You are on page 1of 12

596

Chronic Inflammatory Demyelinating


Polyradiculoneuropathy
Jeffrey Shije, MD1 Thomas H. Brannagan III, MD1

1 Peripheral Neuropathy Center, Neurological Institute, Columbia Address for correspondence Thomas H. Brannagan, III, MD, Peripheral
University, New York, New York Neuropathy Center, Neurological Institute, Columbia University, New York,
NY 10032 (e-mail: Tb2325@cumc.columbia.edu).
Semin Neurol 2019;39:596–607.

Abstract Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is a relatively


common autoimmune disorder affecting the peripheral nerves and nerve roots, often
causing progressive or recurrent weakness with diminished reflexes. Electrodiagnostic
(EDx) studies, cerebral spinal fluid (CSF) analysis, and nerve biopsy may help provide

Downloaded by: University of California, San Francisco. Copyrighted material.


supportive evidence for the diagnosis. Most cases have a favorable response to one of
the three first-line treatments: corticosteroids, IV immunoglobulin (IVIG) and plasma-
Keywords pheresis. Responses to these treatments may vary among individual patients. There is
► chronic inflammatory evidence that a small percentage of CIDP patients with IgG class 4 (IgG4) auto-
demyelinating antibodies to paranodal proteins have characteristic clinical features and poorer
polyradiculoneuro- response to IVIG. Chemotherapy and other immunomodulatory agents, as well as
pathy hematopoietic stem cell transplantation, may be considered in refractory cases.
► CIDP The degree of disability varies, and most patients require ongoing treatment to
► inflammatory maintain stable disease, although long-term remission or cure may be achieved in
neuropathy some patients.

It had been recognized as early as the 1950s that some older patients are more likely to have a progressive course.5
patients with recurrent polyneuropathy would have symp- Ataxia and coarse tremor may be seen, presumably due to
tom improvement from corticosteroids.1 Subsequently, Dyck severe sensory/proprioceptive impairment. Of note, ataxia
et al2 demonstrated histological features of nerve inflamma- and tremor have been reported more frequently in CIDP
tion and chronic demyelination in a group of patients with patients with autoantibodies against paranodal proteins
progressive or recurrent weakness, and they had a favorable such as neurofascin-155 and contactin-1.6–11
response to corticosteroid. They were initially referred to as Cranial neuropathies, and respiratory and/or autonomic
chronic inflammatory polyradiculoneuropathy. We now dysfunction may be present, although they are infrequent.
refer to these cases as chronic inflammatory demyelinating Cranial nerve involvement, including oculomotor, facial, or
polyradiculoneuropathy (CIDP). bulbar, have been reported in 7% to 16% of cases.2–5 Respira-
tory failure is rare in CIDP, although it has been reported.12
One study reported prolonged phrenic nerve latency in most
Clinical Presentation
of the asymptomatic CIDP patients, although abnormal
The clinical features of typical CIDP are summarized pulmonary function tests were more associated with
in ►Table 1. The progressive or recurrent weakness in typical reduced phrenic nerve compound motor action potential
CIDP is relatively symmetrical, often involving distal and (CMAP) amplitude, which was rare.13 Autonomic symptoms
proximal extremities. Absent or diminished reflexes are seen are infrequent and generally mild when present. These may
in nearly all cases. There are accompanying sensory symp- include bowel and bladder complaints, while dry mouth/
toms, such as numbness and paresthesias, in the majority of eyes, orthostatic intolerance, sexual dysfunction, flushing,
the cases, although pain is less frequent.2–5 Younger patients and hyperhidrosis are even rarer. Subclinical abnormalities
are more likely to have relapsing–remitting disease, whereas on autonomic testing may be seen, but there is no correlation

Issue Theme Peripheral Neuropathies; Copyright © 2019 by Thieme Medical DOI https://doi.org/
Guest Editors, Michelle Kaku, MD, and Publishers, Inc., 333 Seventh Avenue, 10.1055/s-0039-1693008.
Peter Siao, MD New York, NY 10001, USA. ISSN 0271-8235.
Tel: +1(212) 584-4662.
CIDP Shije and Brannagan 597

Table 1 Clinical features of typical CIDP

Symptoms Frequency of symptoms Other characteristics


Weakness All Relatively symmetrical, involving proximal and
distal extremities. May be progressive or
relapsing–remitting.
Areflexia/hyporeflexia Nearly all Markedly diminished if not absent
Sensory symptoms Majority Numbness and/or paresthesia. Pain is relatively less
frequent.
Tremor and ataxia, presumably May be associated with paranodal antibody (i.e.,
related to severe sensory/ neurofascin-155, contactin-1, etc.)
proprioceptive impairment
Cranial nerve involvement Infrequent, 7%–16% Oculomotor, facial or bulbar involvement all have
been reported
Respiratory symptoms Infrequent (respiratory failure is Subclinical prolonged phrenic nerve latency may
rare) be present, but abnormal phrenic nerve CMAP
amplitude is more associated with abnormal PFT,
which is rarer.

Downloaded by: University of California, San Francisco. Copyrighted material.


Autonomic symptoms Infrequent Typically mild even if present, i.e., bowel/bladder
symptoms. Sicca syndrome, orthostatic
symptoms, and sexual dysfunction are even rarer.

between autonomic dysfunction and the severity of the and remyelination, as well as perivascular and endoneurial
overall disease.14–18 infiltration by macrophages and T cells.2,29,30 Elevated levels of
Some CIDP patients have an acute-onset presentation that proinflammatory cytokines associated with cell-mediated
is clinically indistinguishable from acute inflammatory immune response have been reported in the serum, CSF, and
demyelinating polyradiculoneuropathy (AIDP) or Guillain– nerve biopsies of CIDP patients during active and subacute
Barre Syndrome (GBS) early in the disease course. One disease.31–35 In an animal model, a spontaneous autoimmune
prospective cohort study showed that GBS patients had no peripheral polyneuropathy (SAPP), similar to CIDP in patho-
more than two treatment-related fluctuations and no pro- physiology, can be produced in mice from unopposed cell-
gression of their neurological impairment beyond mediated immunity.36 The exact antigenic trigger is not
two months.19 Therefore, if a patient was previously thought identified in most cases of CIDP. Immunization with peripheral
to have GBS but continues to worsen two months after myelin proteins (i.e., P0, P2, and PMP22) may produce a chronic
symptom onset, it would raise the suspicion of CIDP instead. experimental allergic neuritis (EAN) in mice and rabbits,37,38
and anti-P0 antibodies from CIDP patients may cause demye-
linating neuropathy when injected into experimental ani-
Epidemiology
mals.39 However, antimyelin protein antibodies have only
The epidemiologic data varies depending on geography. CIDP been reported in a minority of CIDP patients, and these
prevalence ranging from 0.8/100,000 (Tottori Prefecture, autoantibodies have not elicited consistent inflammatory
Japan) to 8.9/100,000 (Olmsted County, Minnesota) have responses in animals.40–43 More recently, autoantibodies
been reported.20–24 The annual incidence may range from against paranodal proteins, that is, neurofascin-155 and con-
0.15 to 1.6/100,000.20,25 The different criteria used to define tactin-1, have been reported in CIDP cases with characteristic
the disease may contribute to the variation in its reported clinical features and responses to treatment, but these only
prevalence and incidence.26 CIDP can affect age groups from account for 2% to 6% of cases.44
childhood to beyond the eighth decade of life. The highest
prevalence reported is in the seventh decade of life, although
Diagnosis
the mean age of onset may be around the fourth to fifth
decade of life; men are more likely to be affected than CIDP may be suspected in someone with progressive or
women.20,25,27 It had been reported that the disease onset recurrent weakness in the proximal and distal extremities
may be preceded by infection or immunization in up to one for at least two months. Albuminocytologic dissociation
third of CIDP cases, which is notably less compared with GBS, (elevated CSF protein without CSF pleocytosis) is often
where nearly half may report an antecedent infection.3,28 present. Aside from nerve biopsy, evidence of nerve demye-
lination may be demonstrated by electrodiagnostic (EDx)
testing. The Edx features of demyelination include significant
Pathophysiology
reduction of motor conduction velocities, presence of con-
There is evidence that CIDP is an autoimmune demyelinating duction block and temporal dispersion, significant prolonga-
disorder of the peripheral nerves and nerve roots. Nerve tion of F-wave latencies, distal motor latencies and distal
biopsies have demonstrated chronic segmental demyelination compound motor action potential (CMAP) duration.

Seminars in Neurology Vol. 39 No. 5/2019


598 CIDP Shije and Brannagan

Various clinical and EDx criteria for CIDP had been proposed criteria, since they do not require nerve biopsy or lumbar
by different investigators early on,2,4,45 including a research puncture if adequate clinical and EDx criteria are met. Thai-
criteria developed by an ad hoc subcommittee of the American setthawatkul et al52 incorporated prolonged distal CMAP
Academy of Neurology (AAN) AIDS Task Force (►Table 2).46 duration into the EDx criteria to further promote diagnostic
The AAN research criteria required three of its four EDx sensitivity. Magda et al53 proposed a set of diagnostic criteria
parameters of demyelination (abnormal conduction velocity, based on minimal EDx changes shared by a collection of CIDP
distal motor latency, F-wave latency, and conduction block/ patients. These were adopted in the Neuropathy Association
temporal dispersion) to be present, with each of those (except guidelines for the diagnosis and treatment of CIDP,54 and it was
conduction block/temporal dispersion) in at least two nerves, inclusive of patients with abnormal EDx findings in three
plus nerve biopsy and CSF analysis, for a definite diagnosis of nerves, with findings diagnostic of demyelination in just one
CIDP. It had been pointed out that the AAN research criteria nerve. The more recent European Federation of Neurologic
may be too stringent and may not be met by many patients Societies/Peripheral Nerve Society (EFNS/PNS) criteria55
with CIDP.47,48 Also, it is not uncommon for the neuropathy in (►Table 4) expanded on the previous criteria, and also allows
CIDP to have substantial secondary axonal loss so that EDx the diagnosis of CIDP with EDx demyelinating findings in one
evaluation of demyelination is no longer feasible.49 There have nerve if additional supportive criteria are met. The EFNS/PNS
been numerous proposed modifications to the diagnostic criteria have been used in recent and ongoing observational
criteria in the interest of improving its sensitivity and speci- studies and clinical trials.
ficity. For example, Saperstein et al48 required only two of the

Downloaded by: University of California, San Francisco. Copyrighted material.


Not all proposed diagnostic criteria are mentioned here.
four EDx parameters of demyelination and did not mandate Attempts to improve the sensitivity of CIDP diagnostic
nerve biopsy to promote diagnostic sensitivity, while having criteria often decrease their specificity, and vice versa. A
more stringent EDx requirements for conduction block to retrospective study comparing CIDP, diabetic neuropathy,
promote specificity. Nicolas et al50 placed more emphasis on and amyotrophic lateral sclerosis patients showed that the
conduction block and temporal dispersion to promote diag- Magda et al/Neuropathy Association guidelines and the
nostic distinction from other demyelinating neuropathies. EFNS/PNS criteria had relatively higher sensitivity (75%
This emphasis on conduction block/temporal dispersion was and 73%, respectively), while maintaining relatively high
also reflected in the criteria by the Inflammatory Neuropathy specificity (83% and 88%, respectively), among the many
Cause and Treatment (INCAT) group (►Table 3),51 which has proposed criteria.56 Several of the aforementioned criteria
been used in large clinical trials. Patients may be diagnosed or (AAN, Saperstein et al, EFNS/PNS) have parameters for
enrolled in clinical trials more promptly based on the INCAT

Table 3 Features of the INCAT diagnostic criteria for CIDP51

Table 2 Features of the AAN research criteria for CIDP46 Clinical features:
Progressive or relapsing motor and sensory dysfunction of
Clinical features: more than one limb for >2 months.
Progressive or relapsing motor and sensory dysfunction of Reduced or absent reflexes.
more than one limb for >2 months.
EDx parameters for demyelination:
Reduced or absent reflexes
a. Reduced conduction velocity <80% of LLN if
3 of the 4 EDx parameters below are required: amplitude > 80% of LLN or <70% of LLN if amplitude
a. Reduced conduction velocity <80% of LLN if <80% of LLN in 2 nerves
amplitude > 80% of LLN or <70% of LLN if amplitude b. Prolonged distal motor latency >125% of ULN if amplitude
<80% of LLN in 2 nerves >80% of LLN or >150% of ULN if amplitude <80% of LLN in
b. Prolonged motor distal latency >125% of ULN if amplitude 2 nerves
>80% of LLN or >150% of ULN if amplitude <80% of LLN in c. Prolonged F-wave latency >120% of ULN if amplitude
2 nerves >80% of LLN or >150% of ULN if amplitude <80% of LLN in
c. Prolonged F-wave latency >120% of ULN if amplitude 2 nerves
>80% of LLN or >150% of ULN if amplitude <80% of LLN in d. Partial conduction block (>20% amplitude reduction from
2 nerves distal to proximal stimulation sites if <15% change in
d. Partial conduction block (>20% amplitude reduction from duration between the CMAPs) or abnormal temporal
distal to proximal stimulation sites if <15% change in dispersion (>15% change in duration between distal and
duration between the CMAPs) or abnormal temporal proximal stimulation sites and >20% amplitude reduction
dispersion (>15% change in duration between distal and from distal to proximal stimulation sites) in 1 nerve
proximal stimulation sites and >20% amplitude reduction
Diagnosis of CIDP requires one of the following:
from distal to proximal stimulation sites) in 1 nerve
• 2 nerves with conduction block or abnormal temporal
Definite CIDP: Clinical and EDx parameters, plus dispersion þ 1 other nerve with either abnormal distal
demyelination on nerve biopsy and CSF (for absence of motor latency, conduction velocity or F-wave response. Or
pleocytosis) • 3 nerves with either abnormal distal motor latency,
Probable CIDP: clinical and EDx parameters plus CSF analysis conduction velocity or F-wave response. Or
support • If only 2 nerves with significant EDx abnormality, need
Possible CIDP: clinical and EDx parameters unequivocal evidence of demyelination on nerve biopsy.

Abbreviations: CMAP, compound motor action potential; EDx, electro- Abbreviations: CMAP, compound motor action potential; EDx, electro-
diagnostic; LLN, lower limit of normal; ULN, upper limit of normal. diagnostic; LLN, lower limit of normal; ULN, upper limit of normal.

Seminars in Neurology Vol. 39 No. 5/2019


CIDP Shije and Brannagan 599

Table 4 Features of the EFNS/PNS diagnostic criteria for CIDP55 they fall short of satisfying all diagnostic parameters for
definite CIDP, considering no one criteria has both perfect
Clinical criteria: sensitivity and specificity.
Typical: progressive, stepwise, or recurrent symmetric Ultimately, the approach to diagnosis is having suspicion
proximal and distal weakness and sensory dysfunction of all
of CIDP when the patient has symmetrical, progressive, or
extremities 2 months and absent or reduced tendon
reflexes, or relapsing weakness for more than two months, involving
Atypical: inclusive of DADS, Lewis-Sumner syndrome, distal and proximal extremities, with diminished or absent
asymmetrical, pure motor, and pure sensory symptoms reflexes. If there is adequate EDx support for an acquired
EDx criteria: demyelinating neuropathy/radiculoneuropathy, with no
Definite: at least one of the following parameters: other apparent etiology, the diagnosis of CIDP is justified.
(a) Conduction velocity 70% of LLN in 2 nerves If EDx findings are equivocal or if nerve conduction
(b) Distal motor latency 150% of ULN (excluding median
responses are inadequate to assess for demyelination, other
neuropathy at the wrist from carpal tunnel syndrome) in
2 nerves tests such as CSF analysis, nerve biopsy, and contrast MRI of
(c) F-wave latency 130% of ULN (150% if CMAP amplitude the nerve roots or a diagnostic trial of immunotherapy may
<80% of LLN) in 2 nerves offer additional diagnostic value.
(d) Absent F-wave if distal CMAP amplitude 20% of LLN in 2
nerves, if there is another demyelinating finding in
Atypical Variants
another nerve

Downloaded by: University of California, San Francisco. Copyrighted material.


(e) Partial conduction block: 50% amplitude reduction The EFNS/PNS criteria has Lewis-Sumner Syndrome, distal
from distal to proximal stimulation sites if distal CMAP acquired demyelinating symmetric (DADS) neuropathy, and
amplitude 20% of LLN in 2 nerves (if seen in only one sensory CIDP included as atypical CIDP.55 Lewis-Sumner
nerve, then requires another demyelinating finding in Syndrome, also known as multifocal acquired demyelinating
another nerve)
sensory and motor neuropathy (MADSAM), was initially
(f) Temporal dispersion: >30% duration increase from distal
to proximal stimulation sites in 2 nerves described by Lewis and colleagues, who reported chronic
(g) Distal CMAP duration increased in at least 1 nerve asymmetric sensorimotor demyelinating neuropathy that
(median 6.6 milliseconds, ulnar 6.7 milliseconds, resembled mononeuritis multiplex but were diagnosed
peroneal 7.6 milliseconds, tibial 8.8 milliseconds) with a demyelinating neuropathy by persistent motor con-
plus one other demyelinating parameter in another
duction block in the nerves affected.57 Patients with Lewis-
nerve
Probable: Sumner Syndrome generally respond to conventional CIDP
Partial conduction block 30% amplitude reduction treatment, and some of them evolve to a more typical CIDP.58
between distal and proximal stimulations sites (excluding DADS neuropathy has been used to describe those with only
the posterior tibial nerve) if distal negative peak CMAP distal extremity involvement. In this population, nearly two-
amplitude 20% of LLN in 2 nerves. (If seen in only one
thirds may have an IgM kappa monoclonal gammopathy, and
nerve, then requires another demyelinating finding in
another nerve.) of those, more than half may have anti–myelin-associated
Possible: glycoprotein (MAG) antibody.59 DADS neuropathy has been
Only 1 nerve meeting one of the definitive EDx parameters reported to progress to classic CIDP,60 although it should be
above emphasized that only those without IgM gammopathy or
Definite CIDP: Clinical criteria (typical or atypical) plus: anti-MAG antibodies are more likely to respond to conven-
• definite EDx criteria, or tional CIDP treatment59,61 and considered an atypical variant
• probable EDx criteria and one supportive criterion, or of CIDP.62,63 Pure sensory CIDP has been described, charac-
• possible EDx criteria and two supportive criteria.
Probable CIDP: clinical criteria (typical or atypical) plus: terized by progressive sensory neuropathy without weak-
• probable EDx criteria, or ness, although many of these cases also had EDx evidence of
• possible EDx criteria and one supportive criterion demyelination involving motor nerves. CSF albuminocytolo-
Possible CIDP: clinical criteria (typical or atypical) plus gic dissociation64 and demyelination on nerve biopsy65 may
possible EDx criteria be seen in these patients as well. It has been reported that
Supportive criteria: 1) CSF protein elevated with <10 WBC. conventional CIDP treatment may stop symptom progression
2) MRI contrast enhancement or hypertrophy of the nerve and improve sensory impairment or sensory ataxia in these
roots or plexus. 3)Sensory conduction velocity slowing, or
patients.65,66 Some of the pure sensory CIDP patients may
reduced median (excluding at the wrist) or radial SNAP with
normal sural amplitude, or delayed SSEP. 4) Objective clinical evolve to have motor symptoms eventually, and have a
improvement after immunomodulatory treatment. 5) Nerve pattern of classic CIDP with proximal and distal weakness,
biopsy showing unequivocal evidence of demyelination/re- although it may be several years from the onset of pure
myelination. sensory symptoms before subsequent motor involvement
Abbreviations: CMAP, compound motor action potential; EDx, electro- becomes apparent.66,67 Furthermore, a particular form of
diagnostic; LLN, lower limit of normal; ULN, upper limit of normal. sensory CIDP, named chronic immune sensory polyradiculo-
pathy (CISP), may only involve the preganglionic sensory
definite, probable, and possible CIDP. While most clinical nerve roots, with therefore normal sensory nerve action
trials may require a definitive diagnosis of CIDP, having potentials, and may be demonstrated by abnormal somato-
parameters for probable and possible CIDP gives clinicians sensory evoked potential testing or MRI contrast enhance-
justification to treat the patient if CIDP is suspected, even if ment of posterior nerve roots. These patients may have CSF

Seminars in Neurology Vol. 39 No. 5/2019


600 CIDP Shije and Brannagan

albuminocytologic dissociation as well, and may respond to often has demyelinating features similar to CIDP,85,86 but
conventional CIDP treatment.68 typically does not respond to conventional CIDP treatment.
Charcot-Marie-Tooth type 1 (CMT1), being a hereditary
Coexisting Disorders and Mimics demyelinating neuropathy, can have similar clinical and
CIDP may coexist with other conditions, and some of these electrodiagnostic features to CIDP. It has been entertained
may cause unrelated neuropathies as well, which may pre- whether CMT1A, being associated with a PMP22 mutation,
sent additional diagnostic challenges. Nevertheless, CIDP increases the risk of developing CIDP, considering anti-
patients with these coexisting conditions respond to con- PMP22 antibodies have been isolated in the serum of CIDP
ventional CIDP treatment. On the other hand, some disorders patients.41 It should be noted that most patients with CMT1
may cause neuropathies that mimic or even satisfy the do not respond to immunotherapy; however, coexisting
diagnostic criteria for CIDP but otherwise do not respond inflammatory neuropathy has been reported in CMT type
to conventional CIDP treatment. It is important to distin- 1A, 1B, and CMT X.87–89 The typical clinical course of CMT1 is
guish the conditions that may simply coexist with CIDP from that of a gradually progressive length-dependent motor and
the mimics. sensory polyneuropathy. However, if the patient has super-
CIDP and diabetic (type 1 and type 2) neuropathy may imposed acute/subacute, relapsing, or stepwise deteriora-
have overlapping clinical and EDx features. The question of tion, in a non–length-dependent manner, a suspicion of
whether diabetes increases the risk of CIDP has been superimposed CIDP may be raised. CIDP and CMT1 may
debated.20,69,70 Nevertheless, diabetic patients with CIDP

Downloaded by: University of California, San Francisco. Copyrighted material.


have similar EDx features of demyelination; however, multi-
respond to conventional CIDP treatment,71,72 although care- focal slowing and conduction block are more likely to be seen
ful consideration of treatment options should be given to in CIDP rather than CMT1.90 These and additional associated
avoid worsening the underlying diabetes (i.e., caution with conditions, and mimics, are listed in ►Table 5.
corticosteroids).
Various forms of neuropathy can occur in different stages
Treatment and Outcome
of human immunodeficiency virus (HIV) infection, and CIDP
has been reported in HIV-positive patients as well.73,74 It is Corticosteroids
important to note that HIV patients with CIDP may have CSF One of the hallmarks of CIDP is that it responds to corticos-
pleocytosis (often with CSF WBC >10/mm3).75 In addition to teroid treatment.1,2 An early randomized controlled trial
treating the underlying HIV infection, the coexisting CIDP demonstrated that patients who received prednisone at an
often responds to conventional CIDP treatment as well.73,76 equivalent of 60 mg a day, tapered over three months,
Monoclonal gammopathy of undetermined significance resulted in a better neuropathy disability score (NDS), later
(MGUS), including immunoglobulin-A (IgA), immunoglobu- renamed neuropathy impairment score (NIS), compared
lin-G (IgG) and immunoglobulin-M (IgM) gammopathy, may with placebo.100 Adverse effects of long-term high dose
coexist or even cause neuropathy. When CIDP patients have steroid use include dyspepsia, hyperglycemia, osteoporosis,
coexisting MGUS, they may respond to conventional treat- weight gain, Cushingoid features, susceptibility for infection,
ment similar to CIDP patients without MGUS.77 However, and mood and sleep disturbance. Subsequently, various
there is a number of IgM class autoantibodies, including anti- investigators reported that pulsed steroid regimens are
MAG, GD1b, and sulfatide antibodies, which can be asso- also effective, while causing relatively less side effects com-
ciated with neuropathies with demyelinating features. Anti- pared with daily oral steroids.101,102 The efficacy of pulsed
MAG antibody is associated with a distal sensorimotor oral steroids was demonstrated in the PREDICT study,103
neuropathy with prominent distal demyelination, and is where pulsed oral dexamethasone 40 mg a day for 4 days,
treated as a separate disorder, often responding better to every 4 weeks, showed similar efficacy but less adverse
rituximab and not intravenous immunoglobulin (IVIG).78–81 effects (i.e., Cushingoid features and sleeplessness) com-
Anti-GD1b antibodies can be associated with chronic-ataxic- pared with daily prednisolone 60 mg tapered over 6 months
neuropathy-ophthalmoplegia-m-protein-agglutination-dis-
ialosyl-antibodies (CANOMAD) syndrome, and is often Table 5 Associated conditions and mimics
responsive to immunotherapy.82,83 Antisulfatide antibodies
may be associated with a predominantly sensory or distal Conditions in which coexisting CIDP may be seen.
sensorimotor neuropathy.84 When a monoclonal protein is Diabetes, HIV, monoclonal gammopathies, Charcot-Marie-
present, it is especially important to rule out a plasma cell Tooth type 1, other autoimmune disorders (i.e., lupus,
dyscrasia (i.e., multiple myeloma), and a skeletal survey is inflammatory bowel disease, membranous
nephropathy),91–93 chronic hepatitis,4 post-organ
recommended to evaluate for osteosclerotic myeloma. Par-
transplant,94 malignancy,95 TNF-α inhibitor use (not known
ticularly, lambda-restricted monoclonal gammopathy may to cause neuropathy otherwise).96 Immune checkpoint
be associated with polyneuropathy, organomegaly, endocri- inhibitor use97,98
nopathy, M-protein, and skin changes (POEMS) syndrome Other neuropathies that may mimic CIDP clinically and
(i.e., hyperpigmentation and hemangioma). An elevated vas- electrodiagnostically
cular endothelial growth factor (VEGF) level can be a diag- Anti-MAG neuropathy, multifocal motor neuropathy,
nostic marker for POEMS syndrome and will also decrease neuropathy in POEMS, hereditary transthyretin amyloid
neuropathy.86,99
with successful treatment. Neuropathy in POEMS syndrome

Seminars in Neurology Vol. 39 No. 5/2019


CIDP Shije and Brannagan 601

for CIDP patients. Subsequently, another randomized con- reasonable alternative. It was shown that SCIG of 0.4 g/kg
trolled trial comparing IV methylprednisolone 0.5 g a day for (over 2–3 infusions) weekly for 5 weeks had similar efficacy
four consecutive days/month and IVIG 0.5 g/kg a day for four as IVIG 2 g/kg over 5 days as initial treatment in improving
consecutive days/month over a 6-month period, demon- muscle strength at 10 weeks, although peak improvement in
strated that in the patients who responded to treatment, patients receiving IVIG occurred sooner with IVIG than in
the degree of improvement was similar between the two those receiving SCIG. Therefore, severely disabled patients
groups.104 The IV methylprednisolone group had more may benefit more from IVIG initially, and SCIG may be
patients drop out of the study, mostly citing lack of improve- considered as maintenance treatment.113 The efficacy of
ment as the reason, compared with the IVIG group. However, SCIG as a maintenance treatment in patients who previously
it should be noted that the previous PREDICT study had responded to IVIG has been demonstrated as well.114,115
shown the median time to improvement from pulsed oral SCIG at a maintenance dose of 0.2 to 0.4 g/kg weekly may
steroid was 17 weeks (ranging from 13–20 weeks). There- be used.116 SCIG had less moderate and/or systemic adverse
fore, the likelihood of improvement from IV methylpredni- effects compared with IVIG based on a meta-analysis,
solone could have been underestimated due to premature although a mild transient injection site reaction may be
treatment discontinuation. Long-term follow-up analysis of more common with SCIG.117
the same study showed that ultimately both groups had a
similar percentage of relapse after treatment discontinua- Plasmapheresis

Downloaded by: University of California, San Francisco. Copyrighted material.


tion, although the IV methylprednisolone group had a sig- Plasmapheresis is effective as well, its efficacy as initial
nificantly longer median time to relapse (median: treatment is comparable to IVIG in a 6-week study.118
14 months, range: 1–31 months) compared with the IVIG Usually, 1 to 1.5 times body volume is exchanged at each
group (median: 4.5 months, range: 1–24 months). session, and 5 to 10 exchanges (2 to 3 exchanges a week as
tolerated) may be considered for an initial course of treat-
Immunoglobulin ment. Plasmapheresis may be used as a first line initial
Randomized controlled studies have demonstrated that CIDP therapy, especially in those with severe symptoms, consider-
patients who received an initial treatment of IVIG could have ing that an improvement is often seen within the first week
improvement of their strength and disability scores within 2 or after a couple of exchanges. However, more than half of
to 4 weeks of the infusion.51,105 It was also shown that patients have relapsing symptoms within 1 to 2 weeks after
relapsing symptoms may occur from 3 to 22 weeks after stopping plasmapheresis.119 Its adverse effects may include
an initial treatment of IVIG at 2 g/kg, and additional IVIG can hemodynamic disturbances, depletion of coagulation fac-
be used to maintain the benefit of the initial treatment.106 tors, and venous access-related complications. Therefore, it
Subsequently, it was demonstrated in the ICE study,107 may be less ideal as a maintenance therapy option compared
starting IVIG with a 2 g/kg loading dose, followed by a with corticosteroids and IVIG.
maintenance regimen of 1 g/kg every 3 weeks, can maintain
the improvement in their disability score at 24 weeks, and up Variability in Treatment Response and Other
to 48 weeks in the extended phase of the study. Nevertheless, Therapy Options
patients may still report relapses sooner than 3 weeks after Steroids, IVIG, and plasma exchange remain the most evi-
infusion. Maintenance IVIG as frequently as every two weeks dence-based, first-line treatment options for CIDP,120–122
has been reported to be effective and well tolerated in some and their dosing strategies are summarized in ►Table 6.
cases.108 One retrospective study showed that of the CIDP patients
Immediate adverse reactions following IVIG administra- (about one-third) who did not respond to an initial first-line
tion are often mild, including transient flu-like symptoms, treatment, about half may respond to treatment after
headache, flushing of the face, change in blood pressure, and switching from steroids to IVIG or plasmapheresis, or after
tachycardia. Minor reactions can often be ameliorated by switching from IVIG or plasmapheresis to corticosteroids.
pretreatment with analgesics, antihistamines, and corticos- Still, there remained 19% of patients who did not response to
teroids, and by slowing the infusion rate. More serious either steroids, IVIG, or plasmapheresis.123 Considering
adverse effects may include thrombosis, renal injury, aseptic about one-fifth of CIDP patients may not respond to steroids,
meningitis, neutropenia, and pseudohyponatremia. Anaphy- IVIG, or plasmapheresis, there is a need for other treatment
laxis is rare but its risk is increased in those with IgA options. Various chemotherapy and other immunomodula-
deficiency. Avoiding sucrose-containing preparations may tory agents, that is, azathioprine,124 mycophenolate mofe-
decrease the risk of renal injury. Decreasing the infusion rate til,108,125 methotrexate,126 cyclophosphamide127(with or
(i.e., at least over 8 hours or no greater than 3–5 g/hr) may without fludarabine108), cyclosporine,128,129 interferon
decrease the risk of thrombosis, renal failure, and aseptic (IFN) α,130,131 IFN β-1a,132 and rituximab,133,134 have been
meningitis.109,110 There is no evidence of any particular reported to offer some benefit in refractory cases. However,
brand or preparation of IVIG being more efficacious or having these treatments lack support from large clinical trials.
more adverse effects.111,112 However, there are anecdotes of Randomized controlled studies with azathioprine,135 IFN
patients tolerating one brand of IVIG better than another. β-1a,136,137 methotrexate,138 and Fingolimod139 failed to
For patients who respond to IVIG but prefer more auton- show significant efficacy for CIDP. It should be noted that
omy, subcutaneous immunoglobulin-G (SCIG) may be a the azathioprine study only evaluated its efficacy as an

Seminars in Neurology Vol. 39 No. 5/2019


602 CIDP Shije and Brannagan

Table 6 First-line treatment options

Rx Dosing options Precautions and considerations with Rx


Corticosteroids Prednisone 60 mg daily for one month, then taper Recommend GI prophylaxis, glycemic and BP
gradually over 3–6 months. monitoring, Ca & Vit-D supplement.
Dexamethasone 40 mg/day  4 days every Improvement may not be seen for 3 to 4 months
four weeks for 6 months, then taper gradually if
able
IV methylprednisolone 0.5 g/day  4 days
very month for 6 months, then taper gradually if
able
Immunoglobulin IVIG 2 g/kg over 4–5 days initially, then 1 g/kg over IgA deficiency increases risk of anaphylaxis.
1–2 days every three weeks as maintenance (may Caution for renal insufficiency. Vigilance for DVT,
increase frequency to every 2 weeks in some cases) neutropenia, hemodynamic instability. Avoid high
SCIG, initial 0.4 g/kg/wk (over 2–3 infusion). rate of infusion. Can pretreat with Benadryl,
Maintenance 0.2 to 0.4 g/kg weekly. Tylenol, and steroid for mild immediate reactions
Plasmapheresis 1 to 1.5 times total body volume per exchange, for Monitor for hemodynamic disturbance and
total of 5 to 10 exchanges (2–3 exchanges/week as complications associated with venous access, i.e.,
tolerated) as initial treatment. thrombosis, thrombophlebitis, line infection/

Downloaded by: University of California, San Francisco. Copyrighted material.


Schedule for maintenance exchanges may vary and sepsis, etc.
depend on relapsing symptoms

Table 7 Other immunomodulatory/chemotherapy agents


adjunctive therapy to prednisone, and the duration of the
reported to be beneficial for CIDP refractory to first-line
study might have been suboptimal.135 A Cochrane review of
treatments, in case reports and retrospective observations
some of these studies concluded that azathioprine, metho-
trexate, and IFN β-1a offer no significant benefit, although a
Azathioprine 100–200 mg/day
small or moderate benefit could not be ruled out.140
One retrospective multicenter observational study in Italy Cyclophosphamide 1 g/m2 IV/month, or 2 mg/kg oral/
day. High dose: 200 mg/kg IV over
showed that of the 110 nonresponders to conventional 4 days. (or 250 mg/m2/day with
treatment (i.e., steroids, IVIG, and plasmapheresis), 37% fludarabine 25 mg/m2/day for 3 days
had a favorable response from other immunosuppressive/ every 5–6 days)
immunomodulatory treatments including azathioprine, Mycophenolate 500–3000 mg/day (typically
cyclophosphamide, methotrexate, cyclosporine, rituximab, mofetil 2000 mg/day), divided twice a day
and IFN-α, with an average improvement of 1.9 points on the Rituximab 375 mg/m2/week  4 weeks
Rankin Scale.124 There was no significant difference in the
Methotrexate 7.5–15 mg/week
treatment responses between these agents. It was also noted
that azathioprine was utilized the most, presumably due to Cyclosporin A 100–300 mg/day or 5 mg/kg/day
(keeping plasma concentration
its ease of use; cyclophosphamide had the highest percent of
between 100 and 150 ng/mL)
treatment responders, methotrexate had the lowest percent
of side effects, and cyclosporine had the highest percent of
discontinuation due to side effects. It should be noted that report142 however, it also has been described to be asso-
some of these agents, such as azathioprine and mycopheno- ciated with demyelinating neuropathy.143 Etanercept, a
late, may not show benefit for many months after the start of tumor necrosis factor (TNF)-α inhibitor, has been reported
treatment. There is no standardized dosing for these treat- to be effective in some CIDP patients refractory to or intol-
ments, and dosing strategies based on the aforementioned erant of first-line treatments144; however, TNF-α inhibitor is
case reports and retrospective study are illustrated in also associated with developing CIDP.96
►Table 7. It is advised that one should be familiar with the Finally, stem cell transplantation may be considered in
potential adverse effects of these immunomodulatory/che- select cases after carefully weighing its potential risks and
motherapy agents prior to use, and collaboration with rheu- benefit. There is a report of allogenic hematopoietic stem cell
matology or oncology may be considered. transplant in a CIDP patient who achieved full recovery
Some additional immunomodulatory treatments have within 8 months and had no relapse at 6.5 year follow
been reported to be effective in case reports and small up.154 There are also reports of autologous stem cell trans-
cohorts, but may not be ideal options for various reasons. plantation leading to disease remission145–147 however,
Alemtuzumab has been reported to decrease the IVIG relapsing disease was also reported in two of those cases
requirement in IVIG-dependent CIDP patients in a small at 2 and 5 years post transplant. In a retrospective review of
cohort study.141 However, its risk of causing other serious 11 patients who received autologous hematopoietic stem
autoimmune disorders may not justify its use when the cell transplant for refractory CIDP, 3 patients had relapsing
patient is otherwise well controlled on maintenance IVIG. disease (1 at 14 months and 2 at 23 months), although 8 of
Tacrolimus has been reported to be beneficial in a case those 11 patients were able to maintain drug-free remission

Seminars in Neurology Vol. 39 No. 5/2019


CIDP Shije and Brannagan 603

at a median follow up time of 28 months.148 One of the Table 8 CIDP Disease Activity Status (CDAS)153
refractory patients achieved remission again after a second
autologous stem cell transplantation. The authors reported 1. Cure: off treatment 5 years
short-term complications that included CMV and EBV reac- A. Normal exam
B. Abnormal exam but at least stable
tivation, and pancreatitis. A phase 2 open-label clinical trial
for nonmyeloablative autologous hematopoietic stem cell 2. Remission: off treatment <5 years
A. Normal exam
transplantation in CIDP patients is ongoing.
B. Abnormal exam but at least stable

Paranodal Antibodies and Response to Treatment 3. Stable active disease: on treatment at least 1 year
A. Normal exam
It is important to be aware that a small percentage of CIDP B. Abnormal exam but at least stable
patients have IgG class 4 (IgG4) autoantibodies to paranodal
4. Improvement: on treatment between 3 months and 1 year
proteins, that is, neurofascin-155 and contactin-1.44 Anti–
A. Normal exam
neurofascin-155 antibody is associated with higher inci- B. Abnormal exam but at least stable
dence of ataxia and tremor, younger age (20–30 s) of
5. Unstable active disease: abnormal exam with progressive
onset,6,8,149 and higher CSF protein levels,9 and a small or relapsing course
percentage of them may have concomitant demyelinating A. Treatment naïve or treatment < 3months
disease involving the central nervous system.7 Anti–contac- B. Off treatment
C. On treatment

Downloaded by: University of California, San Francisco. Copyrighted material.


tin-1 is also associated with a higher incidence of ataxia and
tremor,8,11 and it may have a more rapidly progressive
onset.10,150 CIDP patients with anti–neurofascin-155 and
contactin-1 antibodies have poorer response to IVIG in mediated (antibody-driven) autoimmune process may play a
general, although they may have more a favorable response role in some cases. Perhaps, different CIDP patients may have
to corticosteroids.6–9,11,149 In addition, rituximab has been different pathophysiologies that influence their response to
reported to be effective in CIDP patients with neurofascin- various treatments. This is evidence from the recent discov-
155 and contactin-1 auto-antibodies, including those who ery that CIDP patients who have IgG4 class antibodies to
have failed both IVIG and corticosteroids.10,149,151,152 paranodal proteins, such as neurofascin-155 and contactin-
1, have certain distinct clinical features and respond poorly
Outcome to IVIG but may have a better response to corticosteroids or B
Though over 50% of patients with CIDP are disabled at some cell depleting agents such as rituximab. As our knowledge of
time during the disease,21 the majority of CIDP patients can CIDP continues to evolve, it may be possible to tailor its
remain stable with ongoing treatment. A CIDP Disease treatment based on the immunopathological characteristics
Activity Status (CDAS) classification (►Table 8) was devised of individual patients and improve their overall outcome.
by a panel of experts, based on outcome data from long-
term follow up of 106 CIDP patients.153 Remission, defined Conflict of Interest
as being stable off treatment for less than 5 years, was seen Dr. Brannagan has received compensation for consulting
in 20% of patients. Furthermore, 11% were considered from CSL-Behring, Grifols and Shire.
cured, if clinically stable for at least 5 years off treatment.
Therefore, patients with stable disease on treatment for at
References
least 1 year can be considered for tapering of their ther- 1 Austin JH. Recurrent polyneuropathies and their corticosteroid
apeutic regimen. There is no standardized approach to treatment; with five-year observations of a placebo-controlled
tapering treatments. Gradually decreasing the therapy dos- case treated with corticotrophin, cortisone, and prednisone.
ing and frequency, with close monitoring for relapsing Brain 1958;81(02):157–192
2 Dyck PJ, Lais AC, Ohta M, Bastron JA, Okazaki H, Groover RV.
symptoms, is advised.
Chronic inflammatory polyradiculoneuropathy. Mayo Clin Proc
1975;50(11):621–637
Conclusion 3 McCombe PA, Pollard JD, McLeod JG. Chronic inflammatory
demyelinating polyradiculoneuropathy. A clinical and electro-
Our collective understanding of CIDP has evolved over time. physiological study of 92 cases. Brain 1987;110(Pt 6):1617–1630
Typical cases and atypical variants, as well as other mimics, 4 Barohn RJ, Kissel JT, Warmolts JR, Mendell JR. Chronic inflam-
matory demyelinating polyradiculoneuropathy. Clinical charac-
can be recognized with a thoughtful approach to diagnosis.
teristics, course, and recommendations for diagnostic criteria.
Corticosteroids, IVIG, and plasmapheresis have the best Arch Neurol 1989;46(08):878–884
evidence as first-line treatments, while steroids and IVIG 5 Hattori N, Misu K, Koike H, et al. Age of onset influences clinical
are often preferred as maintenance therapy. However, if a features of chronic inflammatory demyelinating polyneuropa-
favorable response is not seen with one of these first-line thy. J Neurol Sci 2001;184(01):57–63
treatments, switching or escalating treatment, including 6 Querol L, Nogales-Gadea G, Rojas-Garcia R, et al. Neurofascin
IgG4 antibodies in CIDP associate with disabling tremor and poor
utilizing other immunomodulatory/chemotherapy, may
response to IVIg. Neurology 2014;82(10):879–886
offer benefit. CIDP appears to be a heterogeneous group of 7 Ogata H, Yamasaki R, Hiwatashi A, et al. Characterization of IgG4
disorders. While ample evidence points to a prominent cell- anti-neurofascin 155 antibody-positive polyneuropathy. Ann
mediated autoimmune process in most cases, a humoral- Clin Transl Neurol 2015;2(10):960–971

Seminars in Neurology Vol. 39 No. 5/2019


604 CIDP Shije and Brannagan

8 Devaux JJ, Miura Y, Fukami Y, et al. Neurofascin-155 IgG4 in study of a UK population. Muscle Nerve 2009;39(04):
chronic inflammatory demyelinating polyneuropathy. Neurol- 432–438
ogy 2016;86(09):800–807 27 Mahdi-Rogers M, Hughes RAC. Epidemiology of chronic inflam-
9 Kadoya M, Kaida K, Koike H, et al. IgG4 anti-neurofascin155 matory neuropathies in southeast England. Eur J Neurol 2014;21
antibodies in chronic inflammatory demyelinating polyradi- (01):28–33
culoneuropathy: Clinical significance and diagnostic utility of 28 Larsen JP, Kvåle G, Nyland H. Epidemiology of the Guillain-Barré
a conventional assay. J Neuroimmunol 2016;301:16–22 syndrome in the county of Hordaland, Western Norway. Acta
10 Doppler K, Appeltshauser L, Wilhelmi K, et al. Destruction of Neurol Scand 1985;71(01):43–47
paranodal architecture in inflammatory neuropathy with anti- 29 Schmidt B, Toyka KV, Kiefer R, Full J, Hartung HP, Pollard J.
contactin-1 autoantibodies. J Neurol Neurosurg Psychiatry 2015; Inflammatory infiltrates in sural nerve biopsies in Guillain-Barre
86(07):720–728 syndrome and chronic inflammatory demyelinating neuropathy.
11 Miura Y, Devaux JJ, Fukami Y, et al; CNTN1-CIDP Study Group. Muscle Nerve 1996;19(04):474–487
Contactin 1 IgG4 associates to chronic inflammatory demyeli- 30 Schneider-Hohendorf T, Schwab N, Uçeyler N, Göbel K, Sommer
nating polyneuropathy with sensory ataxia. Brain 2015;138(Pt C, Wiendl H. CD8þ T-cell immunity in chronic inflammatory
6):1484–1491 demyelinating polyradiculoneuropathy. Neurology 2012;78
12 Henderson RD, Sandroni P, Wijdicks EFM. Chronic inflammatory (06):402–408
demyelinating polyneuropathy and respiratory failure. J Neurol 31 Misawa S, Kuwabara S, Mori M, Kawaguchi N, Yoshiyama Y,
2005;252(10):1235–1237 Hattori T. Serum levels of tumor necrosis factor-alpha in chronic
13 Macia F, Le Masson G, Rouanet-Larriviere M, et al. A prospective inflammatory demyelinating polyneuropathy. Neurology 2001;
evaluation of phrenic nerve conduction in multifocal motor 56(05):666–669

Downloaded by: University of California, San Francisco. Copyrighted material.


neuropathy and chronic inflammatory demyelinating poly- 32 Beppu M, Sawai S, Misawa S, et al. Serum cytokine and chemo-
neuropathy. Muscle Nerve 2003;28(03):319–323 kine profiles in patients with chronic inflammatory demyelinat-
14 Ingall TJ, McLeod JG, Tamura N. Autonomic function and unmye- ing polyneuropathy. J Neuroimmunol 2015;279:7–10
linated fibers in chronic inflammatory demyelinating polyradi- 33 Rentzos M, Angeli AV, Rombos A, et al. Proinflammatory cyto-
culoneuropathy. Muscle Nerve 1990;13(01):70–76 kines in serum and cerebrospinal fluid of CIDP patients. Neurol
15 Lyu R-K, Tang L-M, Wu Y-R, Chen S-T. Cardiovascular autonomic Res 2012;34(09):842–846
function and sympathetic skin response in chronic inflamma- 34 Mei F-J, Ishizu T, Murai H, et al. Th1 shift in CIDP versus Th2 shift
tory demyelinating polyradiculoneuropathy. Muscle Nerve in vasculitic neuropathy in CSF. J Neurol Sci 2005;228(01):75–85
2002;26(05):669–672 35 Mathey EK, Pollard JD, Armati PJ. TNF alpha, IFN gamma and IL-2
16 Stamboulis E, Katsaros N, Koutsis G, Iakovidou H, Giannakopou- mRNA expression in CIDP sural nerve biopsies. J Neurol Sci 1999;
lou A, Simintzi I. Clinical and subclinical autonomic dysfunction 163(01):47–52
in chronic inflammatory demyelinating polyradiculoneuropa- 36 Salomon B, Rhee L, Bour-Jordan H, et al. Development of spon-
thy. Muscle Nerve 2006;33(01):78–84 taneous autoimmune peripheral polyneuropathy in B7-2-defi-
17 Sakakibara R, Hattori T, Kuwabara S, Yamanishi T, Yasuda K. cient NOD mice. J Exp Med 2001;194(05):677–684
Micturitional disturbance in patients with chronic inflamma- 37 Harvey GK, Pollard JD, Schindhelm K, Antony J. Chronic experi-
tory demyelinating polyneuropathy. Neurology 1998;50(04): mental allergic neuritis. An electrophysiological and histological
1179–1182 study in the rabbit. J Neurol Sci 1987;81(2-3):215–225
18 Figueroa JJ, Dyck PJB, Laughlin RS, et al. Autonomic dysfunction 38 McCombe PA, van der Kreek SA, Pender MP. The effects of
in chronic inflammatory demyelinating polyradiculoneuropa- prophylactic cyclosporin A on experimental allergic neuritis
thy. Neurology 2012;78(10):702–708 (EAN) in the Lewis rat. Induction of relapsing EAN using low
19 Ruts L, Drenthen J, Jacobs BC, van Doorn PA; Dutch GBS Study dose cyclosporin A. J Neuroimmunol 1990;28(02):131–140
Group. Distinguishing acute-onset CIDP from fluctuating Guil- 39 Yan WX, Archelos JJ, Hartung HP, Pollard JD. P0 protein is a target
lain-Barre syndrome: a prospective study. Neurology 2010;74 antigen in chronic inflammatory demyelinating polyradiculo-
(21):1680–1686 neuropathy. Ann Neurol 2001;50(03):286–292
20 Laughlin RS, Dyck PJB, Melton LJ III, Leibson C, Ransom J, Dyck 40 Meléndez-Vásquez C, Redford J, Choudhary PP, et al. Immunologi-
PJB. Incidence and prevalence of CIDP and the association of cal investigation of chronic inflammatory demyelinating polyra-
diabetes mellitus. Neurology 2009;73(01):39–45 diculoneuropathy. J Neuroimmunol 1997;73(1-2):124–134
21 Lunn MPT, Manji H, Choudhary PP, Hughes RAC, Thomas PK. 41 Gabriel CM, Gregson NA, Hughes RA. Anti-PMP22 antibodies in
Chronic inflammatory demyelinating polyradiculoneuropathy: patients with inflammatory neuropathy. J Neuroimmunol 2000;
a prevalence study in south east England. J Neurol Neurosurg 104(02):139–146
Psychiatry 1999;66(05):677–680 42 Sanvito L, Makowska A, Mahdi-Rogers M, et al. Humoral and
22 Mygland A, Monstad P. Chronic polyneuropathies in Vest-Agder, cellular immune responses to myelin protein peptides in chronic
Norway. Eur J Neurol 2001;8(02):157–165 inflammatory demyelinating polyradiculoneuropathy. J Neurol
23 Kusumi M, Nakashima K, Nakayama H, Takahashi K. Epidemiol- Neurosurg Psychiatry 2009;80(03):333–338
ogy of inflammatory neurological and inflammatory neuromus- 43 Khalili-Shirazi A, Atkinson P, Gregson N, Hughes RA. Antibody
cular diseases in Tottori Prefecture, Japan. Psychiatry Clin responses to P0 and P2 myelin proteins in Guillain-Barré syn-
Neurosci 1995;49(03):169–174 drome and chronic idiopathic demyelinating polyradiculo-
24 Chiò A, Cocito D, Bottacchi E, et al; PARCIDP. Idiopathic chronic neuropathy. J Neuroimmunol 1993;46(1-2):245–251
inflammatory demyelinating polyneuropathy: an epidemiologi- 44 Vural A, Doppler K, Meinl E. Autoantibodies against the node of
cal study in Italy. J Neurol Neurosurg Psychiatry 2007;78(12): Ranvier in seropositive chronic inflammatory demyelinating
1349–1353 polyneuropathy: diagnostic, pathogenic, and therapeutic rele-
25 McLeod JG, Pollard JD, Macaskill P, Mohamed A, Spring P, vance. Front Immunol 2018;9:1029
Khurana V. Prevalence of chronic inflammatory demyelinating 45 Albers JW, Kelly JJ Jr. Acquired inflammatory demyelinating
polyneuropathy in New South Wales, Australia. Ann Neurol polyneuropathies: clinical and electrodiagnostic features. Mus-
1999;46(06):910–913 cle Nerve 1989;12(06):435–451
26 Rajabally YA, Simpson BS, Beri S, Bankart J, Gosalakkal JA. 46 Cornblath DR, Asbury AK, Albers JW, et al. Research criteria for
Epidemiologic variability of chronic inflammatory demyeli- diagnosis of chronic inflammatory demyelinating polyneuropa-
nating polyneuropathy with different diagnostic criteria: thy (CIDP). Report from an ad hoc subcommittee of the American

Seminars in Neurology Vol. 39 No. 5/2019


CIDP Shije and Brannagan 605

Academy of Neurology AIDS Task Force. Neurology 1991;41(05): 65 Chin RL, Latov N, Sander HW, et al. Sensory CIDP presenting as
617–618 cryptogenic sensory polyneuropathy. J Peripher Nerv Syst 2004;
47 Bromberg MB. Comparison of electrodiagnostic criteria for pri- 9(03):132–137
mary demyelination in chronic polyneuropathy. Muscle Nerve 66 Berger AR, Herskovitz S, Kaplan J. Late motor involvement in
1991;14(10):968–976 cases presenting as “chronic sensory demyelinating polyneuro-
48 Saperstein DS, Katz JS, Amato AA, Barohn RJ. Clinical spectrum of pathy”. Muscle Nerve 1995;18(04):440–444
chronic acquired demyelinating polyneuropathies. Muscle 67 van Dijk GW, Notermans NC, Franssen H, Wokke JHJ. Develop-
Nerve 2001;24(03):311–324 ment of weakness in patients with chronic inflammatory
49 Latov N. Diagnosis of CIDP. Neurology 2002;59(12, Suppl 6): demyelinating polyneuropathy and only sensory symptoms at
S2–S6 presentation: a long-term follow-up study. J Neurol 1999;246
50 Nicolas G, Maisonobe T, Le Forestier N, Léger J-M, Bouche P. (12):1134–1139
Proposed revised electrophysiological criteria for chronic 68 Sinnreich M, Klein CJ, Daube JR, Engelstad J, Spinner RJ, Dyck PJB.
inflammatory demyelinating polyradiculoneuropathy. Muscle Chronic immune sensory polyradiculopathy: a possibly treata-
Nerve 2002;25(01):26–30 ble sensory ataxia. Neurology 2004;63(09):1662–1669
51 Hughes R, Bensa S, Willison H, et al; Inflammatory Neuropathy 69 Sharma KR, Cross J, Farronay O, Ayyar DR, Shebert RT, Bradley
Cause and Treatment (INCAT) Group. Randomized controlled WG. Demyelinating neuropathy in diabetes mellitus. Arch Neu-
trial of intravenous immunoglobulin versus oral prednisolone in rol 2002;59(05):758–765
chronic inflammatory demyelinating polyradiculoneuropathy. 70 Chiò A, Plano F, Calvo A, Leone M, Mutani R, Cocito D; Piemonte
Ann Neurol 2001;50(02):195–201 and Valle D’Aosta Registry for CIDP (PARCIDP). Comorbidity
52 Thaisetthawatkul P, Logigian EL, Herrmann DN. Dispersion of the between CIDP and diabetes mellitus: only a matter of chance?

Downloaded by: University of California, San Francisco. Copyrighted material.


distal compound muscle action potential as a diagnostic criter- Eur J Neurol 2009;16(06):752–754
ion for chronic inflammatory demyelinating polyneuropathy. 71 Gorson KC, Ropper AH, Adelman LS, Weinberg DH. Influence of
Neurology 2002;59(10):1526–1532 diabetes mellitus on chronic inflammatory demyelinating poly-
53 Magda P, Latov N, Brannagan TH III, Weimer LH, Chin RL, Sander neuropathy. Muscle Nerve 2000;23(01):37–43
HW. Comparison of electrodiagnostic abnormalities and criteria 72 Jann S, Bramerio MA, Facchetti D, Sterzi R. Intravenous immu-
in a cohort of patients with chronic inflammatory demyelinating noglobulin is effective in patients with diabetes and with chronic
polyneuropathy. Arch Neurol 2003;60(12):1755–1759 inflammatory demyelinating polyneuropathy: long term follow-
54 Berger AR, Bradley WG, Brannagan TH, et al; Neuropathy Asso- up. J Neurol Neurosurg Psychiatry 2009;80(01):70–73
ciation, Medical Advisory Committee. Guidelines for the diag- 73 Cornblath DR. Treatment of the neuromuscular complications of
nosis and treatment of chronic inflammatory demyelinating human immunodeficiency virus infection. Ann Neurol 1988;23
polyneuropathy. J Peripher Nerv Syst 2003;8(04):282–284 (Suppl):S88–S91
55 Van Schaik IN, Léger JM, Nobile-Orazio E, et al; Joint Task Force of 74 Ferrari S, Vento S, Monaco S, et al. Human immunodeficiency
the EFNS and the PNS. European Federation of Neurological virus-associated peripheral neuropathies. Mayo Clin Proc 2006;
Societies/Peripheral Nerve Society guideline on management 81(02):213–219
of multifocal motor neuropathy. Report of a joint task force of the 75 Przedborski S, Liesnard C, Voordecker P, et al. Inflammatory
European Federation of Neurological Societies and the Periph- demyelinating polyradiculoneuropathy associated with human
eral Nerve Society—first revision. J Peripher Nerv Syst 2010;15 immunodeficiency virus infection. J Neurol 1988;235(06):359–361
(04):295–301 76 Moodley K, Bill PLA, Patel VB. A comparative study of CIDP in a
56 Breiner A, Brannagan TH III. Comparison of sensitivity and cohort of HIV-infected and HIV-uninfected patients. Neurol
specificity among 15 criteria for chronic inflammatory demye- Neuroimmunol Neuroinflamm 2016;4(02):e315
linating polyneuropathy. Muscle Nerve 2014;50(01):40–46 77 Gorson KC, Allam G, Ropper AH. Chronic inflammatory demye-
57 Lewis RA, Sumner AJ, Brown MJ, Asbury AK. Multifocal demye- linating polyneuropathy: clinical features and response to treat-
linating neuropathy with persistent conduction block. Neurol- ment in 67 consecutive patients with and without a monoclonal
ogy 1982;32(09):958–964 gammopathy. Neurology 1997;48(02):321–328
58 Viala K, Renié L, Maisonobe T, et al. Follow-up study and response 78 Nobile-Orazio E, Latov N, Hays AP, et al. Neuropathy and anti-
to treatment in 23 patients with Lewis-Sumner syndrome. Brain MAG antibodies without detectable serum M-protein. Neurol-
2004;127(Pt 9):2010–2017 ogy 1984;34(02):218–221
59 Katz JS, Saperstein DS, Gronseth G, Amato AA, Barohn RJ. Distal 79 Latov N, Hays AP, Sherman WH. Peripheral neuropathy and anti-
acquired demyelinating symmetric neuropathy. Neurology MAG antibodies. Crit Rev Neurobiol 1988;3(04):301–332
2000;54(03):615–620 80 Nobile-Orazio E, Meucci N, Baldini L, Di Troia A, Scarlato G. Long-
60 Leitch MM, Sherman WH, Brannagan TH III. Distal acquired term prognosis of neuropathy associated with anti-MAG IgM M-
demyelinating symmetric polyneuropathy progressing to classic proteins and its relationship to immune therapies. Brain 2000;
chronic inflammatory demyelinating polyneuropathy and 123(Pt 4):710–717
response to fludarabine and cyclophosphamide. Muscle Nerve 81 Renaud S, Gregor M, Fuhr P, et al. Rituximab in the treatment of
2013;47(02):292–296 polyneuropathy associated with anti-MAG antibodies. Muscle
61 Gable KL, Afshari Z, Sufit RL, Allen JA. Distal acquired demyeli- Nerve 2003;27(05):611–615
nating symmetric neuropathy after vaccination. J Clin Neuro- 82 Nobile-Orazio E, Manfredini E, Carpo M, et al. Frequency and
muscul Dis 2013;14(03):117–122 clinical correlates of anti-neural IgM antibodies in neuropathy
62 Dyck PJ, Dyck PJB. Atypical varieties of chronic inflammatory associated with IgM monoclonal gammopathy. Ann Neurol
demyelinating neuropathies. Lancet 2000;355(9212):1293–1294 1994;36(03):416–424
63 Larue S, Bombelli F, Viala K, et al. Non-anti-MAG DADS neuro- 83 Garcia-Santibanez R, Zaidman CM, Sommerville RB, et al. CANO-
pathy as a variant of CIDP: clinical, electrophysiological, labora- MAD and other chronic ataxic neuropathies with disialosyl
tory features and response to treatment in 10 cases. Eur J Neurol antibodies (CANDA). J Neurol 2018;265(06):1402–1409
2011;18(06):899–905 84 Carpo M, Meucci N, Allaria S, et al. Anti-sulfatide IgM antibodies
64 Oh SJ, Joy JL, Kuruoglu R. “Chronic sensory demyelinating in peripheral neuropathy. J Neurol Sci 2000;176(02):144–150
neuropathy”: chronic inflammatory demyelinating polyneuro- 85 Guo X, Qin X, Zhang Y, Huang C, Yu G. Electrophysiological
pathy presenting as a pure sensory neuropathy. J Neurol Neu- features of POEMS syndrome and chronic inflammatory demye-
rosurg Psychiatry 1992;55(08):677–680 linating polyneuropathy. J Clin Neurosci 2014;21(04):587–590

Seminars in Neurology Vol. 39 No. 5/2019


606 CIDP Shije and Brannagan

86 Lozeron P, Mariani L-L, Dodet P, et al. Transthyretin amyloid 106 Hahn AF, Bolton CF, Zochodne D, Feasby TE. Intravenous immu-
polyneuropathies mimicking a demyelinating polyneuropathy. noglobulin treatment in chronic inflammatory demyelinating
Neurology 2018;91(02):e143–e152 polyneuropathy. A double-blind, placebo-controlled, cross-over
87 Bird SJ, Sladky JT. Corticosteroid-responsive dominantly inher- study. Brain 1996;119(Pt 4):1067–1077
ited neuropathy in childhood. Neurology 1991;41(03):437– 107 Hughes RAC, Donofrio P, Bril V, et al; ICE Study Group. Intrave-
439 nous immune globulin (10% caprylate-chromatography puri-
88 Donaghy M, Sisodiya SM, Kennett R, McDonald B, Haites N, Bell C. fied) for the treatment of chronic inflammatory demyelinating
Steroid responsive polyneuropathy in a family with a novel polyradiculoneuropathy (ICE study): a randomised placebo-
myelin protein zero mutation. J Neurol Neurosurg Psychiatry controlled trial. Lancet Neurol 2008;7(02):136–144
2000;69(06):799–805 108 Kaplan A, Brannagan TH III. Evaluation of patients with refrac-
89 Ginsberg L, Malik O, Kenton AR, et al. Coexistent hereditary and tory chronic inflammatory demyelinating polyneuropathy. Mus-
inflammatory neuropathy. Brain 2004;127(Pt 1):193–202 cle Nerve 2017;55(04):476–482
90 Lewis RA, Sumner AJ. The electrodiagnostic distinctions 109 Brannagan TH III, Nagle KJ, Lange DJ, Rowland LP. Complications
between chronic familial and acquired demyelinative neuropa- of intravenous immune globulin treatment in neurologic dis-
thies. Neurology 1982;32(06):592–596 ease. Neurology 1996;47(03):674–677
91 Rechthand E, Cornblath DR, Stern BJ, Meyerhoff JO. Chronic 110 Orbach H, Katz U, Sherer Y, Shoenfeld Y. Intravenous immuno-
demyelinating polyneuropathy in systemic lupus erythemato- globulin: adverse effects and safe administration. Clin Rev
sus. Neurology 1984;34(10):1375–1377 Allergy Immunol 2005;29(03):173–184
92 Gondim FAA, Brannagan TH III, Sander HW, Chin RL, Latov N. 111 Kuitwaard K, van den Berg LH, Vermeulen M, et al. Randomised
Peripheral neuropathy in patients with inflammatory bowel controlled trial comparing two different intravenous immunoglo-

Downloaded by: University of California, San Francisco. Copyrighted material.


disease. Brain 2005;128(Pt 4):867–879 bulins in chronic inflammatory demyelinating polyradiculoneuro-
93 Wong AHY, Kokubun N, Fukami Y, Miyaji K, Yuki N. Chronic pathy. J Neurol Neurosurg Psychiatry 2010;81(12):1374–1379
inflammatory demyelinating polyneuropathy with membra- 112 Gallia F, Balducci C, Nobile-Orazio E. Efficacy and tolerability of
nous nephropathy. J Peripher Nerv Syst 2015;20(01):63–66 different brands of intravenous immunoglobulin in the main-
94 Echaniz-Laguna A, de Séze J, Chanson J-B. Chronic inflammatory tenance treatment of chronic immune-mediated neuropathies.
demyelinating polyradiculoneuropathy in solid organ transplant J Peripher Nerv Syst 2016;21(02):82–84
recipients: a prospective study. J Neurol Neurosurg Psychiatry 113 Markvardsen LH, Sindrup SH, Christiansen I, Olsen NK, Jakobsen
2012;83(07):699–705 J, Andersen H; Danish CIDP and MMN Study Group. Subcuta-
95 Rajabally YA, Attarian S. Chronic inflammatory demyelinating neous immunoglobulin as first-line therapy in treatment-naive
polyneuropathy and malignancy: a systematic review. Muscle patients with chronic inflammatory demyelinating polyneuro-
Nerve 2018;57(06):875–883 pathy: randomized controlled trial study. Eur J Neurol 2017;24
96 Alshekhlee A, Basiri K, Miles JD, Ahmad SA, Katirji B. Chronic (02):412–418
inflammatory demyelinating polyneuropathy associated with 114 Markvardsen LH, Debost J-C, Harbo T, et al; Danish CIDP and
tumor necrosis factor-α antagonists. Muscle Nerve 2010;41(05): MMN Study Group. Subcutaneous immunoglobulin in respon-
723–727 ders to intravenous therapy with chronic inflammatory demye-
97 Kao JC, Liao B, Markovic SN, et al. Neurological complications linating polyradiculoneuropathy. Eur J Neurol 2013;20(05):
associated with anti-programmed death 1 (PD-1) antibodies. 836–842
JAMA Neurol 2017;74(10):1216–1222 115 Markvardsen LH, Harbo T, Sindrup SH, Christiansen I, Andersen
98 Kolb NA, Trevino CR, Waheed W, et al. Neuromuscular complica- H, Jakobsen J; Danish CIDP and MMN Study Group. Subcutaneous
tions of immune checkpoint inhibitor therapy. Muscle Nerve immunoglobulin preserves muscle strength in chronic inflam-
2018;58:10–22 matory demyelinating polyneuropathy. Eur J Neurol 2014;21
99 Mathis S, Magy L, Diallo L, Boukhris S, Vallat J-M. Amyloid (12):1465–1470
neuropathy mimicking chronic inflammatory demyelinating 116 van Schaik IN, Bril V, van Geloven N, et al; PATH study group.
polyneuropathy. Muscle Nerve 2012;45(01):26–31 Subcutaneous immunoglobulin for maintenance treatment in
100 Dyck PJ, O’Brien PC, Oviatt KF, et al. Prednisone improves chronic chronic inflammatory demyelinating polyneuropathy (PATH): a
inflammatory demyelinating polyradiculoneuropathy more randomised, double-blind, placebo-controlled, phase 3 trial.
than no treatment. Ann Neurol 1982;11(02):136–141 Lancet Neurol 2018;17(01):35–46
101 Lopate G, Pestronk A, Al-Lozi M. Treatment of chronic inflam- 117 Racosta JM, Sposato LA, Kimpinski K. Subcutaneous versus
matory demyelinating polyneuropathy with high-dose intermit- intravenous immunoglobulin for chronic autoimmune neuropa-
tent intravenous methylprednisolone. Arch Neurol 2005;62(02): thies: a meta-analysis. Muscle Nerve 2017;55(06):802–809
249–254 118 Dyck PJ, Litchy WJ, Kratz KM, et al. A plasma exchange versus
102 Muley SA, Kelkar P, Parry GJ. Treatment of chronic inflammatory immune globulin infusion trial in chronic inflammatory demye-
demyelinating polyneuropathy with pulsed oral steroids. Arch linating polyradiculoneuropathy. Ann Neurol 1994;36(06):
Neurol 2008;65(11):1460–1464 838–845
103 van Schaik IN, Eftimov F, van Doorn PA, et al. Pulsed high-dose 119 Hahn AF, Bolton CF, Pillay N, et al. Plasma-exchange therapy in
dexamethasone versus standard prednisolone treatment for chronic inflammatory demyelinating polyneuropathy. A double-
chronic inflammatory demyelinating polyradiculoneuropathy blind, sham-controlled, cross-over study. Brain 1996;119(Pt
(PREDICT study): a double-blind, randomised, controlled trial. 4):1055–1066
Lancet Neurol 2010;9(03):245–253 120 Hughes RA, Mehndiratta MM, Rajabally YA. Corticosteroids for
104 Nobile-Orazio E, Cocito D, Jann S, et al; IMC Trial Group. chronic inflammatory demyelinating polyradiculoneuropathy.
Intravenous immunoglobulin versus intravenous methylpredni- Cochrane Database Syst Rev 2017;11:CD002062
solone for chronic inflammatory demyelinating polyradiculo- 121 Eftimov F, Winer JB, Vermeulen M, de Haan R, van Schaik IN.
neuropathy: a randomised controlled trial. Lancet Neurol 2012; Intravenous immunoglobulin for chronic inflammatory demye-
11(06):493–502 linating polyradiculoneuropathy. Cochrane Database Syst Rev
105 Mendell JR, Barohn RJ, Freimer ML, et al; Working Group on 2013;12(12):CD001797
Peripheral Neuropathy. Randomized controlled trial of IVIg in 122 Mehndiratta MM, Hughes RAC, Pritchard J. Plasma exchange for
untreated chronic inflammatory demyelinating polyradiculo- chronic inflammatory demyelinating polyradiculoneuropathy.
neuropathy. Neurology 2001;56(04):445–449 Cochrane Database Syst Rev 2015;8(08):CD003906

Seminars in Neurology Vol. 39 No. 5/2019


CIDP Shije and Brannagan 607

123 Cocito D, Paolasso I, Antonini G, et al; Italian Network for CIDP inflammatory demyelinating polyradiculoneuropathy (RMC
Register. A nationwide retrospective analysis on the effect of trial): a pilot, multicentre study. Lancet Neurol 2009;8(02):
immune therapies in patients with chronic inflammatory 158–164
demyelinating polyradiculoneuropathy. Eur J Neurol 2010;17 139 Hughes R, Dalakas MC, Merkies I, et al; FORCIDP Trial Investiga-
(02):289–294 tors. Oral fingolimod for chronic inflammatory demyelinating
124 Cocito D, Grimaldi S, Paolasso I, et al; Italian Network for CIDP polyradiculoneuropathy (FORCIDP Trial): a double-blind, multi-
Register. Immunosuppressive treatment in refractory chronic centre, randomised controlled trial. Lancet Neurol 2018;17(08):
inflammatory demyelinating polyradiculoneuropathy. A nation- 689–698
wide retrospective analysis. Eur J Neurol 2011;18(12):1417–1421 140 Mahdi-Rogers M, Brassington R, Gunn AA, van Doorn PA, Hughes
125 Bedi G, Brown A, Tong T, Sharma KR. Chronic inflammatory RA. Immunomodulatory treatment other than corticosteroids,
demyelinating polyneuropathy responsive to mycophenolate immunoglobulin and plasma exchange for chronic inflammatory
mofetil therapy. J Neurol Neurosurg Psychiatry 2010;81(06): demyelinating polyradiculoneuropathy. Cochrane Database Syst
634–636 Rev 2017;5:CD003280
126 Fialho D, Chan YC, Allen DC, Reilly MM, Hughes RA. Treatment of 141 Marsh EA, Hirst CL, Llewelyn JG, et al. Alemtuzumab in the
chronic inflammatory demyelinating polyradiculoneuropathy treatment of IVIG-dependent chronic inflammatory demyelinat-
with methotrexate. J Neurol Neurosurg Psychiatry 2006;77 ing polyneuropathy. J Neurol 2010;257(06):913–919
(04):544–547 142 Ahlmén J, Andersen O, Hallgren G, Peilot B. Positive effects of
127 Gladstone DE, Prestrud AA, Brannagan TH III. High-dose cyclo- tacrolimus in a case of CIDP. Transplant Proc 1998;30(08):4194
phosphamide results in long-term disease remission with 143 Renard D, Gauthier T, Venetz J-P, Buclin T, Kuntzer T. Late onset
restoration of a normal quality of life in patients with severe tacrolimus-induced life-threatening polyneuropathy in a kidney

Downloaded by: University of California, San Francisco. Copyrighted material.


refractory chronic inflammatory demyelinating polyneuropa- transplant recipient patient. Clin Kidney J 2012;5(04):323–326
thy. J Peripher Nerv Syst 2005;10(01):11–16 144 Chin RL, Sherman WH, Sander HW, Hays AP, Latov N. Etanercept
128 Matsuda M, Hoshi K, Gono T, Morita H, Ikeda S. Cyclosporin A in (Enbrel) therapy for chronic inflammatory demyelinating poly-
treatment of refractory patients with chronic inflammatory neuropathy. J Neurol Sci 2003;210(1-2):19–21
demyelinating polyradiculoneuropathy. J Neurol Sci 2004;224 145 Oyama Y, Sufit R, Loh Y, et al. Nonmyeloablative autologous
(1-2):29–35 hematopoietic stem celltransplantation for refractory CIDP.
129 Odaka M, Tatsumoto M, Susuki K, Hirata K, Yuki N. Intractable Neurology 2007;69(18):1802–1803
chronic inflammatory demyelinating polyneuropathy treated 146 Vermeulen M, Van Oers MH. Successful autologous stem cell
successfully with ciclosporin. J Neurol Neurosurg Psychiatry transplantation in a patient with chronic inflammatory demye-
2005;76(08):1115–1120 linating polyneuropathy. J Neurol Neurosurg Psychiatry 2002;72
130 Gorson KC, Allam G, Simovic D, Ropper AH. Improvement (01):127–128
following interferon-alpha 2A in chronic inflammatory demye- 147 Vermeulen M, van Oers MHJ. Relapse of chronic inflammatory
linating polyneuropathy. Neurology 1997;48(03):777–780 demyelinating polyneuropathy 5 years after autologous stem
131 Pavesi G, Cattaneo L, Marbini A, Gemignani F, Mancia D. Long- cell transplantation. J Neurol Neurosurg Psychiatry 2007;78(10):
term efficacy of interferon-alpha in chronic inflammatory 1154
demyelinating polyneuropathy. J Neurol 2002;249(06):777–779 148 Press R, Askmark H, Svenningsson A, et al. Autologous haema-
132 Vallat J-M, Hahn AF, Léger J-M, et al. Interferon beta-1a as an topoietic stem cell transplantation: a viable treatment option for
investigational treatment for CIDP. Neurology 2003;60(08, CIDP. J Neurol Neurosurg Psychiatry 2014;85(06):618–624
Suppl 3):S23–S28 149 Garg N, Park SB, Yiannikas C, et al. Neurofascin-155 IGG4
133 Benedetti L, Briani C, Franciotta D, et al. Rituximab in patients neuropathy: pathophysiological insights, spectrum of clinical
with chronic inflammatory demyelinating polyradiculoneuro- severity and response to treatment. Muscle Nerve 2018;57(05):
pathy: a report of 13 cases and review of the literature. J Neurol 848–851
Neurosurg Psychiatry 2011;82(03):306–308 150 Querol L, Nogales-Gadea G, Rojas-Garcia R, et al. Antibodies to
134 Roux T, Debs R, Maisonobe T, et al. Rituximab in chronic contactin-1 in chronic inflammatory demyelinating polyneuro-
inflammatory demyelinating polyradiculoneuropathy with pathy. Ann Neurol 2013;73(03):370–380
associated diseases. J Peripher Nerv Syst 2018;23(04):235–240 151 Burnor E, Yang L, Zhou H, et al. Neurofascin antibodies in
135 Dyck PJ, O’Brien P, Swanson C, Low P, Daube J. Combined autoimmune, genetic, and idiopathic neuropathies. Neurology
azathioprine and prednisone in chronic inflammatory-demye- 2018;90(01):e31–e38
linating polyneuropathy. Neurology 1985;35(08):1173–1176 152 Querol L, Rojas-García R, Diaz-Manera J, et al. Rituximab in
136 Hadden RDM, Sharrack B, Bensa S, Soudain SE, Hughes RAC. treatment-resistant CIDP with antibodies against paranodal
Randomized trial of interferon beta-1a in chronic inflammatory proteins. Neurol Neuroimmunol Neuroinflamm 2015;2(05):
demyelinating polyradiculoneuropathy. Neurology 1999;53 e149
(01):57–61 153 Gorson KC, van Schaik IN, Merkies ISJ, et al. Chronic inflamma-
137 Hughes RAC, Gorson KC, Cros D, et al; Avonex CIDP Study Group. tory demyelinating polyneuropathy disease activity status:
Intramuscular interferon beta-1a in chronic inflammatory recommendations for clinical research standards and use in
demyelinating polyradiculoneuropathy. Neurology 2010;74 clinical practice. J Peripher Nerv Syst 2010;15(04):326–333
(08):651–657 154 Remenyi P, Masszi T, Borbenyi Z, et al. CIDP cured by allogenic
138 Mahdi-Rogers M, Rutterford C, Hughes RAC, et al; RMC Trial hematopoietic stem cell transplantation. Eur J Neurol 2007;14
Group. Randomised controlled trial of methotrexate for chronic (08):e1–e2

Seminars in Neurology Vol. 39 No. 5/2019

You might also like