 (2015).
of life in patients with severe refractory chronic 223. Graham, R. C., Hughes, R. A. & White, C. M. 245. Chan, Y. C., Allen, D. C., Fialho, D., Mills, K. R. &
inflammatory demyelinating polyneuropathy. A prospective study of physiotherapist prescribed Hughes, R. A. Predicting response to treatment in
J. Peripher. Nerv. Syst. 10, 11–16 (2005). community based exercise in inflammatory chronic inflammatory demyelinating
199. McHutchison, J. G. et al. Interferon alfa-2b alone or peripheral neuropathy. J. Neurol. 254, 228–235 polyradiculoneuropathy. J. Neurol. Neurosurg.
in combination with ribavirin as initial treatment for (2007). Psychiatry 77, 114–116 (2006).
chronic hepatitis C. N. Engl. J. Med. 339, 1485–1492 224. Khan, F. et al. Outcomes of high- and low-​intensity 246. Ng, J. K. et al. Neurofascin as a target for autoantibodies
(1998). rehabilitation programme for persons in chronic in peripheral neuropathies. Neurology 79, 2241–2248
200. Leitch, M. M., Sherman, W. H. & Brannagan, T. H. 3rd. phase after Guillain-​Barré syndrome: a randomized (2012).
Fludarabine in the treatment of refractory chronic controlled trial. J. Rehabil. Med. 43, 638–646 247. Uncini, A. & Vallat, J. M. Autoimmune nodo-​
inflammatory demyelinating neuropathies. J. Clin. (2011). paranodopathies of peripheral nerve: the concept is
Neuromuscul. Dis. 17, 1–5 (2015). 225. Markvardsen, L. H. et al. Resistance training and gaining ground. J. Neurol. Neurosurg. Psychiatry 89,
201. Dyck, P. J., O’Brien, P., Swanson, C., Low, P. & Daube, J. aerobic training improve muscle strength and aerobic 627–635 (2017).
Combined azathioprine and prednisone in chronic capacity in chronic inflammatory demyelinating 248. Misawa, S. et al. Serum levels of tumor necrosis
inflammatory-​demyelinating polyneuropathy. polyneuropathy. Muscle Nerve 57, 70–76 (2017). factor-alpha in chronic inflammatory demyelinating
Neurology 35, 1173–1176 (1985). 226. Garssen, M. P. et al. Physical training and fatigue, polyneuropathy. Neurology 56, 666–669 (2001).
202. Kieseier, B. C. & Stuve, O. A critical appraisal of fitness, and quality of life in Guillain-​Barré syndrome 249. Hartung, H. P., Reiners, K., Schmidt, B., Stoll, G. &
treatment decisions in multiple sclerosis — old versus and CIDP. Neurology 63, 2393–2395 (2004). Toyka, K. V. Serum interleukin-2 concentrations in
new. Nat. Rev. Neurol. 7, 255–262 (2011). 227. Cocito, D. et al. Multifocal motor neuropathy during Guillain-​Barré syndrome and chronic idiopathic
203. Hirst, C., Raasch, S., Llewelyn, G. & Robertson, N. treatment with infliximab. J. Peripher. Nerv. Syst. 10, demyelinating polyradiculoneuropathy: comparison
Remission of chronic inflammatory demyelinating 386–387 (2005). with other neurological diseases of presumed
polyneuropathy after alemtuzumab (Campath 1H). 228. Wilson, J. R., Conwit, R. A., Eidelman, B. H., immunopathogenesis. Ann. Neurol. 30, 48–53
J. Neurol. Neurosurg. Psychiatry 77, 800–802 Starzl, T. & Abu-​Elmagd, K. Sensorimotor neuropathy (1991).
(2006). resembling CIDP in patients receiving FK506. 250. Willison, H. J. & Yuki, N. Peripheral neuropathies and
204. Marsh, E. A. et al. Alemtuzumab in the treatment of Muscle Nerve 17, 528–532 (1994). anti-​glycolipid antibodies. Brain 125, 2591–2625
IVIG-​dependent chronic inflammatory demyelinating 229. Merkies, I. S. et al. Understanding the consequences (2002).
polyneuropathy. J. Neurol. 257, 913–919 (2010). of chronic inflammatory demyelinating 251. Wakerley, B. R., Uncini, A. & Yuki, N. Guillain-​Barré
205. Engelhardt, B. & Ransohoff, R. M. The ins and outs of polyradiculoneuropathy from impairments to activity and Miller Fisher syndromes — new diagnostic
T-​lymphocyte trafficking to the CNS: anatomical sites and participation restrictions and reduced quality classification. Nat. Rev. Neurol. 10, 537–544 (2014).
and molecular mechanisms. Trends Immunol. 26, of life: the ICE study. J. Peripher. Nerv. Syst. 15, 252. Harschnitz, O. et al. MMN: from immunological
485–495 (2005). 208–215 (2010). cross-​talk to conduction block. J. Clin. Immunol. 34,
206. Vallat, J. M. et al. Natalizumab as a disease-​modifying 230. Forsberg, A., Press, R. & Holmqvist, L. W. Residual S112–S119 (2014).
therapy in chronic inflammatory demyelinating disability 10 years after falling ill in Guillain-​Barré 253. Kuwabara, S. et al. IgG anti-​GM1 antibody is
polyneuropathy — a report of three cases. Eur. Neurol. syndrome: a prospective follow-​up study. J. Neurol. Sci. associated with reversible conduction failure and
73, 294–302 (2015). 317, 74–79 (2012). axonal degeneration in Guillain-​Barré syndrome.
207. Wolf, C. et al. Natalizumab treatment in a patient with 231. White, C. M., Hadden, R. D., Robert-​Lewis, S. F., Ann. Neurol. 44, 202–208 (1998).
chronic inflammatory demyelinating polyneuropathy. McCrone, P. R. & Petty, J. L. Observer blind 254. Ho, T. W. et al. Anti-​GD1a antibody is associated with
Arch. Neurol. 67, 881–883 (2010). randomised controlled trial of a tailored home exercise axonal but not demyelinating forms of Guillain-​Barré
208. Leger, J. M., Guimaraes-​Costa, R. & Muntean, C. programme versus usual care in people with stable syndrome. Ann. Neurol. 45, 168–173 (1999).
Immunotherapy in peripheral neuropathies. inflammatory immune mediated neuropathy. 255. Yuki, N., Kuwabara, S., Koga, M. & Hirata, K.
Neurotherapeutics 13, 96–107 (2016). BMC Neurol. 15, 147 (2015). Acute motor axonal neuropathy and acute motor-​
209. Koski, C. L. Initial and long-​term management of 232. Merkies, I. S. & Kieseier, B. C. Fatigue, pain, sensory axonal neuropathy share a common
autoimmune neuropathies. CNS Drugs 19, anxiety and depression in Guillain-​Barré syndrome immunological profile. J. Neurol. Sci. 168, 121–126
1033–1048 (2005). and chronic inflammatory demyelinating (1999).
210. Umapathi, T., Hughes, R. A., Nobile-​Orazio, E. polyradiculoneuropathy. Eur. Neurol. 75, 199–206 256. Nagashima, T., Koga, M., Odaka, M., Hirata, K. &
& Leger, J. M. Immunosuppressive treatment for (2016). Yuki, N. Continuous spectrum of pharyngeal-​cervical-
multifocal motor neuropathy. Cochrane Database 233. Merkies, I. S., Schmitz, P. I., Samijn, J. P., brachial variant of Guillain-​Barré syndrome.
Syst. Rev. 2, CD003217 (2002). van der Meche, F. G. & van Doorn, P. A. Fatigue in Arch. Neurol. 64, 1519–1523 (2007).
211. Kieseier, B. C., Dalakas, M. C. & Hartung, H. P. immune-mediated polyneuropathies. European 257. van Schaik, I. N., Bossuyt, P. M., Brand, A. &
Immune mechanisms in chronic inflammatory Inflammatory Neuropathy Cause and Treatment Vermeulen, M. Diagnostic value of GM1 antibodies
demyelinating neuropathy. Neurology 59, S7–S12 (INCAT) Group. Neurology 53, 1648–1654 (1999). in motor neuron disorders and neuropathies:
(2002). 234. Drory, V. E., Bronipolsky, T., Bluvshtein, V., Catz, A. a meta-analysis. Neurology 45, 1570–1577 (1995).
212. Nobile-​Orazio, E., Cappellari, A. & Priori, A. & Korczyn, A. D. Occurrence of fatigue over 20 years 258. Garcia-​Santibanez, R. et al. CANOMAD and other
Multifocal motor neuropathy: current concepts after recovery from Guillain-​Barré syndrome. chronic ataxic neuropathies with disialosyl antibodies
and controversies. Muscle Nerve 31, 663–680 J. Neurol. Sci. 316, 72–75 (2012). (CANDA). J. Neurol. 265, 1402–1409 (2018).
(2005). 235. Renoir, T., Hasebe, K. & Gray, L. Mind and body: how 259. Querol, L. et al. Antibodies to contactin-1 in chronic
213. Federico, P., Zochodne, D. W., Hahn, A. F., the health of the body impacts on neuropsychiatry. inflammatory demyelinating polyneuropathy.
Brown, W. F. & Feasby, T. E. Multifocal motor Front. Pharmacol. 4, 158 (2013). Ann. Neurol. 73, 370–380 (2013).
neuropathy improved by IVIg: randomized, double-​ 236. Kuitwaard, K., Ruts, L., van Doorn, P. A. & 260. Miura, Y. et al. Contactin 1 IgG4 associates to chronic
blind, placebo-​controlled study. Neurology 55, van der Pol, W. L. Individual patients who experienced inflammatory demyelinating polyneuropathy with
1256–1262 (2000). both Guillain-​Barré syndrome and CIDP. J. Peripher. sensory ataxia. Brain 138, 1484–1491 (2015).
214. Harbo, T. et al. Subcutaneous versus intravenous Nerv. Syst. 14, 66–68 (2009). 261. Devaux, J. J. et al. Neurofascin-155 IgG4 in chronic
immunoglobulin in multifocal motor neuropathy: 237. Draak, T. H. P., Faber, C. G. & Merkies, I. S. J. inflammatory demyelinating polyneuropathy.
a randomized, single-​blinded cross-​over trial. Quality of life in inflammatory neuropathies: the Neurology 86, 800–807 (2016).
Eur. J. Neurol. 16, 631–638 (2009). IN-QoL. J. Neurol. Neurosurg. Psychiatry 89, 262. Hiraga, A. et al. Patterns and serial changes in
215. Harbo, T., Andersen, H. & Jakobsen, J. Long-​term 256–262 (2018). electrodiagnostic abnormalities of axonal Guillain-​
therapy with high doses of subcutaneous 238. Mahdi-​Rogers, M., McCrone, P. & Hughes, R. A. Barré syndrome. Neurology 64, 856–860 (2005).
immunoglobulin in multifocal motor neuropathy. Economic costs and quality of life in chronic 263. Berciano, J. et al. Severe Guillain-​Barré syndrome:
Neurology 75, 1377–1380 (2010). inflammatory neuropathies in southeast England. sorting out the pathological hallmark in an
216. Cocito, D. et al. Subcutaneous immunoglobulin in Eur. J. Neurol. 21, 34–39 (2014). electrophysiological axonal case. J. Peripher. Nerv.
CIDP and MMN: a short-​term nationwide study. 239. Forsberg, A., de Pedro-​Cuesta, J. & Widen Holmqvist, L. Syst. 14, 54–63 (2009).
J. Neurol. 261, 2159–2164 (2014). Use of healthcare, patient satisfaction and burden of 264. Grimm, A. et al. Ultrasound and electrophysiologic
217. Chaudhry, V. & Cornblath, D. R. An open-​label care in Guillain-​Barré syndrome. J. Rehabil. Med. 38, findings in patients with Guillain-​Barre syndrome
trial of rituximab (Rituxan®) in multifocal motor 230–236 (2006). at disease onset and over a period of six months.
neuropathy. J. Peripher. Nerv. Syst. 15, 196–201 240. Merkies, I. S. et al. Health-related quality-​of-life Clin. Neurophysiol. 127, 1657–1663 (2016).
(2010). improvements in CIDP with immune globulin IV 265. Adachi, Y. et al. Brachial and lumbar plexuses in
218. Niermeijer, J. M. et al. Intermittent cyclophosphamide 10%: the ICE Study. Neurology 72, 1337–1344 chronic inflammatory demyelinating
with prednisone versus placebo for polyneuropathy (2009). polyradiculoneuropathy: MRI assessment including
with IgM monoclonal gammopathy. Neurology 69, 241. Appeltshauser, L., Weishaupt, A., Sommer, C. & apparent diffusion coefficient. Neuroradiology 53,
50–59 (2007). Doppler, K. Complement deposition induced by 3–11 (2011).

22 | Article citation ID: (2018) 4:31 www.nature.com/nrdp

0123456789();
Primer

266. Sugimoto, T. et al. Ultrasonographic nerve Author contributions Grifols, Kedrion, Sanofi-​Genzyme, Shire and Pfizer; personal
enlargement of the median and ulnar nerves and the Introduction (B.C.K. and H.-P.H.); Epidemiology (B.C.K., fees for consulting for Air Liquide, Alnylam, Astellas,
cervical nerve roots in patients with demyelinating E.K.M. and C.S.); Mechanisms/pathophysiology (B.C.K., Baxalta and UCB; and research support to the institution
Charcot-​Marie-Tooth disease: distinction from patients E.K.M. and H.-P.H.); Diagnosis, screening and prevention from Kedrion. H.-P.H. reports CSL Behring fees for serving
with chronic inflammatory demyelinating (B.C.K. and C.S.); Management (B.C.K., E.K.M., C.S. and on a steering committee and from UCB for participating on
polyneuropathy. J. Neurol. 260, 2580–2587 (2013). H.-P.H.); Quality of life (B.C.K.); Outlook (B.C.K. and H.-P.H.); advisory boards.
267. Zaidman, C. M. & Pestronk, A. Nerve size in chronic Overview of Primer (B.C.K.).
inflammatory demyelinating neuropathy varies with Publisher’s note
disease activity and therapy response over time: a Competing interests Springer Nature remains neutral with regard to jurisdictional
retrospective ultrasound study. Muscle Nerve 50, B.C.K. reports receiving personal fees for consulting for claims in published maps and institutional affiliations.
733–738 (2014). Grifols and UCB. E.K.M. reports receiving funding from the
268. Corse, A. M. et al. Sensory nerve pathology in multifocal GBS/CIDP Foundation International and the National Reviewer information
motor neuropathy. Ann. Neurol. 39, 319–325 (1996). Health and Medical Research Council of Australia and is a Nature Reviews Disease Primers thanks G. Lauria,
269. Gordon, P. H. & Wilbourn, A. J. Early electrodiagnostic member of the Scientific Committee of the Trish Multiple E. Nobile-Orazio, A. Uncini, J. Vallat and the other anony-
findings in Guillain-​Barré syndrome. Arch. Neurol. 58, Sclerosis Research Foundation. C.S. reports receiving per- mous reviewer(s) for their contribution to the peer review of
913–917 (2001). sonal fees for educational talks for Alnylam, CSL Behring, this work.

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:31 23

0123456789();

Das könnte Ihnen auch gefallen