ArticlePDF Available

Abstract and Figures

Antibodies against myelin-associated glycoprotein (MAG) almost invariably appear in the context of an IgM monoclonal gammopathy associated neuropathy. Very few cases of anti-MAG neuropathy lacking IgM-monoclonal gammopathy have been reported. We investigated the presence of anti-MAG antibodies in 69 patients fulfilling diagnostic criteria for CIDP. Anti-MAG antibodies were tested by ELISA and confirmed by immunohistochemistry. We identified four (5.8%) anti-MAG positive patients without detectable IgM-monoclonal gammopathy. In two of them, IgM-monoclonal gammopathy was detected at 3 and 4-year follow-up coinciding with an increase in anti-MAG antibodies titers. In conclusion, anti-MAG antibody testing should be considered in chronic demyelinating neuropathies, even if IgM-monoclonal gammopathy is not detectable.
This content is subject to copyright. Terms and conditions apply.
1
SCIENTIFIC REPORTS | (2019) 9:6155 | https://doi.org/10.1038/s41598-019-42545-8
www.nature.com/scientificreports
Clinical and laboratory features
of anti-MAG neuropathy without
monoclonal gammopathy
Elba Pascual-Goñi
1, Lorena Martín-Aguilar1, Cinta Lleixà1, Laura Martínez-Martínez4,
Manuel J. Simón-Talero3, Jordi Díaz-Manera1,2, Elena Cortés-Vicente1,2, Ricard Rojas-García1,2,
Esther Moga4, Cándido Juárez4, Isabel Illa1,2 & Luis Querol
1,2
Antibodies against myelin-associated glycoprotein (MAG) almost invariably appear in the context
of an IgM monoclonal gammopathy associated neuropathy. Very few cases of anti-MAG neuropathy
lacking IgM-monoclonal gammopathy have been reported. We investigated the presence of anti-MAG
antibodies in 69 patients fullling diagnostic criteria for CIDP. Anti-MAG antibodies were tested by
ELISA and conrmed by immunohistochemistry. We identied four (5.8%) anti-MAG positive patients
without detectable IgM-monoclonal gammopathy. In two of them, IgM-monoclonal gammopathy
was detected at 3 and 4-year follow-up coinciding with an increase in anti-MAG antibodies titers. In
conclusion, anti-MAG antibody testing should be considered in chronic demyelinating neuropathies,
even if IgM-monoclonal gammopathy is not detectable.
Polyneuropathy associated with IgM monoclonal gammopathy of uncertain signicance (MGUSP) is a rare
form of chronic immune-mediated neuropathy. More than 50% of these patients harbor antibodies against
myelin-associated glycoprotein (MAG)1,2. Patients with anti-MAG+ MGUSP present with a predominantly sen-
sory neuropathy with ataxia and tremor with poor response to immunotherapy3.
Anti-MAG antibodies were described to be invariably associated with IgM monoclonal gammopathy4, and
clinical practice guidelines recommend to test them in patients with detectable IgM monoclonal gammopathy5.
Anecdotal cases of neuropathy with anti-MAG antibodies lacking monoclonal gammopathy were reported68.
A recent Japanese study8 reported a prevalence of 5.6% of anti-MAG positive patients in a cohort of 36 patients
with chronic demyelinating polyneuropathy with no monoclonal gammopathy. Antibodies in these patients were
tested by enzyme-linked immunosorbent assay (ELISA) and conrmed by Western blot analysis.
Here we investigate the presence of anti-MAG antibodies in patients fullling diagnostic criteria for chronic
inammatory demyelinating polyradiculoneuropathy (CIDP) without IgM monoclonal gammopathy. Also, we
describe the clinical, electrophysiological and laboratory ndings of four patients with anti-MAG associated neu-
ropathy without any detectable monoclonal gammopathy at the time of diagnosis.
Results
Patients. We detected 69 patients (61% males, mean age 58 years) fulfilling CIDP diagnostic criteria.
Flowchart of the study population is represented in Fig.1A. Briey, nine patients with antibodies toward NF155
(n = 4; 5.8%), NF140/186 (n = 2; 2.9%), CNTN1 (n = 2, 2.9%) or CNTN1/CASPR1 (n = 1; 1.4%), all of them neg-
ative for anti-MAG antibodies, were excluded from the seronegative cohort. irteen patients had monoclonal
gammopathy (IgA n = 1; IgG n = 9; IgM n = 2; IgA + IgG n = 1) at diagnosis. e two CIDP patients with IgM
monoclonal gammopathy were anti-MAG negative. Finally, we tested anti-MAG antibodies by ELISA in 58 CIDP
seronegative patients. Anti-MAG antibodies were detected in four patients (6.9% of the seronegative patients;
5.8% of the whole CIDP cohort) without IgM monoclonal gammopathy.
1Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat
Autònoma de Barcelona, Barcelona, Spain. 2Centro para la Investigación Biomédica en Red en Enfermedades Raras
(CIBERER), Madrid, Spain. 3Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma
de Barcelona, Barcelona, Spain. 4Department of Immunology, Hospital de la Santa Creu i Sant Pau, Universitat
Autònoma de Barcelona, Barcelona, Spain. Correspondence and requests for materials should be addressed to L.Q.
(email: lquerol@santpau.cat)
Received: 31 January 2019
Accepted: 2 April 2019
Published: xx xx xxxx
OPEN
There are amendments to this paper
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
SCIENTIFIC REPORTS | (2019) 9:6155 | https://doi.org/10.1038/s41598-019-42545-8
www.nature.com/scientificreports
www.nature.com/scientificreports/
Clinical and neurophysiological features. Clinical and epidemiological features of all four patients are
summarized in Table1. All of them were males, with ages ranging from 58 to 70 years. Patients 1 and 2 presented
with progressive distal sensory disturbances, while patient 4 presented with gait imbalance due to sensory ataxia.
Patient 3 was diagnosed of essential tremor and had an incipient neuropathy with impaired vibration sensation in
the lower limbs. Physical examination revealed mild to moderate sensory ataxia and mild to severe action tremor
in all patients. During follow-up, patients 1, 2 and 4 developed distal motor involvement. Nerve conduction stud-
ies (Table1) demonstrated prolonged distal motor latencies in patients 1 and 4 and mild to moderate reduction
of motor or sensory nerve conduction velocities in all four patients. Also, F-waves showed prolonged latencies in
patients 1, 2 and 3; and were absent in patient 4. Temporal dispersion was observed in patient 1, and compound
muscle action potentials or sensory nerve action potentials were reduced in all four patients. An additional le
shows nerve conduction studies in more detail (Supplementary Table1).
All patients were treated with intravenous immunoglobulin (IVIg) (2 g/Kg), and good response was observed
in patient 1 and 4, while partial response was observed in patients 2 and 3. Patient 2 was treated with azathio-
prine without response. Upon IgM MGUS detection, rituximab (375 mg/m2, once weekly for 4 weeks followed
by 1 additional dose 1 month later) was started and we observed disease stabilization. IVIg were suspended in
patient 4, due to toxicodermia and neither prednisone (1 mg/Kg/d), nor cyclosporine (125 mg/12 h) showed any
signicant benet.
Antibody assays. Anti-MAG antibodies tested positive at diagnosis in four patients by ELISA.
Immunoxation did not detect monoclonal gammopathy at diagnosis in any of these patients, and total IgM
levels were only mildly elevated in patient 3 (301 mg/dL, upper limit 230 mg/dL). Anti-MAG antibody titers and
presence of IgM monoclonal gammopathy by immunoxation was tested periodically in these patients depending
on their visit schedules. Follow-up anti-MAG antibody titers are shown in Fig.1B. Antibodies to sulfatides and
gangliosides were negative in all four patients.
In patients 1 and 2 we detected an IgM monoclonal gammopathy aer 3 and 4 years of follow-up respectively
(Fig.1B), while in patients 3 and 4 no monoclonal gammopathy has been detected yet (follow-up of 5 and 2
years respectively). In patient 1, the detection of monoclonal gammopathy coincided with a signicant increase in
anti-MAG antibody titers. At that time, hematological malignancy screening tests performed in patients 1 and 2
were negative. Both had a serum IgM-kappa monoclonal protein of less than 1 g/L, a negative Bence-Jones protein
Figure 1. Flowchart of the study population (A). Serial anti-MAG antibody titers during follow-up (B). e
asterisks highlight the detection of IgM MGUS in patients 1 and patient 2. e arrow indicates rituximab
administration. Immunohistochemistry studies with serum from patients 1–4 showing IgM binding on the
myelin sheaths. Immunouorescence intensity increased in patients 1 and 2 aer MGUS detection (C). Staining
pattern of patients anti-MAG- sulfatides+ MGUSP used as control are shown. Titers of anti-MAG and anti-
sulfatides antibodies are represented. (Anti-IgM, 20x and 40x original magnication). BTU Bühlmann test
units; IgM immunoglobulin M; MAG myelin-associated glycoprotein; MGUS monoclonal gammopathy of
uncertain signicance.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
SCIENTIFIC REPORTS | (2019) 9:6155 | https://doi.org/10.1038/s41598-019-42545-8
www.nature.com/scientificreports
www.nature.com/scientificreports/
urine test, and a radiographic X-ray skeletal survey without bone lesions. Accordingly, both patients were diag-
nosed of IgM MGUS and underwent hematological follow-up. Neither of them developed malignancy to date.
Immunohistochemistry. At diagnosis, serum from all four patients showed a typical anti-MAG reactivity
pattern in the immunohistochemistry assays. Immunostaining reactivity was indistinguishable from patients
with monoclonal gammopathy associated anti-MAG neuropathy and dierent from patients with anti-sulfatide
antibody- associated neuropathy (Fig.1C). e intensity of the myelin staining increased signicantly aer IgM
monoclonal gammopathy detection in patients 1 and 2.
Discussion
In this study, we identied anti-MAG antibodies in four patients fullling CIDP diagnostic criteria and no evi-
dence of monoclonal gammopathy. Only one patient had slightly increased total IgM levels at diagnosis, and
in two patients we detected an IgM MGUS aer 3 and 4 years of follow-up. All patients presented with clin-
ical, electrophysiological and serological features indistinguishable from those described in patients with
anti-MAG + MGUSP3.
Since their description, anti-MAG antibodies have always been associated to IgM monoclonal gammopathy4.
Anti-MAG have been shown to be specic for the diagnosis of MGUSP, while they are negative in healthy con-
trols2. Indeed, clinical guidelines5,9 only recommend to test anti-MAG antibodies in patients with IgM monoclo-
nal gammopathy. Although the association of IgM monoclonal gammopathy and anti-MAG antibodies is very
strong, these recommendations likely generate a selection bias. Our observations suggest that there is a subset
of patients with anti-MAG + polyneuropathy without any detectable monoclonal gammopathy that may remain
undiagnosed. A few other cases of anti-MAG neuropathy in the absence of monoclonal gammopathy have been
described68, supporting our observations.
In two of our patients an IgM monoclonal gammopathy was detected by serum immunoxation years aer
diagnosis, and in patient 1 it clearly coincided with an increase in anti-MAG titers. Longer follow-ups may lead
to detectable gammopathy in the other two patients but this remains to be conrmed. is phenomenon may
Patient 1 Patient 2 Patient 3 Patient 4
Age at onset, yr; Sex 58; M 70; M 70; M 68; M
Past medical history No Hypertension, diabetes Osteoarthritis,
Essential tremor Arthritis (methotrexate)
Clinical manifestations
Initial symptoms Distal sensory
disturbance
upper > lower limbs
Distal sensory disturbance
upper > lower limbs Postural tremor Gait ataxia
Limb weakness Distal > proximal mild Distal mild No Distal > proximal moderate
distal atrophy
Gait ataxia Mild Moderate Mild Moderate
Intention tremor Mild Moderate, upper limbs Severe, head and
upper limbs Moderate, upper limbs
Electrophysiological ndings
Prolonged motor distal
latencies + − +
Reduction of NCV + + + +
Prolonged F-wave latencies + + + +
Conduction block NA
Temporal dispersion + − +
Reduced CMAPs + + +
Reduced SNAPs + + + +
Laboratory ndings
Cerebrospinal uid ndings
(protein; cell count) 1,1 g/L; 2cells/mm31,4 g/L; 2cells/mm3NA 0,44 g/L; 0cells/mm3
IgM levels * (presentation) 163 mg/dL 174 mg/dL 301 mg/dL 63 mg/dL
Anti-sulfatides and
gangliosides Negative Negative Negative Negative
Anti-MAG Abs titers
(presentation) 2500 9000 2050 1300
Monoclonal protein, levels
(follow-up) IgM-κ, < 1 g/L IgM-κ, < 1 g/L No No
Malignancy screening Negative Negative NA NA
Treat ment and response IVIg: good IVIg: partial azathioprine:
no rituximab: good IVIg: partial IVIg: good steroids: no
cyclosporine: no
Table 1. Summary of clinical and laboratory ndings of patients with anti-MAG neuropathy without
monoclonal gammopathy. *IgM normal values: 40–230 mg/dL. CMAPs: compound muscle action potential;
IVIg: intravenous immunoglobulin; κ: kappa light chain; M: male; NA: not available; NCV: nerve conduction
velocities; SNAPs: sensory nerve action potential; yr: years.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
SCIENTIFIC REPORTS | (2019) 9:6155 | https://doi.org/10.1038/s41598-019-42545-8
www.nature.com/scientificreports
www.nature.com/scientificreports/
imply that, either early IgM gammopathy is not detectable with current immunoxation techniques or that an
early antigen-driven autoimmune process is subsequently followed by a clonal expansion and appearance of the
monoclonal gammopathy. Whatever the case, these two patients suggest that anti-MAG antibody polyneuropathy
displays a spectrum of disease that includes patients that test negative for the presence of gammopathy. Either
early testing of anti-MAG or repeated testing of monoclonal gammopathy by immunoxation have to be con-
sidered then, especially in patients with clinical and electrophysiological features resembling typical anti-MAG+
MGUSP.
e prevalence of anti-MAG+ patients in our CIDP cohort (5.8%) was similar to that recently reported by
a Japanese group (5.7%)8. It is also comparable to the amount of anti-NF155+ patients in our study population
(4/69) and the prevalence of anti-NF155+ patients reported in other CIDP cohorts10. ese ndings support the
concept that CIDP is a heterogeneous disease in terms of immunopathology, clinical presentation and treatment
response. erefore, autoantibody proling, including detection of anti-MAG antibodies, is useful to guide diag-
nosis, prognosis and treatment selection in patients with chronic demyelinating neuropathy.
e treatment strategy in anti-MAG associated neuropathies is limited due to the low response rate to cur-
rent therapies. Treatment with IVIg, plasma exchange, prednisone or rituximab have shown benets in some
patients3,11. Two of our patients were initially diagnosed of seronegative CIDP and unsuccessfully treated with
immunosuppressant drugs such as azathioprine and cyclosporine that are not considered eective in anti-MAG+
MGUSP. us, within the standard therapies used in CIDP, anti-MAG antibodies helped us choose those thera-
pies that could yield better results (e.g IVIg). It would be interesting to assess in larger cohorts if early treatment
of these patients with B-cell depleting therapies, such as rituximab, would be more ecacious than if patients are
treated aer the development of the monoclonal gammopathy11,12. Moreover, due to the association of anti-MAG
antibodies to the presence of MGUS, all four patients underwent hematological follow-up to study the appearance
of monoclonal gammopathy.
In conclusion, we report four patients with anti-MAG neuropathy in the absence of IgM-monoclonal gam-
mopathy. Given these observations, we suggest to test anti-MAG antibodies in patients with chronic demyeli-
nating neuropathy, regardless of the detection of IgM monoclonal gammopathy, especially in those with distal,
sensory-ataxic involvement.
Methods
Patients, informed consent and protocol approvals. Patients prospectively observed during routine
neuromuscular practice between 2007–2017 fullling EFNS/PNS diagnostic criteria for CIDP were included.
We tested the presence of anti-MAG antibodies in serum. Patients with antibodies towards neurofascin-155
(NF155), nodal neurofascin-140 and 186 (NF140/186), contactin-1 (CNTN1), contactin-1/caspr-1 complex
(CNTN1/CASPR1) were excluded from the seronegative cohort. is study was conducted according to a pro-
tocol approved by the Institutional Ethics’ Committee of the Hospital de la Santa Creu i Sant Pau. All experi-
ments were performed in accordance with the relevant guidelines and regulations. Written informed consent
were obtained from all subjects.
Clinical and neurophysiological features. In anti-MAG+ patients we collected the age at onset, sex,
past medical history and clinical manifestations including initial symptoms and the presence of limb weakness
(proximal/distal), gait ataxia or intention tremor. We analyzed neurophysiological ndings including motor distal
latencies, nerve conduction velocities, F-wave latencies, and the presence of conduction blocks, temporal disper-
sion, reduced CMAPs or reduced SNAPs. We also collected therapies and response to them.
Antibody assays. e presence of monoclonal gammopathy (IgA, IgG or IgM) was evaluated by serum
protein electrophoresis and serum immunoxation electrophoresis studies (Sebia, France) at diagnosis and
follow-up. Antibodies against NF155, NF140/186, CNTN1 and CNTN1/CASPR1 were investigated by immu-
nocytochemistry as previously described13. Anti-MAG antibodies were tested by ELISA (Bühlmann laboratories
AG, Schönenbuch, Switzerland). We used a cut-o value of 1000 Bühlmann Titer Units (BTU), according to the
manufacturer’s instructions. In anti-MAG+ patients, antibodies to sulfatides and gangliosides were also inves-
tigated by ELISA as previously described14. Further, total levels of IgM in serum were investigated (Immage 800
Nephelometer Beckman Coulter).
Immunohistochemistry. Monkey peripheral nerve tissue slides (Inova Diagnostics, Inc., San Diego, CA)
were blocked with 5% normal goat serum in PBS, incubated with patients’ sera at 1:10 for 1 hour at room temper-
ature, washed and incubated with Alexa Fluor 594 goat antihuman IgM secondary antibody at 1:1000 for 1 hour.
Slides were mounted with Fluoromount medium (Sigma-Aldrich, St. Louis, MO). Immunostaining patterns were
analyzed and compared with controls. Sera from patients with anti-MAG+ MGUSP and anti-MAG- sulfati-
des + MGUSP were used as disease controls.
Data Availability
All data generated or analyzed during this study are included in this published article (and its Supplementary
Information Files).
References
1. Nobile-Orazio, E. et al. Frequency and clinical correlates of anti-neural IgM antibodies in neuropathy associated with IgM
monoclonal gammopathy. Ann Neurol 36, 416–424 (1994).
2. uijf, M. L. et al. Detection of anti-MAG antibodies in polyneuropathy associated with IgM monoclonal gammopathy. Neurology
73, 688–695 (2009).
3. Dalaas, M. C. Advances in the diagnosis, immunopathogenesis and therapies of IgM-anti-MAG antibody-mediated neuropat hies.
er. Adv. Neurol. Disord. 11, 175628561774664 (2018).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
SCIENTIFIC REPORTS | (2019) 9:6155 | https://doi.org/10.1038/s41598-019-42545-8
www.nature.com/scientificreports
www.nature.com/scientificreports/
4. Ellie, E. et al. Neuropathy associated with ‘benign’ anti-myelin-associated glycoprotein IgM gammopathy: clinical, immunological,
neurophysiological pathological ndings and resp onse to treatment in 33 cases. J Neurol 243, 34–43 (1996).
5. Joint Tas Force of the, E. & the, P. N. S. European Federation of Neurological Societies/Peripheral Nerve Society Guideline on
management of paraproteinemic demyelinating neuropathies. eport of a Joint Tas Force of the European Federation of
Neurological Societies and the Peripheral Nerve Societ. J Peripher Ner v Syst 15, 185–195 (2010).
6. Nobile-Orazio, E. et al. Neuropathy and anti-MAG antibodies without detectable serum M-protein. Neurology 34, 218–221 (1984).
7. Gabriel, J. M. et al. Confocal microscopic localization of anti-myelin-associated glycoprotein autoantibodies in a patient with
peripheral neuropathy initially lacing a detectable IgM gammopathy. Acta Neuropathol 95, 540–546 (1998).
8. Saamoto, Y., Shimizu, T., Tobisawa, S. & Isozai, E. Chronic demyelinating neuropathy with anti-myelin-associated glycoprotein
antibody without any detectable M-protein. Neurol Sci 38, 2165–2169 (2017).
9. Van den Bergh, P. Y. et al. European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of
chronic inflammatory demyelinating polyradiculoneuropathy: report of a joint tas force of the European Federation of
Neurological Societies and the Peripher. Eur J Neurol 17, 356–363 (2010).
10. Devaux, J. J. et al. Neurofascin-155 IgG4 in chronic inammatory demyelinating polyneuropathy. Neurology 86, 800–807 (2016).
11. Dalaas, M. C. et al. Placebo-controlled trial of rituximab in IgM anti-myelin-associated glycoprotein antibody demyelinating
neuropathy. Ann Neurol 65, 286–293 (2009).
12. Leger, J.-M. et al. Placebo-controlled trial of rituximab in IgM anti-myelin-associated glycoprotein neuropathy. Neurology 80,
2217–2225 (2013).
13. Querol, L. et al. Antibodies against peripheral nerve antigens in chronic inammatory demyelinating polyradiculoneuropathy. Sci
Rep 7, 14411 (2017).
14. Willison, H. J. et al. Inter-laborator y validation of an ELISA for the determination of serum anti-ganglioside antibodies. Eur J Neurol
6, 71–77 (1999).
Acknowledgements
is project was supported by Fondo de Investigaciones Sanitarias (FIS), Instituto de Salud Carlos III, Spain and
FEDER under grant FIS16/00627, and personal grant SLT006/17/00131 of the Pla estratègic de recerca i innovació
en salut (PERIS), Departament de Salut, Generalitat de Catalunya, IP Luis Querol.
Author Contributions
E.P.G. and L.Q. were involved in study conceptualization, data collection, draing, analysis, and revising the
manuscript for intellectual content. L.M.A., C.L., L.M.M., M.S.T., J.D.M., E.C.V., R.R.G., E.M., C.J., I.I. were
involved in data collection and revising the manuscript for intellectual content.
Additional Information
Supplementary information accompanies this paper at https://doi.org/10.1038/s41598-019-42545-8.
Competing Interests: EP-G, LM-A, CL, LM-M, MJS-T, JD-M, EC-V, RR-G, EM and CJ declare no competing
interests.LQ has provided expert testimony for Grifols, Genzyme and CSL Behring and received research funds
from Novartis Spain and Grifols (Spin Award). II provided expert testimony and received speaking fees and
travel grants from Pzer.
Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Cre-
ative Commons license, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons license and your intended use is not per-
mitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the
copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
© e Author(s) 2019
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Two earlier studies, encompassing a combined total of 113 patients, identified six patients who met the diagnostic criteria for CIDP and tested positive for anti-MAG antibodies without IgM paraproteinemia. 64,65 The clinical presentation and progression in these patients were similar to that of anti-MAG positive patients who had an IgM paraproteinemia. Three out of these six patients tested positive for an M-protein later during follow-up. ...
Article
Full-text available
Diagnosing Chronic Inflammatory Demyelinating Polyneuropathy (CIDP) poses numerous challenges. The heterogeneous presentations of CIDP variants, its mimics, and the complexity of interpreting electrodiagnostic criteria are just a few of the many reasons for misdiagnoses. Early recognition and treatment are important to reduce the risk of irreversible axonal damage, which may lead to permanent disability. The diagnosis of CIDP is based on a combination of clinical symptoms, nerve conduction study findings that indicate demyelination, and other supportive criteria. In 2021, the European Academy of Neurology (EAN) and the Peripheral Nerve Society (PNS) published a revision on the most widely adopted guideline on the diagnosis and treatment of CIDP. This updated guideline now includes clinical and electrodiagnostic criteria for CIDP variants (previously termed atypical CIDP), updated supportive criteria, and sensory criteria as an integral part of the electrodiagnostic criteria. Due to its many rules and exceptions, this guideline is complex and misinterpretation of nerve conduction study findings remain common. CIDP is treatable with intravenous immunoglobulins, corticosteroids, and plasma exchange. The choice of therapy should be tailored to the individual patient’s situation, taking into account the severity of symptoms, potential side effects, patient autonomy, and past treatments. Treatment responses should be evaluated as objectively as possible using disability and impairment scales. Applying these outcome measures consistently in clinical practice aids in recognizing the effectiveness (or lack thereof) of a treatment and facilitates timely consideration of alternative diagnoses or treatments. This review provides an overview of the current perspectives on the diagnostic process and first-line treatments for managing the disease.
... Since anti-MAG antibodies are found in more than 70% of patients with typical M component-related demyelinating polyneuropathy [4], in clinical practice, an anti-MAG antibody test is performed only in patients with detectable IgM monoclonal gammopathy. However, some cases of anti-MAG neuropathy can lack IgM monoclonal gammopathy [44][45][46][47] with the disclosure of the IgM monoclonal protein after years of follow-up [45]. Although these reports are anecdotical, it is suggested to test anti-MAG antibodies in patients with distal chronic sensorimotor demyelinating neuropathy, regardless of the detection of IgM monoclonal gammopathy. ...
Article
Full-text available
Chronic dysimmune neuropathies encompass a group of neuropathies that share immune-mediated pathomechanism. Chronic dysimmune antibody-related neuropathies include anti-MAG neuropathy, multifocal motor neuropathy, and neuropathies related to immune attack against paranodal antigens. Such neuropathies exhibit distinguishing pathomechanism, clinical and response to therapy features with respect to chronic inflammatory demyelinating polyradiculoneuropathy and its variants, which represent the most frequent form of chronic dysimmune neuropathy. This narrative review provides an overview of pathomechanism; clinical, electrophysiological, and biochemical features; and treatment response of the antibody-mediated neuropathies, aiming to establish when and why to look for antibodies in chronic dysimmune neuropathies.
Article
Full-text available
Serological tests are important to detect autoantibodies (autoAbs) in patients with autoimmune neuropathies (AN) and myasthenia gravis (MG) as they are biomarkers for diagnosis, stratification, treatment selection, and monitoring. However, tests to detect autoAbs frequently lack proper standardization and results differ across diagnostic laboratories. We compared results for tests routinely performed in Spanish diagnostic laboratories to detect AN and MG autoAbs. In the Spanish Society of Immunology Autoimmunity Group national workshop, serum samples from 13 patients with AN or MG were tested for anti-ganglioside, anti-myelin-associated glycoprotein (MAG), anti-nicotinic acetylcholine receptor (AChR), and anti-muscle-specific kinase (MuSK) autoAbs using reference methods and were distributed for analysis to 27 participating laboratories using their routine methods. Overserved were inter-laboratory variability and worryingly low sensitivity, especially for anti-ganglioside immunoglobulin G and anti-MAG autoAb detection. This pilot study reflects autoAbs detection state of the art in AN and MG testing in leading diagnostic laboratories in Spain, highlighting the need for standardization prior to clinical use.
Article
The nodes of Ranvier (NoR) are essential domains for nerve conduction and their disruption plays a key role in the pathophysiology of immune-mediated neuropathies. Our understanding of the specialised nodal regions and the immune mechanisms that affect them is growing and has led to the update of peripheral neuropathy classification to include the autoimmune nodopathies, defined by the site of the autoimmune attack. Autoantibodies directed against molecules of the nodal region (as neurofascin-140/186, neurofascin-155, contactin-1, contactin-associated protein 1, contactin-associated protein 2, gangliosides, LGI4 or myelin-associated glycoprotein), macrophage-induced paranodal demyelination, and phenotypic changes of the nodal domains of Schwann cells have been identified as key mechanisms in the pathogenesis of the autoimmune neuropathies. This review explores the current knowledge of the autoimmune vulnerability of the NoR, including the underlying mechanisms leading to dysfunction in the diverse autoimmune disorders. This article is protected by copyright. All rights reserved.
Article
Full-text available
Dysfunction of the immune system can result in damage to the peripheral nervous system. The immunological mechanisms, which include macrophage infiltration, inflammation and proliferation of Schwann cells, result in variable degree of demyelination and axonal degeneration. Etiology is diverse and, in some cases, may be precipitated by infection. Various animal models have helped elucidate the pathophysiological mechanisms in acute and chronic inflammatory polyradiculoneuropathies (Guillain-Barre Syndrome and chronic inflammatory de-myelinating polyradiculoneuropathy, respectively). Presence of specific anti-glycoconjugate antibodies indicates an underlying process of molecular mimicry and sometimes assists in the classification of these disorders which often merely supports the clinical diagnosis. Electro-physiological presence of conduction block is important in characterizing another sub-group of treatable motor neuropathies (multifocal motor neuropathy with conduction block), which is distinct from Lewis-Sumner syndrome (multifocal acquired demyelinating sensory and motor neuropathy) in its response to treatment modalities as well as electrophysiological features. Furthermore, paraneoplastic neuropathies are also immune-mediated and are the result of an immune reaction to tumor cells that express onconeural antigens and mimic molecules expressed on the surface of neurons. The detection of specific paraneoplastic antibodies often assists the clinician in the investigation of an underlying, sometimes specific, malignancy. This review aims to discuss the immunological and pathophysiological mechanisms that are thought to be crucial in the etiology of dysimmune neuropathies as well as their individual electrophysiological characteristics, laboratory features and existing treatment options. Here, we aim to present a balanced discussion from these diverse angles that may be helpful in categorizing disease and establishing prognosis.
Article
Autoimmune neuropathies are a heterogeneous group of rare and disabling diseases in which the immune system targets peripheral nervous system antigens and that respond to immune therapies. This review focuses on Guillain-Barré syndrome, chronic inflammatory demyelinating polyneuropathy, multifocal motor neuropathy, polyneuropathy associated with IgM monoclonal gammopathy, and autoimmune nodopathies. Autoantibodies targeting gangliosides, proteins in the node of Ranvier, and myelin-associated glycoprotein have been described in these disorders, defining subgroups of patients with similar clinical features and response to therapy. This topical review describes the role of these autoantibodies in the pathogenesis of autoimmune neuropathies and their clinical and therapeutic importance.
Chapter
Autoimmune neuromuscular disorders are very heterogeneous both in their clinical presentation and their pathogenesis. The discovery of specific autoantibodies targeting the nicotinic acetylcholine receptor in myasthenia gravis led the way in the description of a wide variety of autoantibodies that helped diagnosing, classifying, and treating autoimmune disorders of the neuromuscular junction, inflammatory neuropathies, and myositis. The description of antibodies against muscle-specific kinase in myasthenia, anti-ganglioside antibodies, or the new antibodies against nodal and paranodal structures in inflammatory neuropathies and the anti-synthetase or anti-HMGCoA antibodies in myositis were critical for the detection of distinct diseases and phenotypes within classical clinical syndromes. Searching for highly disease-specific autoantibodies should be a priority in autoimmune neuromuscular disorders to understand their pathogenesis and base diagnosis in biomarkers rather than clinical criteria.KeywordsMyastheniaAChRMusKGuillain-Barre syndromeCIDPContactin-1NF155Pan-neurofascinAnti-ganglioside antibodiesMyositisAnti-synthetaseAnti-HMGCoALambert-Eaton
Article
Introduction: Despite extensive research, multiple inter-related diagnostic and management challenges remain for chronic inflammatory demyelinating polyneuropathy (CIDP). Areas covered: A literature review was performed on diagnosis and treatment in CIDP. The clinical features and disease course were evaluated. Investigative techniques, including electrophysiology, cerebrospinal fluid examination, neuropathology, imaging and neuroimmunology, were considered in relation to technical aspects, sensitivity, specificity, availability and cost. Available evidenced-based treatments and those with possible efficacy despite lack of evidence, were considered, as well as current methods for evaluation of treatment effects. Expert opinion: CIDP remains a clinical diagnosis, supported first and foremost by electrophysiology. Other investigative techniques have limited impact. Most patients with CIDP respond to available first-line treatments and immunosuppression may be efficacious in those who do not. Consideration of the natural history and of the high reported remission rate, of under-recognised associated disabling features, of treatment administration modalities and assessment methods, require enhanced attention.
Article
The acquired chronic demyelinating neuropathies include a growing number of disease entities that have characteristic, often overlapping, clinical presentations, mediated by distinct immune mechanisms, and responding to different therapies. After the discovery in the early 1980s, that the myelin associated glycoprotein (MAG) is a target antigen in an autoimmune demyelinating neuropathy, assays to measure the presence anti-MAG antibodies were used as the basis to diagnose the anti-MAG neuropathy. The route was open for describing the clinical characteristics of this new entity as a chronic distal large fiber sensorimotor neuropathy, for studying its pathogenesis and devising specific treatment strategies. The initial use of chemotherapeutic agents was replaced by the introduction in the late 1990s of rituximab, a monoclonal antibody against CD20⁺ B-cells. Since then, other anti-B cells agents have been introduced. Recently a novel antigen-specific immunotherapy neutralizing the anti-MAG antibodies with a carbohydrate-based ligand mimicking the natural HNK-1 glycoepitope has been described.
Article
Full-text available
Polyneuropathy with immunoglobulin M (IgM) monoclonal gammopathy is the most common paraproteinemic neuropathy, comprising a clinicopathologically and immunologically distinct entity. The clinical spectrum spans from distal paresthesias and mild gait imbalance to more severe sensory ataxia, with falls and a varying degree of distal sensorimotor deficits. In approximately 75% of patients, the monoclonal IgM immunoreacts with myelin-associated glycoprotein (MAG) and sulfoglucuronyl glycosphingolipid (SGPG), or other peripheral nerve glycolipids that serve as antigens. These antibodies are considered pathogenic because IgM and complement are deposited on the myelin sheath, splitting the myelin lamellae, while adoptive transfer of patients’ IgM into susceptible host animals causes sensory ataxia and reproduces the human pathology. In spite of the apparently convincing pathogenicity of these antibodies, the response to immunotherapies remains suboptimal. Clorambuscil, cladibrine, cyclophospamide and intravenous immunoglobulin may help some patients but the benefits are minimal and transient. Open-label studies in >200 patients indicate that rituximab is helpful in 30–50% of these patients, even with long-term benefits, probably by suppressing IgM anti-MAG antibodies or inducing immunoregulatory T cells. Two controlled studies with rituximab did not however meet the primary endpoint, mostly because of the poor sensitivity of the scales used; they did however show statistical improvement in secondary endpoints and improved clinical functions in several patients. This review provides an overview of the clinical phenotypes and immunoreactivity of IgM to glycolipids or glycoproteins of peripheral nerve myelin, summarizes the progress on treatment with rituximab as a promising therapy, discusses the pitfalls of scales used, identifies possible biomarkers of response to therapy and highlights the promising new anti-B cell or target-specific immunotherapies.
Article
Full-text available
Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is a heterogeneous disease in which diverse autoantibodies have been described but systematic screening has never been performed. Detection of CIDP-specific antibodies may be clinically useful. We developed a screening protocol to uncover novel reactivities in CIDP. Sixty-five CIDP patients and 28 controls were included in our study. Three patients (4.6%) had antibodies against neurofascin 155, four (6.2%) against contactin-1 and one (1.5%) against the contactin-1/contactin-associated protein-1 complex. Eleven (18.6%) patients showed anti-ganglioside antibodies, and one (1.6%) antibodies against peripheral myelin protein 2. No antibodies against myelin protein zero, contactin-2/contactin-associated protein-2 complex, neuronal cell adhesion molecule, gliomedin or the voltage-gated sodium channel were detected. In IgG experiments, three patients (5.3%) showed a weak reactivity against motor neurons; 14 (24.6%) reacted against DRG neurons, four of them strongly (7.0%), and seven (12.3%) reacted against Schwann cells, three of them strongly (5.3%). In IgM experiments, six patients (10.7%) reacted against DRG neurons, while three (5.4%) reacted against Schwann cells. However, results were not statistically significant when compared to controls. Immunoprecipitation experiments identified CD9 and L1CAM as potential antigens, but reactivity could not be confirmed with cell-based assays. In summary, we describe a diverse autoantibody repertoire in CIDP patients, reinforcing the hypothesis of CIDP’s pathophysiological heterogeneity.
Article
Previous case reports and studies have shown that anti-myelin-associated glycoprotein (MAG) antibody can be detected in patients with polyneuropathy without any detectable M-protein. Nevertheless, the frequency of and related factors have not yet been adequately investigated. The objectives of this study are to examine the prevalence of anti-MAG antibody in patients with demyelinating neuropathy without M-protein and to determine their clinical characteristics. From January, 2004, to September, 2016, consecutive patients with chronic demyelinating neuropathy were recruited. Anti-MAG antibody presence was tested at the first evaluation. We determined the prevalence of anti-MAG antibody without M-protein among included patients and evaluated the clinical characteristics. A total of 44 patients were included in the present study (12 women; median age at first visit 60 years [interquartile range 47–67 years]; median duration between onset and first visit 9 months [3–26 months]). M-protein was found in eight patients (18%) at the first evaluation. Anti-MAG antibody was present in 2 of remaining 36 (5.6 [95% confidence interval 0–13.0] %) patients without M-protein. Patients with anti-MAG antibody exhibited slowly progressive and distal dominant neuropathy with elevated serum IgM levels and refractory to immunotherapy. There were no differences in clinical features between patients having anti-MAG antibody without M-protein, and those with M-protein. One patient with the anti-MAG antibody showed a delayed appearance of M-protein during a 4-year follow-up after diagnosis. The prevalence of the anti-MAG antibody in chronic demyelinating neuropathy without any detectable M-protein was 5.6%. Anti-MAG antibody may be detectable earlier than M-protein.
Article
Objective: We report the clinical and serologic features of Japanese patients with chronic inflammatory demyelinating polyneuropathy (CIDP) displaying anti-neurofascin-155 (NF155) immunoglobulin G4 (IgG4) antibodies. Methods: In sera from 533 patients with CIDP, anti-NF155 IgG4 antibodies were detected by ELISA. Binding of IgG antibodies to central and peripheral nerves was tested. Results: Anti-NF155 IgG4 antibodies were identified in 38 patients (7%) with CIDP, but not in disease controls or normal participants. These patients were younger at onset as compared to 100 anti-NF155-negative patients with CIDP. Twenty-eight patients (74%) presented with sensory ataxia, 16 (42%) showed tremor, 5 (13%) presented with cerebellar ataxia associated with nystagmus, 3 (8%) had demyelinating lesions in the CNS, and 20 of 25 (80%) had poor response to IV immunoglobulin. The clinical features of the antibody-positive patients were statistically more frequent as compared to negative patients with CIDP (n = 100). Anti-NF155 IgG antibodies targeted similarly central and peripheral paranodes. Conclusion: Anti-NF155 IgG4 antibodies were associated with a subgroup of patients with CIDP showing a younger age at onset, ataxia, tremor, CNS demyelination, and a poor response to IV immunoglobulin. The autoantibodies may serve as a biomarker to improve patients' diagnosis and guide treatments.
Article
Objective: To determine whether rituximab 375 mg/m(2) was efficacious in patients with immunoglobulin M (IgM) anti-myelin-associated glycoprotein antibody demyelinating neuropathy (IgM anti-MAG demyelinating neuropathy). Methods: Fifty-four patients with IgM anti-MAG demyelinating neuropathy were enrolled in this randomized, double-blind, placebo-controlled trial. The inclusion criteria were inflammatory neuropathy cause and treatment (INCAT) sensory score (ISS) ≥4 and visual analog pain scale >4 or ataxia score ≥2. The primary outcome was mean change in ISS at 12 months. Results: Twenty-six patients were randomized to a group receiving 4 weekly infusions of 375 mg/m(2) rituximab, and 28 patients to placebo. Intention-to-treat analysis, with imputation of missing ISS values by the last observation carried forward method, showed a lack of mean change in ISS at 12 months, 1.0 ± 2.7 in the rituximab group, and 1.0 ± 2.8 in the placebo group. However, changes were observed, in per protocol analysis at 12 months, for the number of patients with an improvement of at least 2 points in the INCAT disability scale (p = 0.027), the self-evaluation scale (p = 0.016), and 2 subscores of the Short Form-36 questionnaire. Conclusions: Although primary outcome measures provide no evidence to support the use of rituximab in IgM anti-MAG demyelinating neuropathy, there were improvements in several secondary outcomes in per protocol analysis. Level of evidence: This study provides Class I evidence that rituximab is ineffective in improving ISS in patients with IgM anti-MAG demyelinating neuropathy.
Article
Anti-ganglioside antibodies are frequently sought in the sera of patients with autoimmune peripheral neuropathy, using an enzyme-linked immunosorbent assay (ELISA) as the principal method for antibody detection. Wide variations in assay performance between laboratores have been reported. In this study, we established a standardized ELISA method between laboratories within the European Inflammatory Neuropathy Cause and Treatment (INCAT) group and determined the inter-laboratory variance in assay performance using both the standardized INCAT method and in-house local methods. As expected, the inter-laboratory variances were greater using local methods than using the standardized method, producing titre estimates which could be 24.8 or 7.6 times larger or smaller, respectively, than the true means for these laboratories. Using the standardized method, the within laboratory measurement error accounted for 41% of the inter-laboratory variation, providing a theoretical upper limit to which technical improvements within laboratories could reduce inter-laboratory variation. These data describe the intrinsic weaknesses within the widely used ganglioside antibody ELISA methods and reinforce the importance of inter-laboratory cooperation within this area. Standardized serological reagents used in this study are available from INCAT members.
Article
Consensus guidelines on the definition, investigation, and treatment of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) have been previously published in European Journal of Neurology and Journal of the Peripheral Nervous System. To revise these guidelines. Disease experts, including a representative of patients, considered references retrieved from MEDLINE and Cochrane Systematic Reviews published between August 2004 and July 2009 and prepared statements that were agreed in an iterative fashion. The Task Force agreed on Good Practice Points to define clinical and electrophysiological diagnostic criteria for CIDP with or without concomitant diseases and investigations to be considered. The principal treatment recommendations were: (i) intravenous immunoglobulin (IVIg) (Recommendation Level A) or corticosteroids (Recommendation Level C) should be considered in sensory and motor CIDP; (ii) IVIg should be considered as the initial treatment in pure motor CIDP (Good Practice Point); (iii) if IVIg and corticosteroids are ineffective, plasma exchange (PE) should be considered (Recommendation Level A); (iv) if the response is inadequate or the maintenance doses of the initial treatment are high, combination treatments or adding an immunosuppressant or immunomodulatory drug should be considered (Good Practice Point); (v) symptomatic treatment and multidisciplinary management should be considered (Good Practice Point).
Article
Detection of serum antibodies to myelin-associated glycoprotein (MAG) by Western blot (WB) is a valuable assay to diagnose a distinct type of demyelinating polyneuropathy with immunoglobulin M (IgM) monoclonal gammopathy. In this study, the diagnostic accuracy of a new and more practical ELISA to detect these antibodies was validated. Routine WBs from 2 independent laboratories and ELISA were used to detect anti-MAG IgM in serum from 207 patients with neuropathy and controls. The sensitivity and specificity of these assays were compared and related to the patient clinical and electrophysiologic characteristics. In ELISA, anti-MAG antibodies were found in serum from 49 (72%) of 68 patients with demyelinating polyneuropathy and IgM monoclonal gammopathy. However, in this subgroup of patients, only 30 (44%) and 37 (54%) were positive in the 2 WBs. All of the patients positive in the 2 WBs were also positive in ELISA. A high correlation was found for IgM activity in ELISA to MAG and sulfate-3-glucuronyl paragloboside (SGPG) (Spearman rho = 0.72, p < 0.0001), supporting the notion that the shared sulfated glucuronic acid moiety of MAG and SGPG is preserved. Most patients positive in anti-MAG ELISA had a slowly progressive sensory-motor demyelinating polyneuropathy, even if the WB was negative. In control groups, however, 4 WB-negative patients with a nondemyelinating monoclonal gammopathy-related polyneuropathy were positive in anti-MAG ELISA. The remaining samples were negative in ELISA. ELISA is more sensitive than Western blot to diagnose anti-myelin-associated glycoprotein related polyneuropathy, although a positive serology may be found in other forms of polyneuropathy as well.
Article
Report a double-blind, placebo-controlled study of rituximab in patients with anti-MAG demyelinating polyneuropathy (A-MAG-DP). Twenty-six patients were randomized to four weekly infusions of 375 mg/m(2) rituximab or placebo. Sample size was calculated to detect changes of > or = 1 Inflammatory Neuropathy Course and Treatment (INCAT) leg disability scores at month 8. IgM levels, anti-MAG titers, B cells, antigen-presenting cells, and immunoregulatory T cells were monitored every 2 months. Thirteen A-MAG-DP patients were randomized to rituximab and 13 to placebo. Randomization was balanced for age, electrophysiology, disease duration, disability scores, and baseline B cells. After 8 months, by intention to treat, 4 of 13 rituximab-treated patients improved by > or = 1 INCAT score compared with 0 of 13 patients taking placebo (p = 0.096). Excluding one rituximab-randomized patient who had normal INCAT score at entry, and thus could not improve, the results were significant (p = 0.036). The time to 10m walk was significantly reduced in the rituximab group (p = 0.042) (intention to treat). Clinically, walking improved in 7 of 13 rituximab-treated patients. At month 8, IgM was reduced by 34% and anti-MAG titers by 50%. CD25+CD4+Foxp3+ regulatory cells significantly increased by month 8. The most improved patients were those with high anti-MAG titers and most severe sensory deficits at baseline. Rituximab is the first drug that improves some patients with A-MAG-DP in a controlled study. The benefit may be exerted by reducing the putative pathogenic antibodies or by inducing immunoregulatory T cells. The results warrant confirmation with a larger trial.
Article
Anti-MAG IgM antibodies were detected by ELISA in a patient with slowly progressive peripheral neuropathy. Serum IgM content was normal, and no M-protein was detected by serum protein electrophoresis, immunoelectrophoresis, or immunostaining. By immunoblot analysis, the anti-MAG antibodies were IgMk; they reacted with human and bovine MAG but not with mouse MAG. The data suggest that there was an anti-MAG IgM M-protein in concentration too low to be detected by conventional techniques. Tests for anti-MAG antibodies should be done in patients with slowly progressive neuropathy of unknown etiology, even in the absence of detectable serum M-protein.