Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Special Report

For reprint orders, please contact reprints@expert-reviews.com

Chronic inflammatory
demyelinating polyneuropathy:
diagnosis andmanagement
Expert Rev. Clin. Immunol. 6(3), 373380 (2010)

Eduardo A De Sousa Over the course of 8weeks, a 50year-old man developed progressive bilateral leg and arm
Neuromuscular Medicine, Department weakness, with numbness and tingling of the feet and hands. His symptoms persisted for 6months,
of Neurology, Jefferson Medical with impaired manual dexterity, arm weakness when brushing his teeth, tripping when walking,
College, Thomas Jefferson University, inability to climb stairs and gait imbalance. On examination, there is mild proximal and distal
900 Walnut Street, Ste 200,
Philadelphia, PA 19107, USA
weakness of the upper and lower extremity muscles, length-dependent sensory loss of vibratory
Tel.: +1 215 955 7952 perception and joint position sense, areflexia, positive Romberg test and steppage gait with bilateral
Fax: +1 215 955 9976 foot drop. Motor nerve conduction studies of the arms and legs show partial conduction blocks in
eduardo.desousa@jefferson.edu several nerves with nonuniform slowing, and sensory responses are absent in the hands, however,
normal sural responses are noted. Lumbar puncture reveals acellular cerebrospinal fluid with elevated
protein. After 2 months following treatment, his strength and gait improved significantly, and his
sensory symptoms resolved.

Keywords : acquired chronic inflammatory demyelinating polyneuropathy demyelinating demyelination


inflammatory neuropathy peripheral polyneuropathy polyradiculoneuropathy weakness

Chronic inflammatory demyelinating polyneuro Depending on the population studied and


pathy (CIDP) is a treatable, potentially disa- the diagnostic criteria employed, the incidence
bling, acquired immune-mediated disease of the of CIDP may be as high as GuillainBarr
peripheral nerves and/or their roots. In 1929, syndrome (acute inflammatory demyelinat-
Harris and Newcomb described pathological ing polyneuropathy [AIDP]), at approximately
peripheral nerve demyelination and remyelin 1.6 per 100,000 [13] or as low as 0.15 per
ation in adults with recurrent or progressive 100,000[14] . The prevalence varies from 1.9 to
nonfamilial hypertrophic neuropathies [1] . In 8.9 per 100,000 [1318] and CIDP can affect all
1958, Austin described relapsing polyradiculo age groups, men slightly more so than women.
neuropathy responsive to corticosteroids [2] Clinical presentation may vary with age [19] ,
and in 1969, Thomas reported recurrent with either more motor or more sensory dis-
and chronic relapsing GuillainBarr poly turbance and, as is common in any peripheral
neuritis [3] . The term chronic inflammatory neuropathy, reflexes are often hypoactive or
polyradiculoneuropathy was coined in 1975 absent. CIDP and multiple sclerosis (MS) have
by Peter Dyck [4] . The AIDS taskforce from some similarities: both are acquired demyeli-
the American Academy of Neurology published nating diseases, and both can have secondary
research diagnostic guidelines for CIDP [5] , axonal injury from the demyelinating process,
which are frequently used owing to their high leading to more permanent disability (axonal
specificity applicable for research, however, neuropathic component in CIDP and black
they may be of low sensitivity when applied in holes in MS). In CIDP, immune-mediated
clinical practice[6,7] , and this could leave some attack occurs against peripheral myelin pro-
patients undiagnosed without early treatment. duced by Schwann cells, while in MS it is CNS
Delay in treatment may lead to axonal damage myelin from oligodendrocytes. Unlike MS,
with increased morbidity [8] . Several diagnostic CIDP does not have much of a well-defined geo-
criteria have been proposed [4,5,912] , and dif- graphic variation[13,14,16,18] . There are reports of
fer mainly in electrod iagnostic definitions and patients with acquired peripheral and central
the number of demyelinating findings on nerve demyelination syndromes [20] , although these
conductionstudies. are not common.

www.expert-reviews.com 10.1586/ECI.10.13 2010 Expert Reviews Ltd ISSN 1744-666X 373


Special Report De Sousa

Clinical features CIDP spectrum & ancillary tests


Motor dysfunction Chronic inflammatory demyelinating polyneuropathy can have
Typical and atypical CIDP is defined based on the distribution diverse presentations [31] , such as purely sensory [32,33] , cranial
of clinical features. Typical CIDP has symmetric painless proxi- nerve symptoms [34] or pain [35] . Nerve conduction studies may
mal and distal weakness. This pattern resembles proximal weak- unmask additional motor or sensory neuropathic dysfunction.
ness from motor neuron disease (progressive muscular atrophy or When presenting with purely motor signs, symptoms, and/or
adult onset of spinal muscular atrophy), neuromuscular junction electrophysiology, CIDP must be distinguished from multifocal
disease (myasthenia gravis) or muscle disease (myopathies). The motor neuropathy with or without conduction block [36] , which,
presence of sensory dysfunction cannot be due to motor neuron, compared with CIDP, has a different treatment response, and may
neuromuscular junction or muscle disease, and is more suggestive worsen with treatment with corticosteroids [37] or plasma exchange
ofCIDP. (PLEX) [38] , but may respond to intravenous immunoglobulin
Atypical CIDP can present as distal or multifocal disease [9] (IVIg) [39] .
and treatment responses may be similar to typical CIDP [21,22] . Although onset is defined as at least over 8weeks, CIDP may
Distal CIDP is characterized by a lack of proximal weakness, present acutely as GuillainBarr syndrome (acute-onset CIDP),
predominantly sensory length-dependent dysfunction, with or with an eventual progressive or relapsing chronic course [40,41] .
without distal weakness [23] . Distal weakness may be mild with This must be distinguished from treatment-related fluctuations
toe extension weakness or more pronounced with foot drop, and of GuillainBarr syndrome [42] .
may mimic the more common distal symmetric axonal poly Chronic inflammatory demyelinating polyneuropathy may
neuropathy that is associated with diabetes and thyroid disease, also develop in patients with a previously existing hereditary
among other causes. neuropathy, such as Charcot-Marie-Tooth disease (CMT)[43,44] ,
Multifocal CIDP [24,25] is characterized by sensory disturbance when there is a clear change in symptoms and signs. Diabetes
and asymmetric weakness resembling the clinical pattern of vas- mellitus does not appear to be a major risk factor for the develop
culitic neuropathy (mononeuritis multiplex) associated with mixed ment of CIDP [13,45] , despite earlier reports[46] , but individuals
cryoglobulinemia (particularly hepatitis C), systemic lupus ery- with diabetes mellitus can develop CIDP, which is a treatable
thematosus, Wegeners granulomatosis, ChurgStrauss syndrome condition even in the context of diabetes[47] . CIDP may develop
or polyarteritis nodosa [26] . However, vasculitic neuropathy needs as a complication of IFN-a therapy for hepatitisC patients [4850] ,
more aggressive and urgent treatment in order to avoid additional although paradoxically there are reports of IFN-a being used
infarctions or inflammatory lesions in organ systems other than as a successful treatment of CIDP patients with hepatitis C [51] .
the peripheral nerves. Patients presenting with peripheral neuropathy, including
Without nerve conduction studies, atypical CIDP may go patients with CIDP, may have associated conditions detectable
undiagnosed, especially when weakness is only distal and the via blood tests. Diabetes mellitus is the most common cause of
clinical pattern is thought to be of a common distal symmetric peripheral neuropathy worldwide. Other treatable disorders include
axonal polyneuropathy. Distal CIDP was reported as being more hypothyroidism, vitamin B12 deficiency and viral infections, such
common in older CIDP patients [27] . Nerve biopsy may be useful as HIV and hepatitis C. Laboratory tests for patients with symp-
in atypical CIDP, particularly when mononeuritis multiplex or toms of peripheral neuropathy include a 2-h oral glucose tolerance
vasculitic neuropathy is considered [28] . test, serum thyroid-stimulating hormone with reflex thyroxine,
and vitamin B12 with methylmalonic acid. Based on the January
Sensory dysfunction 2010 American Diabetes Association guidelines, the results of a
Sensory symptoms of CIDP can also mimic those of distal sensory standardized glycohemoglobin (or hemoglobin A1C) test allow
symmetric polyneuropathy, with length-dependent numbness, tin- the diagnosis of diabetes (6.5% or greater) or at risk for diabetes
gling or other dysesthesias and, rarely, pain. These sensory symptoms (5.76.4%) [52] . The presence of a monoclonal protein (Mpro-
can be slightly asymmetric or multifocal. On sensory examination, tein) may be detected with serum protein electrophoresis, although
vibratory perception and joint position sense are reduced or absent at serum and urine immunoelectrophoresis with immunofixation are
the toes, ankles and fingers. When more severe, vibratory perception preferred [53] . Quantitative immunoglobulins may also be help-
may be absent or impaired more proximally at fibular heads, knees, ful. Additional tests depend on the clinical context, and include
wrists or elbows. A useful bedside tool is the RydelSeiffer graduated tests for Lyme antibody, hepatitis B surface antigen, hepatitis C
tuning fork, with published normative data [29] and high inter- and virus antibody and HIV, along with ELISA and reflex Western
intra-rater reliability [30] . Length-dependent reduced sensation to blot, C-reactive protein, anti-nuclear antibody test (ANA), extract-
pinprick, cool temperature and light touch is common in CIDP. able nuclear antigens test (ENA), angiotensin-converting enzyme
Sensory ataxia may be seen in both the typical sensorimotor (ACE) test, anti-gliadin and tissue transglutaminase antibodies
CIDP as well as in the sensory variant of CIDP. Neurologists are (celiac antibodies) tests, among others. Serum-free light chains may
trained to distinguish peripheral from CNS dysfunction on clini- have a complementary role in the evaluation of Mproteins, and
cal examination and, when not clear, CNS pathology (particularly although not a substitute for serum or urine protein electrophoresis,
spinal cord disease affecting the dorsal columns) must be excluded the test for these has been used by hematologists for monitoring
with MRI of the neuroaxis. and diagnosing k or l chain disorders, such as multiple myeloma

374 Expert Rev. Clin. Immunol. 6(3), (2010)


CIDP: diagnosis &management Special Report

and amyloid light chain amyloidosis [54,55] , however, its role in the Fas-mediated T-cell apoptosis may distinguish acute CIDP from
evaluation of CIDP patients is less well defined. Consultation with GuillainBarr syndrome based on a recent study, but the technique
a hematologist and bone marrow biopsy should be considered in is complex, requiring cell cultures and analysis of Fas function over
patients with Mproteins. When a monoclonal protein is identified, a time period of more than 2weeks [62] .
bone lesions should be evaluated by skeletal survey or Tc99m ses- For clinicians not familiar with neurophysiology, electro
tamibi scan, which are more useful for the detection of plasmocy- diagnostic tests, such as nerve conduction studies, present results for
tomas than conventional bone scans [53] . Even when the Mprotein CIDP that can be explained as somewhat similar to the electrophysi-
is presumed to be essential (monoclonal gammopathy of uncertain ologic findings present in hereditary demyelinating neuropathies
significance), diagnostic workup should be repeated periodically like CMT, but instead of a generalized (uniform) process affecting
because of the risk of transformation into less benign plasma cell all nerves similarly, patchy involvement analogous to Guillain
disorders. Conditions associated with an Mprotein and peripheral Barr syndrome (AIDP) is seen in CIDP, with nonuniform demy-
neuropathy include the POEMS or CrowFukase syndrome elinating findings including prominent slowing or prolongation of
(polyneuropathy, organomegaly, endocrinopathy, M-protein and conduction velocities, distal latencies and F-wave responses. As a
skin abnormalities syndrome), osteosclerotic myeloma, multiple rule, differently from CMT, acquired demyelinating neuropathies
myeloma syndrome, Waldemstrom macroglobulinemia, chronic such as CIDP may reveal abnormal temporal dispersion and persist-
lymphocytic leukemia or related lymphoma, among others. In ent conduction block. These are thought to be electrical signatures
POEMS syndrome, VEGF levels are frequently elevated, and nor- of focal acquired demyelinating pathology causing differential slow-
mal levels raise suspicion for an alternative diagnosis, particularly ing and interruption of normal physiologic saltatory conduction;
when an Mprotein is not present [56] . however, there are rare reports of CMT with partial conduction
Chronic inflammatory demyelinating polyneuropathy with IgG block. Several criteria exist for the electrodiagnosis of CIDP [5,9,12] .
or IgA Mproteins may respond to therapy similarly to idiopathic Cerebrospinal fluid commonly shows albuminocytologic dis-
CIDP [53] . The presence of an IgM monoclonal protein may herald sociation, with elevated CSF protein and normal cell count. In
the diagnosis of a different type of neuropathy, such as anti-myelin- CIDP with HIV, cell count may be elevated.
associated glycoprotein (anti-MAG) neuropathy, with or without Imaging studies may show diffuse or focal enlargement of nerves
Waldenstrms macroglobulinemia. Anti-MAG antibodies should or roots with MRI [63] or ultrasonography [64,65] . Nerve hyper-
be tested for, especially when nerve conductions show severely pro- trophy in CIDP may be sufficiently profound to cause lumbar
longed distal latencies in most or all tested motor nerves, with less stenosis [66] or even cord compression and long tract signs [67] .
frequent distal temporal dispersion than CIDP [57] . Patients with Ultrasound studies are particularly attractive because of the pos-
IgM monoclonal proteins and/or anti-MAG antibodies usually do sibility of almost concomitant image testing along with electro-
not respond well to standard therapies for CIDP (corticosteroids, diagnostic testing (nerve conduction studies).
PLEX or IVIg), but may respond to rituximab [58] . Nerve biopsies may be nondiagnostic owing to the patchy
Antiganglioside antibodies, such as anti-GM1 antibodies, are nature of the disease: the biopsied nerve segment may not show
more useful for the diagnosis of multifocal motor neuropathy inflammation or demyelination. When abnormal, common find-
(with GM1-IgM antibodies) or acute motor axonal neuropathy ings on light microscopy are endoneurial edema and onion bulbs,
(the axonal form of GuillainBarr syndrome, with GM1-IgG with inflammation ranging from 0 to 55% in different series [4,68] .
antibodies). GM1 antibodies are not specific for motor neuro Teased nerve fiber preparation may reveal segmental demyeli-
pathies, and can be present in postpolio syndrome, previous para- nation, remyelination or variability of fiber thickness. Electron
lytic polio, GuillainBarr syndrome, motor neuron disease and microscopy may show myelin stripping or naked axons.
even healthy controls [59] .
When patients with presumed CIDP are refractory to standard Differential diagnosis/associated conditions
therapy, familial amyloid polyneuropathy with transthyretin gene Once the diagnosis of CIDP has been established, associated con-
mutation (TTR-FAP) should be considered in the differential ditions must be evaluated, since CIDP could be a secondary mani-
diagnosis, because such patients may be mistakenly thought to festation of another disease. However, the presence of elevated
have CIDP [60] . Liver transplant may be helpful but does not autoantibodies or antibodies against infections of the IgG class
prevent deterioration in all patients. should be interpreted cautiously in CIDP patients when the test
Testing for anti-Hu antibodies may not be useful in the context of is performed after IVIg therapy, since these may originate from
CIDP with motor weakness, given that the anti-Hu syndrome does the infused IVIg and may not be produced by the patient. The
not cause a sensorimotor neuropathy, but rather a purely sensory half-life of IgG antibodies is approximately 30days, but it may
neuronopathy (ganglionopathy) without motor weakness. Anti-Hu be longer in primary immunodeficiencies [69] .
syndrome may also present as a cerebellar syndrome. This expensive
blood test is therefore unnecessary for typical CIDP patients. Treatment
Biomarkers for the diagnosis of CIDP or for disease activity are The goal of treatment is to establish functional recovery, mini-
not yet well established, but promising results have recently been mizing secondary axonal injury. This may be accomplished with
published for peripheral blood flow cytometry analysis of tran- immunomodulatory treatments that may prevent further demy-
scription factor expression in CD4 + Tlymphocytes[61] . Defective elinating immune-mediated attack. Analogous to CNS disease

www.expert-reviews.com 375
Special Report De Sousa

in stroke and MS, time is nerve, and secondary axonal injury from over a decade ago suggest that approximately 90% of all
may lead to disability [8] . First-line (standard) treatments are IVIg, CIDP patients initially respond to treatment, and that favorable
PLEX and corticosteroids: the first two have demonstrated effi- prognostics factors include milder weakness at onset and symptoms
cacy in randomized placebo-controlled trials, and the latter was for less than a year [74,75] , which probably relates to less secondary
shown to be effective in a placebo-controlled trial [12] . Evidence of axonal injury. Partial or complete remission after stopping medi-
superiority of one treatment over another is lacking. Also lacking is cation treatment was seen in 40% [76] and 6387% walk without
detailed information on the use of more than one treatment simul- assistance [4,14,15] . Recent 5-year follow-up suggests that the long-
taneously, even though this may be carried out in some refractory term prognosis is favorable, although approximately 39% needed
cases. Corticosteroids can cause well-established short- and long- continued immunomodulatory therapy and approximately 13%
term side effects, including psychosis, gastrointestinal bleeding, had severe disability [77] .
osteopenia/osteoporosis and premature cataracts.
Plasma exchange requires sophisticated machinery available in Expert commentary
hospital settings, while IVIg can be administered at home. PLEX Chronic inflammatory demyelinating polyneuropathy is usually
can be given as the initial treatment for CIDP, but its use for sta- a treatment-responsive peripheral neuropathy that can be diag-
bilization as maintenance therapy presents disadvantages, includ- nosed based on clinical examination and nerve conduction studies.
ing bleeding from reduced clotting factors, difficulty with venous Additional tests include nerve biopsies and imaging studies, which
access, use of citrate and hemodynamic changes [12] . A Cochrane are not mandatory but may give additional supportive evidence.
review of the subject reports that, based on double-blind rand- A biological marker for diagnosis and disease activity is not well
omized controlled trials, PLEX can be given as ten exchanges over established, but would be extremely useful. Once diagnosed, CIDP
4weeks or sixexchanges over 3weeks [70] . In my practice, I often use patients must be assessed for secondary causes, which are less com-
three to five plasma exchanges, given on alternate days, as tolerated. mon than essential CIDP but may have serious implicationsespe-
Common side effects of IVIg include flu-like symptoms, rash cially when associated with malignancy or active viral infection,
and headaches from aseptic meningitis. IVIg infusions can be such as hepatitis C or HIV. Patients typically respond to corticos-
given at the initial dose of 2g/kg over 35days, followed by teroids, IVIg or PLEX, and failure of an initial standard treatment
maintenance doses of 2g/kg every 4weeks, 1g/kg every 3weeks modality should lead to a trial of another standard therapy, until
or 0.5g/kg every 2weeks. The landmark ICE study (10% capr- all three standard modalities have been attempted. Patients that
ylate-chromatography purified immune globulin intravenous do no respond to any of the three should have their diagnosis of
CIDP efficacy study) demonstrated the short- and long-term CIDP questioned and, if no alternative diagnosis arises, combina-
benefits of IVIg [71] . In my practice, doses are administered until tion treatment or nonstandard therapies could be attempted. An
maximal improvement is achieved, and then tapering down of important mimicker is a form of familial amyloidosis (TTR-FAP).
dose and/or frequency is attempted. Targeted immunomodulating therapies are not yet well defined
A third of the patients who fail one of the standard three for CIDP.
treatment modalities will respond to a second or third standard
modality [72] . Five-year view
In stable CIDP patients, maintenance treatment may not need Some of the anticipated diagnostic developments for CIDP
to be continued indefinitely, although this should be individual- include accurate biomarkers for early disease identification and
ized for each patient. Once optimal results are achieved after a monitoring of disease activity. In 5years time, clinicians and
prolonged course, I use the lowest effective g/kg dose or the least scientists are likely to have a better understanding of CIDP at the
frequent treatment interval for maintenance before I attempt to molecular level and targets other than peripheral nerve myelin
discontinue therapy. may be unveiled. It may be possible to recognize predisposed sub-
jects (host factors) and predisposing conditions (environmental
CIDP refractory to standard treatment factors) associated with CIDP.
When one or more combined standard treatments fail, less well- As we increase our understanding of the pathophysiology of
studied alternative treatments may be used, including myco secondary axonal injury, prevention or reduction of disability
phenolate, azathioprine, IFN-a, cyclosporine and monthly or will be tangible.
high-dose cyclophosphamide, among others. These should only Studying larger patient cohorts with standardized methods will
be prescribed by physicians with experience in immunosuppressive determine whether dividing or combining CIDP subcategories
therapy and for patients who can be closely monitored [73] . would result in different patient outcomes.
Ultrasonography, among other real-time imaging techniques,
Prognosis will become more widely available as a bedside tool for the diag-
The original report from Dyck in 1975 suggested that complete nosis and monitoring of disease activity. Establishing imaging
recovery was infrequent, 60% were able to walk and work, 25% end points for clinical trials will allow investigators to monitor
were wheelchair or bed bound and 10% died of their disease [4] . At patients more closely in shorter time intervals as it is a noninvasive
present, with increased early diagnostic awareness and treatment and painless test. Other noninvasive real-time imaging techniques
availability, current figures are probably more optimistic. Reports may be developed for neuromuscular disorders.

376 Expert Rev. Clin. Immunol. 6(3), (2010)


CIDP: diagnosis &management Special Report

Functional evaluation of peripheral nerves with electro Information resources


diagnostic tests will remain a key factor for the diagnosis of The Neuropathy Association: www.neuropathy.org
CIDP. Nerve conduction studies are unique for testing nerve
function. New electrodiagnostic techniques and re-evaluation CIDP International Foundation: www.cidpusa.org
of current methods may be developed as the techniques for real- GBS/CIDP Foundation International: www.gbs-cidp.org
time imaging become used more frequently alongside nerve
conduction studies.
Finer modulation of the misguided immune-mediated attack Financial & competing interests disclosure
against key phases of peripheral nerve damage will be accom- The author has no relevant affiliations or financial involvement with any
plished as targeted therapies are developed, such as better mono- organization or entity with a financial interest in or financial conflict with
clonal antibodies, small molecule therapies and antisense oligo- the subject matter or materials discussed in the manuscript. This includes
nucleotides. Corticosteroids may still be used in the next 5years, employment, consultancies, honoraria, stock ownership or options, expert
but earlier intervention to maximize bone health and minimize testimony, grants or patents received or pending, or royalties.
side effects should become more widespread. No writing assistance was utilized in the production of this manuscript.

Key issues
In chronic inflammatory demyelinating polyneuropathy (CIDP), proximal limb weakness with sensory signs or symptoms is common
and can be treated with immunomodulatory therapy. Less commonly, CIDP may present without proximal weakness (distal CIDP) or
with asymmetric weakness (multifocal CIDP). Response to treatment in typical and atypical CIDP may not differ significantly.
Patients with apparent distal symmetric, predominantly sensory neuropathy should be evaluated with electrodiagnostic tests (including
nerve conduction studies) as they may have the distal variant of CIDP, with predominantly sensory symptoms and with or without
distalweakness.
Multifocal CIDP with sensory and motor dysfunction must be carefully distinguished from vasculitic neuropathy, which requires
aggressive and prompt treatment.
Nerve biopsy and lumbar puncture are not mandatory but may help support the diagnosis of CIDP.
First-line treatments are intravenous immunoglobulin, plasma exchange and corticosteroids. If a patient fails to respond to one of the
three types of treatment, a second or third type of treatment, or a combination of them, should be attempted before using
alternative nonstandard therapies.
When there is failure to respond to standard treatments, reassessing the diagnosis and investigating possible secondary causes of
CIDP may be useful.
Relatively common secondary causes of CIDP include hepatitis C, plasma cell dyscrasia and lymphoma.
A syndrome that may resemble CIDP but that responds differently to therapy is anti-myelin-associated glycoprotein neuropathy,
which has a distinct pathophysiology (and a different therapeutic response) from CIDP. Testing for anti-myelin-associated
glycoprotein antibodies may be useful in patients with Waldenstrms macroglobulinemia and neuropathy, particularly when nerve
conduction studies show very prolonged distal motor latencies. Neuropathy may improve with rituximab, even when hematologic
disease is mild.
Onconeural antibodies, also known as the paraneoplastic antibody panel, are not commonly present in CIDP and may not be useful.

4 Dyck PJ, Lais AC, Ohta M, Bastron JA, One of the top cited research criteria
References Okazaki H, Groover RV. Chronic forCIDP, consolidating some of the
Papers of special note have been highlighted as:
inflammatory polyradiculoneuropathy. mostcurrently used
of interest
Mayo Clin. Proc. 50(11), 621637 (1975). electrodiagnosticparameters.
of considerable interest
1 Harris W, Newcomb WD. A case of The original complete manuscript that 6 Bromberg MB. Comparison of
relapsing interstitial hypertrophic defined what is now known as chronic electrodiagnostic criteria for primary
polyneuritis. Brain 52, 108116 (1929). inflammatory polyradiculoneuropathy demyelination in chronic polyneuropathy.
(CIDP), describing clinical, Muscle Nerve 14(10), 968976 (1991).
2 Austin JH. Recurrent polyneuropathies and
electrophysiological and pathological data. 7 Sander HW, Latov N. Research criteria
their corticosteroid treatment; with
five-year observations of a placebo- 5 Ad Hoc Subcommittee of the American for defining patients with CIDP.
controlled case treated with corticotrophin, Academy of Neurology AIDS Task Force. Neurology 60(8 Suppl. 3), S8S15
cortisone, and prednisone. Brain 81(2), Research criteria for the diagnosis of (2003).
157192 (1958). chronic inflammatory demyelinating 8 Nagamatsu M, Terao S, Misu K etal.
polyradiculoneuropathy (CIDP). Report Axonal and perikaryal involvement in
3 Thomas PK, Lascelles RG, Hallpike JF,
from an Ad Hoc Subcommittee of the chronic inflammatory demyelinating
Hewer RL. Recurrent and chronic
American Academy of Neurology AIDS polyneuropathy. J.Neurol. Neurosurg.
relapsing GuillainBarre polyneuritis.
Task Force. Neurology 41, 617618 (1991). Psychiatry 66(6), 727733 (1999).
Brain 92, 589606 (1969).

www.expert-reviews.com 377
Special Report De Sousa

9 Saperstein DS, Katz JS, Amato AA, 18 Rajabally YA, Simpson BS, Beri S, and Electrodiagnostic Medicine, and
BarohnRJ. Clinical spectrum of chronic BankartJ, Gosalakkal JA. Epidemiologic American Academy of Physical Medicine
acquired demyelinating polyneuropathies. variability of chronic inflammatory and Rehabilitation. Neurology 72(2),
Muscle Nerve 24, 311324 (2001). demyelinating polyneuropathy with 177184 (2009).
10 Nicolas G, Maisonobe T, Le Forestier N, different diagnostic criteria: study of a UK 29 Martina IS, van Koningsveld R, Schmitz
Lger JM, Bouche P. Proposed revised population. Muscle Nerve 39(4), 432438 PI, van der Mech FG, van Doorn PA.
electrophysiological criteria for chronic (2009). Measuring vibration threshold with a
inflammatory demyelinating polyradiculo- 19 Hattori N, Misu K, Koike H etal. Age of graduated tuning fork in normal aging and
neuropathy. Muscle Nerve 25, 2630 onset influences clinical features of chronic in patients with polyneuropathy. European
(2002). inflammatory demyelinating Inflammatory Neuropathy Cause and
11 Magda P, Latov N, Brannagan TH III, polyneuropathy. J.Neurol. Sci. 184(1), Treatment (INCAT) group. J.Neurol.
Weimer LH, Chin RL, Sander HW. 5763 (2001). Neurosurg. Psychiatry 65(5), 743747
Comparison of electrodiagnostic 20 Sharma KR, Saadia D, Facca AG, Bhatia R, (1998).
abnormalities and criteria in a cohort of Ayyar DR, Sheremata W. Chronic 30 Pestronk A, Florence J, Levine T etal.
patients with chronic inflammatory inflammatory demyelinating Sensory exam with a quantitative tuning
demyelinating polyneuropathy. Arch. polyradiculoneuropathy associated with fork: rapid, sensitive and predictive of
Neurol. 60, 17551759 (2003). multiple sclerosis. J.Clin. Neuromuscul. Dis. SNAP amplitude. Neurology 62(3),
12 Joint Task Force of the EFNS and the PNS. 9(4), 385396 (2008). 461464 (2004).
European Federation of Neurological 21 Dyck PJ, Dyck PJ. Atypical varieties of 31 Rotta FT, Sussman AT, Bradley WG,
Societies/Peripheral Nerve Society chronic inflammatory demyelinating RamAyyar D, Sharma KR, Shebert RT.
guideline on management of chronic neuropathies. Lancet 355(9212), The spectrum of chronic inflammatory
inflammatory demyelinating 12931294 (2000). demyelinating polyneuropathy. J.Neurol.
polyradiculoneuropathy. Report of a joint 22 Brannagan TH 3rd. Current treatments of Sci. 173(2), 129139 (2000).
task force of the European Federation of chronic immune-mediated demyelinating 32 Boukhris S, Magy L, Khalil M, Sindou P,
Neurological Societies and the Peripheral polyneuropathies. Muscle Nerve 39(5), Vallat JM. Pain as the presenting
Nerve Society. J.Peripher. Nerv. Syst. 10, 563578 (2009). symptom of chronic inflammatory
220228 (2005). demyelinating polyradiculoneuropathy
A well-organized and concise review of
A very organized and relatively recent set current treatments for CIDP. (CIDP). J.Neurol. Sci. 254(12), 3338
of guidelines for the diagnosis and (2007).
23 Katz JS, Saperstein DS, Gronseth G,
treatment of CIDP. A revised, updated 33 Chin RL, Latov N, Sander HW etal.
Amato AA, Barohn RJ. Distal acquired
manuscript is scheduled for publication Sensory CIDP presenting as cryptogenic
demyelinating symmetric neuropathy.
in2010. Neurology 54(3), 615620 (2000). sensory polyneuropathy. J.Peripher. Nerv.
13 Laughlin RS, Dyck PJ, Melton LJ 3rd, Syst. 9(3), 132137 (2004).
24 Lewis RA, Sumner AJ, Brown MJ,
Leibson C, Ransom J, Dyck PJ. AsburyAK. Multifocal demyelinating 34 Sinnreich M, Klein CJ, Daube JR,
Incidenceand prevalence of CIDP and neuropathy with persistent conduction Engelstad J, Spinner RJ, Dyck PJ.
theassociation of diabetes mellitus. block. Neurology 32(9), 958964 (1982). Chronic immune sensory
Neurology73(1), 3945 (2009). polyradiculopathy: a possibly treatable
The original description of sensory ataxia. Neurology 63(9),
14 McLeod JG, Pollard JD, Macaskill P,
multifocalCIDP. 16621669 (2004).
Mohamed A, Spring P, Khurana V.
Prevalence of chronic inflammatory 25 Saperstein DS, Amato AA, Wolfe GI etal. 35 Varela H, Rubin DI. Facial and
demyelinating polyneuropathy in New Multifocal acquired demyelinating sensory trigeminal neuropathies as the initial
South Wales, Australia. Ann. Neurol. and motor neuropathy: the LewisSumner manifestation of chronic inflammatory
46(6), 910913 (1999). syndrome. Muscle Nerve 22(5), 560966 demyelinating polyradiculopathy. J.Clin.
(1999). Neuromuscul. Dis. 10(4), 194198
15 Lunn MP, Manji H, Choudhary PP,
Hughes RA, Thomas PK. Chronic 26 Said G, Lacroix C. Primary and secondary (2009).
inflammatory demyelinating vasculitic neuropathy. J.Neurol. 252(6), 36 Nobile-Orazio E. Whats new in
polyradiculoneuropathy: a prevalence 633641 (2005). multifocal motor neuropathy in 2007
study in South East England. J.Neurol. 27 Hattori N, Misu K, Koike H etal. Age of 2008? J.Peripher. Nerv. Syst. 13(4),
Neurosurg. Psychiatry 66(5), 677680 onset influences clinical features of chronic 261263 (2008).
(1999). inflammatory demyelinating 37 Donaghy M, Mills KR, Boniface SJ etal.
16 Mygland A, Monstad P. Chronic polyneuropathy. J.Neurol. Sci. 184(1), Pure motor demyelinating neuropathy:
polyneuropathies in Vest-Agder, Norway. 5763 (2001). deterioration after steroid treatment and
Eur.J.Neurol. 8(2), 157165 (2001). 28 England JD, Gronseth GS, Franklin G improvement with intravenous
17 Iijima M, Koike H, Hattori N etal.; etal. American Academy of Neurology. immunoglobulin. J.Neurol. Neurosurg.
Refractory Peripheral Neuropathy Study Practice Parameter: evaluation of distal Psychiatry 57(7), 778783 (1994).
Group of Japan. Prevalence and incidence symmetric polyneuropathy: role of 38 Carpo M, Cappellari A, Mora G etal.
rates of chronic inflammatory autonomic testing, nerve biopsy, and skin Deterioration of multifocal motor
demyelinating polyneuropathy in the biopsy (an evidence-based review). Report neuropathy after plasma exchange.
Japanese population. J.Neurol. Neurosurg. of the American Academy of Neurology, Neurology 50(5), 14801482 (1998).
Psychiatry 79(9), 10401043 (2008). American Association of Neuromuscular

378 Expert Rev. Clin. Immunol. 6(3), (2010)


CIDP: diagnosis &management Special Report

39 Joint Task Force of the EFNS and the 49 Meriggioli MN, Rowin J. Chronic 60 Plant-Bordeneuve V, Ferreira A, Lalu T etal.
PNS. European Federation of Neurological inflammatory demyelinating Diagnostic pitfalls in sporadic transthyretin
Societies/Peripheral Nerve Society polyneuropathy after treatment with familial amyloid polyneuropathy (TTR-
Guideline on management of multifocal interferon-a. Muscle Nerve 23, 433435 FAP). Neurology 69(7), 693698 (2007).
motor neuropathy. Report of a joint task (2000). 61 Madia F, Frisullo G, Nociti V etal.
force of the European Federation of 50 Hirotani M, Nakano H, Ura S etal. pSTAT1, pSTAT3, and T-bet as markers of
Neurological Societies and the Peripheral Chronic inflammatory demyelinating disease activity in chronic inflammatory
Nerve Society. J.Peripher. Nerv. Syst. 11(1), polyneuropathy after treatment with demyelinating polyradiculoneuropathy.
18 (2006). interferon-a. Intern. Med. 48(5), 373375 J.Peripher. Nerv. Syst. 14(2), 107117
40 Mori K, Hattori N, Sugiura M etal. (2009). (2009).
Chronic inflammatory demyelinating 51 Corcia P, Barbereau D, Guennoc AM, 62 Comi C, Osio M, Ferretti M etal.
polyneuropathy presenting with features of deToffol B, Bacq Y. Improvement of a Defective Fas-mediated T-cell apoptosis
GBS. Neurology 58(6), 979982 (2002). CIDP associated with hepatitis C virus predicts acute onset CIDP. J.Peripher.
41 Hantson P, Kevers L, Fabien N, infection using antiviral therapy. Neurology Nerv. Syst. 14(2), 101106 (2009).
VanDenBergh P. Acute-onset chronic 63(1), 179180 (2004). 63 Naganuma M, Doi S, Shima K,
inflammatory demyelinating 52 No authors listed. Summary of revisions for Matsumoto A, Tashiro K. [Chronic
polyneuropathy with cranial nerve the 2010 Clinical Practice inflammatory demyelinating
involvement, dysautonomia, respiratory Recommendations. Diabetes Care polyradiculoneuropathy associated with
failure, and autoantibodies. Muscle Nerve 33(Suppl. 1), S3 (2010). multifocal nerve hypertrophy report of a
41(3), 423426 (2010). case with MRI study]. Rinsho Shinkeigaku
53 Joint Task Force of the EFNS and the
42 Ruts L, van Koningsveld R, van Doorn PA. PNS. European Federation of Neurological 31(11), 11861191 (1991).
Distinguishing acute-onset CIDP from Societies/Peripheral Nerve Society 64 Taniguchi N, Itoh K, Wang Y etal.
GuillainBarr syndrome with treatment Guideline on management of Sonographic detection of diffuse
related fluctuations. Neurology 65(1), paraproteinemic demyelinating peripheral nerve hypertrophy in chronic
138140 (2005). neuropathies. Report of a joint task force inflammatory demyelinating
This manuscript helps to of the European Federation of polyradiculoneuropathy. J.Clin.
distinguishacute-onset CIDP Neurological Societies and the Peripheral Ultrasound 28(9), 488491 (2000).
fromGuillainBarr syndrome. Nerve Society. J.Peripher. Nerv. Syst. 65 Zaidman CM, Al-Lozi M, Pestronk A.
11(1), 919 (2006). Peripheral nerve size in normals and patients
43 Rajabally Y, Vital A, Ferrer X etal.
Chronicinflammatory demyelinating 54 Mollee P. Current trends in the diagnosis, with polyneuropathy: an ultrasound study.
polyneuropathy caused by HIV infection in therapy and monitoring of the monoclonal Muscle Nerve 40(6), 960966 (2009).
a patient with asymptomatic CMT 1A. gammopathies. Clin. Biochem. Rev. 30(3), 66 Goldstein JM, Parks BJ, Mayer PL,
J.Peripher. Nerv. Syst. 5(3), 158162 (2000). 93103 (2009). KimJH, Sze G, Miller RG. Nerve root
44 Ryan MM, Jones HR Jr. CMTX mimicking 55 Sanchorawala V. Light-chain (AL) hypertrophy as the cause of lumbar
childhood chronic inflammatory amyloidosis: diagnosis and treatment. Clin. stenosis in chronic inflammatory
demyelinating neuropathy with tremor. J. Am. Soc. Nephrol. 1(6), 13311341 demyelinating polyradiculoneuropathy.
Muscle Nerve 31(4), 528530 (2005). (2006). Muscle Nerve 19(7), 892896 (1996).
45 Chi A, Plano F, Calvo A, Leone M, 56 Briani C, Scarlato M, Pavan A, 67 Echaniz-Laguna A, Philippi N. Teaching
Mutani R, Cocito D; Piemonte and Valle Marcolongo R, Rondinone R, Adami F. neuroimages: chronic inflammatory
DAosta Registry for CIDP (PARCIDP). Polyneuropathy, organomegaly, demyelinating polyradiculoneuropathy
Comorbidity between CIDP and diabetes endocrinopathy, M protein, skin changes: causing spinal cord compression.
mellitus: only a matter of chance? Eur. J. not always a POEMS syndrome. Neurology 72(24), e121 (2009).
Neurol. 16(6), 752754 (2009). Haematologica 91(1), 32 (2006). 68 Dalakas MC, Engel WK. Polyneuropathy
46 Sharma KR, Cross J, Farronay O, 57 Gondim FA, De Sousa EA, Latov N, with monoclonal gammopathy: studies of 11
AyyarDR, Shebert RT, Bradley WG. Sander HW, Chin RL, Brannagan TH. patients. Ann. Neurol. 10(1), 4552 (1981).
Demyelinating neuropathy in diabetes Anti-MAG/SGPG associated neuropathy 69 Schiff RI, Rudd C. Alterations in the
mellitus. Arch. Neurol. 59(5), 758765 does not commonly cause distal nerve half-life and clearance of IgG during
(2002). temporal dispersion. J.Neurol. Neurosurg. therapy with intravenous g-globulin in 16
Psychiatry 78(8), 902904 (2007). patients with severe primary humoral
47 Ayyar DR, Sharma KR. Chronic
inflammatory demyelinating 58 Dalakas MC, Rakocevic G, Salajegheh M immunodeficiency. J.Clin. Immunol. 6(3),
polyradiculoneuropathy in diabetes etal. Placebo-controlled trial of rituximab 256264 (1986).
mellitus. Curr. Diab. Rep. 4(6), in IgM anti-myelin-associated 70 Mehndiratta MM, Hughes RA, Agarwal P.
409412 (2004). glycoprotein antibody demyelinating Plasma exchange for chronic inflammatory
neuropathy. Ann. Neurol. 65(3), 286293 demyelinating polyradiculoneuropathy.
48 Marzo E, Tintor M, Fabregues O,
(2009). Cochrane Database Syst Rev. 3, CD003906
Montalban X, Codina A. Chronic
inflammatory demyelinating 59 Farbu E, Rekand T, Tysnes OB, Aarli JA, (2004).
polyneuropathy during treatment with Gilhus NE, Vedeler CA. GM1 antibodies 71 Hughes RA, Donofrio P, Bril V etal.
interferon-a. J.Neurol. Neurosurg. in post-polio syndrome and previous (ICEStudy Group). Intravenous immune
Psychiatry 65(4), 604 (1998). paralytic polio. J.Neuroimmunol. globulin (10% caprylate-chromatography
139(12), 141144 (2003).

www.expert-reviews.com 379
Special Report De Sousa

purified) for the treatment of chronic 73 Toothaker TB, Brannagan TH 3rd. 76 Barohn RJ, Kissel JT, Warmolts JR,
inflammatory demyelinating Chronic inflammatory demyelinating Mendell JR. Chronic inflammatory
polyradiculoneuropathy (ICE study): a polyneuropathies: current treatment demyelinating polyradiculoneuropathy.
randomised placebo-controlled trial. Lancet strategies. Curr. Neurol. Neurosci. Rep. 7(1), Clinical characteristics, course, and
Neurol. 7(2), 136144 (2008). Erratum in: 6370 (2007). recommendations for diagnostic criteria.
Lancet Neurol. 7(9), 771 (2008). 74 Hahn AF, Bolton CF, Pillay N etal. Arch. Neurol. 46(8), 878884 (1989).
72 Gorson KC, Allam G, Ropper AH. Chronic Plasma-exchange therapy in chronic 77 Kuwabara S, Misawa S, Mori M,
inflammatory demyelinating inflammatory demyelinating TamuraN, Kubota M, Hattori T. Long
polyneuropathy: clinical features and polyneuropathy. A double-blind, sham- term prognosis of chronic inflammatory
response to treatment in 67 consecutive controlled, cross-over study. Brain demyelinating polyneuropathy: a five year
patients with and without a monoclonal 119(Pt4), 10551066 (1996). follow up of 38 cases. J.Neurol. Neurosurg.
gammopathy. Neurology 48(2), 321328 75 Hahn AF, Bolton CF, Zochodne D, Psychiatry 77(1), 6670 (2006).
(1997). FeasbyTE. Intravenous immunoglobulin
The authors showed that when patients treatment in chronic inflammatory
fail the first treatment modality, be it demyelinating polyneuropathy. A double-
corticosteroids, intravenous blind, placebo-controlled, cross-over study.
immunoglobulin or plasma exchange, a Brain 119(Pt4), 10671077 (1996).
second or third type of therapy should
beattempted.

380 Expert Rev. Clin. Immunol. 6(3), (2010)


Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

You might also like