ArticlePDF Available

Obinutuzumab, a new anti-CD20 antibody, and chlorambucil are active and effective in anti-MAG antibody polyneuropathy

Wiley
European Journal of Neurology
Authors:

Abstract

Background Rituximab, a chimeric anti‐CD20 monoclonal antibody (mAb), has been used in polyneuropathy associated with anti‐MAG antibody polyneuropathy with controversial results. Herein, we report on two patients with anti‐MAG antibody neuropathy and concurrent chronic lymphocytic leukemia (CLL), who dramatically responded to obinutuzumab, a novel glycoengineered humanized anti‐CD20 mAb. Methods Patient 1 was a 82‐year‐old man with severe demyelinating sensory‐motor neuropathy. He was wheelchair‐bound, with loss of sensation up to knees. He had a CLL, IgM lambda monoclonal gammopathy, anti‐MAG antibodies >70,000BTU. Patient 2 was a 84‐year‐old women with demyelinating neuropathy, paresthesias and gait instability. She had CLL and IgM kappa paraprotein with anti‐MAG antibodies >70,000BTU. Both patients were treated with obinutuzumab intravenously at 100mg on day +1, 900mg +2, then at 1,000mg on day 8 and 15 of cycle 1 and day 1 of cycles 2‐6; chlorambucil orally at 0.5mg/kg at day 1 and 15 of cycles 1‐6. Results Patients 1 at cycle 6 was able to stand, gait was possible with monolateral support, hypoesthesia and strength improved. M‐protein and IgM level decreased. In patient 2 already after 3 cycles the monoclonal component disappeared and there was dramatic improvement of symptoms and gait normalization. At the end to therapy anti‐MAG Ab titer decreased to 5,462BTU. Neurophysiology also improved. Conclusions In our patients obinutuzumab was effective as first‐line treatment of anti‐MAG antibody polyneuropathy. CLL might have had a role in the response to therapy, but the associations might be considered in future trials. This article is protected by copyright. All rights reserved.
Accepted Article
This article has been accepted for publication and undergone full peer review but has not
been through the copyediting, typesetting, pagination and proofreading process, which may
lead to differences between this version and the Version of Record. Please cite this article as
doi: 10.1111/ene.13838
This article is protected by copyright. All rights reserved.
PROF. CHIARA BRIANI (Orcid ID : 0000-0001-8035-0200)
DR. ALESSANDRO SALVALAGGIO (Orcid ID : 0000-0002-1273-7566)
Article type : Short Communications
Obinutuzumab, a new anti-CD20 antibody, and chlorambucil are
active and effective in anti-MAG antibody polyneuropathy
Briani C1*, Visentin A2, Salvalaggio A1, Cacciavillani M3, Trentin L2.
1Neurology Unit, Department of Neuroscience, University of Padova. 2Hematology and
Clinical Immunology Unit, Department of Medicine, University of Padova; 3CEMES, Data
Medica Group, Padova, Italy.
*Correspondence to:
Chiara Briani, MD
Department of Neurosciences: Sciences, University of Padova
Via Giustiniani, 5, 35128 Padova - Italy
Tel.: +39-049-8213600 Fax: +39-049-8751770
E-mail: chiara.briani@unipd.it
Accepted Article
This article is protected by copyright. All rights reserved.
Keywords: Obinutuzumab, anti-MAG neuropathy, paraprotein, chronic lymphocytic
leukemia
Running title: Obinutuzumab in anti-MAG antibody neuropathy
Abstract
Background. Rituximab, a chimeric anti-CD20 monoclonal antibody (mAb), has been used
in polyneuropathy associated with anti-MAG antibody polyneuropathy with controversial
results.
Herein, we report on two patients with anti-MAG antibody neuropathy and concurrent
chronic lymphocytic leukemia (CLL), who dramatically responded to obinutuzumab, a novel
glycoengineered humanized anti-CD20 mAb.
Methods. Patient 1 was a 82-year-old man with severe demyelinating sensory-motor
neuropathy. He was wheelchair-bound, with loss of sensation up to knees. He had a CLL,
IgM lambda monoclonal gammopathy, anti-MAG antibodies >70,000BTU. Patient 2 was a
84-year-old women with demyelinating neuropathy, paresthesias and gait instability. She had
CLL and IgM kappa paraprotein with anti-MAG antibodies >70,000BTU. Both patients
were treated with obinutuzumab intravenously at 100mg on day +1, 900mg +2, then at
1,000mg on day 8 and 15 of cycle 1 and day 1 of cycles 2-6; chlorambucil orally at 0.5mg/kg
at day 1 and 15 of cycles 1-6.
Results. Patients 1 at cycle 6 was able to stand, gait was possible with monolateral support,
hypoesthesia and strength improved. M-protein and IgM level decreased. In patient 2 already
after 3 cycles the monoclonal component disappeared and there was dramatic improvement
Accepted Article
This article is protected by copyright. All rights reserved.
of symptoms and gait normalization. At the end to therapy anti-MAG Ab titer decreased to
5,462BTU. Neurophysiology also improved.
Conclusions. In our patients obinutuzumab was effective as first-line treatment of anti-MAG
antibody polyneuropathy. CLL might have had a role in the response to therapy, but the
associations might be considered in future trials.
Introduction.
Rituximab, a chimeric anti-CD20 monoclonal antibody (mAb), has been used in
polyneuropathy associated with anti-myelin-associated-glycoprotein (MAG) antibodies with
controversial results [1]. Obinutuzumab, a novel, type II glycoengineered humanized anti-
CD20 mAb [2], in combination with chlorambucil or bendamustine, is able to induce higher
response and longer progression-free survival in chronic lymphocytic leukemia (CLL) [3], as
compared with rituximab.
We report on two therapy-naïve anti-MAG antibody neuropathy patients who had been
successfully treated with obintuzumab and chlorambucil.
Patient 1 was a 82-year-old man, who had been complaining of gait disturbances for several
years. When we first saw the patient, he used wheelchair to travel outdoors (Inflammatory
Neuropathy Cause and Treatment, INCAT leg disability score 4), was incapable of standing
and able to walk a few steps only with bilateral support. He had bilateral distal weakness
(tibialis anterior, extensor hallucis longus, gastrocnemius 2/5 MRC bilaterally), tactile
hypoestesia and loss of vibration up to the knees, areflexia, upper limbs tremor.
Neurophysiology (performed maintaining the skin temperature at 36 °C with Keypoint
Workstation Dantec, Skovlunde, Denmark) [4] showed distal demyelinating neuropathy, with
Accepted Article
This article is protected by copyright. All rights reserved.
severe secondary axonal degeneration at lower limbs (Table). The patient had a mild clonal B
lymphocytosis CD5+ CD19+ CD23+, compatible with CLL, IgM lambda monoclonal
gammopathy. Anti-MAG antibodies were >70,000BTU. Bone marrow biopsy showed a
small infiltrate of low grade lymphoma that at immunophenotype was consistent with CLL.
IGHV gene was mutated and TP53 abnormalities, deletion and mutations, were absent. The
presence of favorable prognostic CLL markers [5] and comorbidities prompt us to use
chlorambucil+obinutuzumab. Obinutuzumab was given intravenously at 100mg on day +1,
900mg +2, then at 1000mg on day 8 and 15 of cycle 1 and day 1 of cycles 2-6; chlorambucil
orally at 0.5mg/kg at day 1 and 15 of cycles 1-6. At cycle 6 the patient was able to stand, gait
was possible with monolateral support (INCAT leg disability 3), tactile hypoestesia was
limited to the feet, distal strength slightly improved (3/5 MRC), and vibration was regained at
toes. Monoclonal protein decreased from 15.8g/L to 8.46g/L. Similarly, total IgM (14.8 vs
6.7g/L), and lymphocyte (6,340 vs 900/µl) decreased. Anti-MAG antibody titer remained
stable. Two months after the end of treatment the patients died of pneumonia.
Patient 2 was a 84-year-old women with a 10 months history of painful paresthesias at lower
limbs and gait instability (INCAT leg disability 1). Neurophysiology revealed distal
symmetric demyelinating neuropathy (Table). Blood tests revealed the presence of CLL
(9,100/µL clonal B lymphocytes CD5+ CD19+ CD23+ CD200+) and IgM kappa paraprotein.
Anti-MAG antibodies were >70,000BTU. Bone marrow biopsy confirmed the presence of
15% CLL cells. Chlorambucil+obinutuzumab were administered as the same schedule as in
the first patient.
Already after 3 cycles the monoclonal component disappeared and the patient reported
dramatic improvement of symptoms and gait normalization (INCAT leg disability 0). At the
end of cycle 6 lymphocytes decreased to 1,260/mL, monoclonal component persisted absent,
Accepted Article
This article is protected by copyright. All rights reserved.
IgM 0.6g/L, anti-MAG antibodies were 5,462BTU. Neurophysiological findings also
improved (Table, Figure).
Both patients completed all the planned treatment. Both developed grade 2 neutropenia (i.e.
neutrophils count between 500-1,000/L), one reported 2 grade 3 pneumonias requiring
hospitalization and one grade 2 anemia (i.e. Hb 80-100g/l). None reported infusion-related
drug reactions.
Discussion.
Anti-MAG antibody neuropathy is a slowly progressive neuropathy, that may become
disabling when sensory ataxia worsen or motor impairment occur. Anti-MAG antibodies are
pathogenic [6], and Rituximab seems to be effective regardless of the associated
hematological disease [7]. However rituximab provides benefit only in a subgroup of
patients, and repeated cycles are often necessary with progressive loss of efficacy. We have
described two patients with anti-MAG antibody neuropathy and CLL successfully treated
with obinutuzumab-chlorambucil as first therapy. Obinutuzumab is a type II, glycoengineered
humanized anti-CD20 mAb significantly more potent than rituximab in depleting B-cells [2-
3]. In the phase III CLL11 study, 781 patients with untreated elderly CLL and comorbidities
were randomized to chlorambucil vs chlorambucil-rituximab vs chlorambucil-obinutuzumab
[3]. Chlorambucil-obinutuzumab achieved higher complete remission rate, longer progression
free survival, and improved overall survival. Both our patients developed neutropenia and
one developed a fatal pneumonia, known expected adverse events for elderly CLL patients
with comorbidities receiving chemo-immunotherapy [8].
Accepted Article
This article is protected by copyright. All rights reserved.
Rakocevic et al. recently described two patients unsuccessfully treated with obinutuzumab,
after failure or loss of efficacy of rituximab [9]. Despite loss of clinical efficacy, the authors
reported a great effect of obinutuzumab on anti-MAG antibodies titer and on IgM levels,
indicating the biological efficacy of the drug. As the authors themselves hypothesized, the
presence of axonal damage and the long history of the neuropathy might have contributed to
the little response. Also in our first patient the neuropathy had progressed to severe motor
impairment, but the clinical response at 6 months therapy was evident. Unfortunately the
patients later died of pneumonia. In patient 2, who started with less severe neuropathy, the
efficacy was clear already at the third cycle (third month) of therapy and further improved
after the sixth cycle. Also neurophysiology improved. Our data indicate that obinutuzumab-
chlorambucil may be effective in anti-MAG antibody polyneuropathy as first-line treatment,
independently of the neuropathy severity. The associated CLL might have had a role in the
response to therapy, and also chlorambucil itself might have contributed to the response [10].
Results on a wider sample of patients are however needed. Caution is warranted for potential
adverse effect in older patients.
Acknowledgements: None
Conflicts of interest and source of funding: None
Accepted Article
This article is protected by copyright. All rights reserved.
References
1. Lunn MP, Nobile-Orazio E. Immunotherapy for IgM anti-myelin-associated
glycoprotein paraprotein-associated peripheral neuropathies. Cochrane Database Syst
Rev 2016;10:CD002827.
2. Mossner E, Brunker P, Moser S, et al. Increasing the efficacy of CD20 antibody
therapy through the engineering of a new type II anti-CD20 antibody with enhanced
direct and immune effector cell-mediated B-cell cytotoxicity. Blood. 2010, 4393
4402.
3. Goede V, Fischer K, Engelke A, et al. Obinutuzumab as frontline treatment of
chronic lymphocytic leukemia: updated results of the CLL11 study. Leukemia 2015,
16021604.
4. Frigeni B, Cacciavillani M, Ermani M, et al. Neurophysiological examination of
dorsal sural nerve. Muscle Nerve. 2012;46:895-898.
5. Visentin A, Facco M, Frezzato F, et al. Integrated CLL Scoring System, a New and
Simple Index to Predict Time to Treatment and Overall Survival in Patients With
Chronic Lymphocytic Leukemia. Clin Lymphoma Myeloma Leuk. 2015;15:612-
20.e1-5
6. Dalakas MC. Advances in the diagnosis, immunopathogenesis and therapies of IgM-
anti-MAG antibody-mediated neuropathies. Ther Adv Neurol Disord 2018;
15;11:1756285617746640.
7. Campagnolo M, Zambello R, Nobile-Orazio E, et al. IgM MGUS and Waldenstrom-
associated anti-MAG neuropathies display similar response to rituximab therapy. J
Neurol Neurosurg Psychiatry 2017;88:1094-1097.
Accepted Article
This article is protected by copyright. All rights reserved.
8. Visentin A, Compagno N, Cinetto F, et al. Clinical profile associated with infections
in patients with chronic lymphocytic leukemia. Protective role of immunoglobulin
replacement therapy. Haematologica 2015;100:e515e518.
9. Rakocevic G, Martinez-Outschoorn U, Dalakas MC. Obinutuzumab, a potent anti-B-
cell agent, for rituximab-unresponsive IgM anti-MAG neuropathy. Neurol
Neuroimmunol Neuroinflamm. 2018;5(4):e460.
10. Oksenhendler E, Chevret S, Léger JM, Louboutin JP, Bussel A, Brouet JC. Plasma
exchange and chlorambucil in polyneuropathy associated with monoclonal IgM
gammopathy. IgM-associated Polyneuropathy Study Group. J Neurol Neurosurg
Psychiatry. 1995;59:243-247.
Legend to Table: DL: distal latency; CV: conduction velocity; cMAP: compound motor
action potential; SAP: sensory action potential; BE: below elbow; AE: above elbow; BFH:
below fibular head; LPF: lateral popliteal fossa; PF: popliteal fossa; APB: abductor pollicis
brevis; ADM: abductor digiti minimi; EDB: extensor digitorum brevis; EDL: extensor
digitorum longus; TA, tibialis anterior; AH, abductor hallucis; NR: no response; R: right; L:
left; m: motor; s: sensory; TLI: terminal latency index.
Normal values: median nerve DL ≤3,5 ms; SCV ≥48 m/s; SAP ≥15 µV; MCV ≥50 m/s;
cMAP ≥6 mV; ulnar nerve DL ≤3,1 ms; SCV ≥48 m/s; SAP ≥10 µV; MCV ≥50 m/s; cMAP
≥4 mV; radial nerve SCV ≥40 m/s; SAP ≥10 µV; peroneal nerve DL ≤5,5 ms; MCV ≥40 m/s;
cMAP ≥3 mV; tibial nerve DL ≤6 ms; MCV ≥40 m/s; cMAP ≥4 mV; sural nerve SCV ≥50
m/s; SAP ≥5 µV; F-wave median/ulnar < 32 ms; F-wave peroneal < 56 ms.
Accepted Article
This article is protected by copyright. All rights reserved.
SA: spontaneous activity; Rec: recruitment; Amp: amplitude; Dur: duration; Poly:
polyphasia; N: normal; 0 absent; ++ moderate; : increased; ↓: slight reduction; ↓↓: moderate
reduction; ↓↓↓: marked reduction; NR: no motor units recruited.
Legend to Figure: left (A,C) and right (B,D) peroneal nerve conduction studies in patient 2.
To note the significant reduction of Distal Latency (clearly evident when comparing the
latency to negative peak) before (A,B) and after (C,D) therapy.
Accepted Article
This article is protected by copyright. All rights reserved.
Patient 1
Nerve conduction studies
Needle EMG
Nerve
Stimulation
site
DL
(ms)
CV
(m/s)
Amplitude
(cMAP=mV
SAP=µV)
Total
duration
(ms)
F
Latency
(ms)
Muscle
SA
Rec
Amp
Dur
Poly
L Median (m)
Wrist
6.43
5.6
14.8
39
L Deltoid
0
N
N
N
N
Elbow
32
4.3
17.9
R Deltoid
0
N
N
N
N
L Ulnar (m)
Wrist
4.27
4.0
13.6
41
L Biceps brachii
0
N
N
N
N
BE
35
3.6
17.1
L Triceps brachii
0
N
N
N
N
AE
32
3.4
17.3
L 1st dorsal interosseous
0
N
N
N
N
L Peroneal (m)
Ankle
NR
R 1st dorsal interosseous
0
N
N
N
N
L Peroneal (m)
BFH
17.1
0.74
25.6
L Gluteus maximus
0
N
N
N
N
LPF
23
0.74
27.2
R Gluteus maximus
0
N
N
N
N
R Tibial (m)
Ankle
NR
L Biceps femoralis
0
N
N
N
N
PF
R Biceps femoralis
0
N
N
N
N
L Median (s)
1th finger
NR
L Vastus lateralis
0
N
N
N
N
3rd finger
NR
R Vastus lateralis
0
N
N
N
N
L Ulnar (s)
5th finger
NR
L Vastus medialis
0
N
N
N
N
L Radial (s)
1th finger
NR
R Vastus medialis
0
N
N
N
N
R Sural (s)
Mild calf
NR
L Tibialis anterior
0
↓↓↓
↓↓↓
L Sural (s)
Mild calf
NR
R Tibialis anterior
0
↓↓↓
↓↓↓
L Extensor digitorum comunis
++
↓↓↓
↓↓↓
R Extensor digitorum comunis
++
↓↓↓
↓↓↓
L Gastrocnemius
++
↓↓↓
↓↓↓
R Gastrocnemius
++
↓↓↓
↓↓↓
L Extensor digitorum brevis
++
NR
Accepted Article
This article is protected by copyright. All rights reserved.
R Extensor digitorum brevis
++
NR
L Abductor hallucis
++
NR
R Abductor hallucis
++
NR
Patient 2
Nerve Conduction Studies
Nerve Conduction Studies, follow up
Nerve
Stimulation
site
DL
(ms)
CV
(m/s)
Amplitude
(cMAP=mV
SAP=µV)
Total
duration
(ms)
F
Latency
(ms)
DL
(ms)
CV
(m/s)
Amplitude
(cMAP=mV
SAP=µV)
Total
duration
(ms)
F
Latency
(ms)
L Median (m)
Wrist
15.0
2.4
17.2
49
9.52
3.3
15.6
38
Elbow
34
2.4
17.5
46
3.3
16.1
R Median (m)
Wrist
14.3
1.0
16.1
7.85
2.4
15.9
36
Elbow
37
1.4
16.1
43
2.4
16.8
R Ulnar (m)
Wrist
4.37
5.6
12.7
35
2.93
4.4
12.2
32
BE
48
5.4
13.6
53
4.4
13.0
AE
44
5.2
14.0
49
4.3
13.5
L Peroneal (m)
Ankle
8.31
2.5
15.8
79
5.73
2.5
12.9
65
LPF
30
1.7
19.3
35
1.8
21.1
R Peroneal (m)
Ankle
8.21
3.3
12.7
5.16
4.4
10.3
LPF
40
3.3
14.5
38
3.0
12.8
L Tibial (m)
Ankle
11.8
0.2
27.5
9.56
0.3
15.9
PF
27
0.2
29.1
35
0.3
19.4
R Tibial (m)
Ankle
13.2
0.7
34.7
9.34
0.7
16.7
PF
28
0.6
35.9
34
1.1
16.9
L Median (s)
1th finger
NR
NR
3rd finger
NR
NR
R Median (s)
1th finger
NR
NR
Accepted Article
This article is protected by copyright. All rights reserved.
3rd finger
NR
NR
L Ulnar (s)
5th finger
NR
2.85
40
2.2
1.88
R Ulnar (s)
5th finger
NR
2.70
38
2.0
1.52
L Radial (s)
1th finger
2.29
36
5.0
1.36
2.07
44
6.4
1.48
R Radial (s)
1th finger
2.37
39
2.4
1.83
2.37
39
2.4
1.83
R Sural (s)
Mild calf
3.34
42
3.5
1.83
2.93
48
3.6
1.56
L Sural (s)
Mild calf
NR
3.28
43
2.8
1.56
Accepted Article
This article is protected by copyright. All rights reserved.
... Therefore, rituximab is currently used in the clinical practice either alone or in combination with cyclophosphamide [36], fludarabine [37] or bendamustine [38]. Obinutuzumab, a humanized glycoengineered anti-CD20 monoclonal antibody, has also been used as a possible alternative treatment in patients with anti-MAG antibody neuropathy, with controversial results and concerns regarding possible toxicity [39,40]. ...
... Development of peripheral neuropathy was more common in previously treated subjects, with TP53 deletion and serum monoclonal gammopathy [95]. Two CLL patients with concurrent anti-MAG antibody polyneuropathy were treated with six cycles of chlorambucil-obinutuzumab [40], showing persistent hematological and neurological remissions (Table 1). However, both developed pneumonia that required hospitalization [40]. ...
... Two CLL patients with concurrent anti-MAG antibody polyneuropathy were treated with six cycles of chlorambucil-obinutuzumab [40], showing persistent hematological and neurological remissions (Table 1). However, both developed pneumonia that required hospitalization [40]. Despite the efficacy, caution is warranted for potential adverse effects in aged patients. ...
Article
Full-text available
Simple Summary The peripheral nervous systems may be involved by several hematological diseases ranging from preneoplastic diseases to overt malignancies or paraneoplastic syndromes. In most cases, a monoclonal paraprotein plays a pivotal role in the damage of peripheral nervous systems through different mechanisms. For these reasons, the multidisciplinary approach between hematologist and neurologist is fundamental to correctly diagnose and treat monoclonal gammopathies of neurological significance. We reviewed the biologic, clinic and neurophysiological features, as well as tailored treatments of monoclonal gammopathies of neurological significance. Abstract Monoclonal gammopathy and peripheral neuropathy are common diseases of elderly patients, and almost 10% of patients with neuropathy of unknown cause have paraprotein. However, growing evidence suggests that several hematological malignancies synthesize and release monoclonal proteins that damage the peripheral nervous system through different mechanisms. The spectrum of the disease varies from mild to rapidly progressive symptoms, sometimes affecting not only sensory nerve fibers, but also motor and autonomic fibers. Therefore, a multidisciplinary approach, mainly between hematologists and neurologists, is recommended in order to establish the correct diagnosis of monoclonal gammopathy of neurological significance and to tailor therapy based on specific genetic mutations. In this review, we summarize the spectrum of monoclonal gammopathies of neurological significance, their distinctive clinical and neurophysiological phenotypes, the most relevant pathophysiological events and new therapeutic approaches.
... 44 Obinutuzumab, a third-generation humanized anti-CD20 therapy approved for CLL, has been suggested to achieve more effective depletion (compared with rituximab) not only of peripheral B cells but also in lymphoid tissue. 45 Here, obinutuzumab and chlorambucil were used as first-line treatment in our patient with CLL-MG, and therefore, we cannot derive a comparative statement regarding anti-CD20 therapies (as it has been done for other CLL-associated/ autoimmune diseases, e.g., autoimmune hemolytic anemia, immune thrombocytopenia, 46 rheumatoid arthritis, 47 or anti-MAG neuropathy 48,49 ). Nevertheless, our patient with CLL-MG was successfully treated early on, and obinutuzumab and chlorambucil had lasting effects and were safe. ...
Article
Full-text available
Background and Objectives Myasthenia gravis (MG) can in rare cases be an autoimmune phenomenon associated with hematologic malignancies such as chronic lymphocytic leukemia (CLL). It is unclear whether in patients with MG and CLL, the leukemic B cells are the ones directly driving the autoimmune response against neuromuscular endplates. Methods We identified patients with acetylcholine receptor antibody–positive (AChR ⁺ ) MG and CLL or monoclonal B-cell lymphocytosis (MBL), a precursor to CLL, and described their clinical features, including treatment responses. We generated recombinant monoclonal antibodies (mAbs) corresponding to the B-cell receptors of the CLL phenotype B cells and screened them for autoantigen binding. Results A computational immune cell screen revealed a subgroup of 5/38 patients with MG and 0/21 healthy controls who displayed a CLL-like B-cell phenotype. In follow-up hematologic flow cytometry, 2 of these 5 patients were diagnosed with an MBL. An additional patient with AChR ⁺ MG as a complication of manifest CLL presented at our neuromuscular clinic and was successfully treated with the anti-CD20 therapy obinutuzumab plus chlorambucil. We investigated the specificities of expanding CLL-like B-cell clones to assess a direct causal link between the 2 diseases. However, we observed no reactivity of the clones against the AChR, antigens at the neuromuscular junction, or other common autoantigens. Discussion Our study suggests that AChR autoantibodies are produced by nonmalignant, polyclonal B cells The new anti-CD20 treatment obinutuzumab might be considered in effectively treating AChR ⁺ MG. Classification of Evidence This is a single case study and provides Class IV evidence that obinutuzumab is safe to use in patients with MG.
... These include BCL2 inhibitors such as venetoclax [197], anti-CXCR4 monoclonal antibodies (ulocuplumab) [198], next-generation proteasome inhibitors [199] and next-generation anti-CD20 agents, such as ofatumumab and obinutuzumab [200]. Obinutuzumab causes enhanced B-cell-depleting activity compared to rituximab [201] and may be effective in treating (rituximab non-responding) IgM gammopathy anti-MAG polyneuropathy [202,203]. ...
Article
Full-text available
With increasing age, the chances of developing either MGUS or polyneuropathy increase as well. In some cases, there is a causative relationship between the IgM M-protein and polyneuropathy. In approximately half of these cases, IgM targets the myelin-associated glycoprotein (MAG). This results in chronic polyneuropathy with slowly progressive, predominantly sensory neurological deficits and distally demyelinating features in nerve conduction studies. Despite the disease being chronic and developing slowly, it can cause considerable impairment. We reviewed English medical publications between 1980 and May 2022 on IgM gammopathy-associated polyneuropathy, with special attention to studies addressing the pathophysiology or treatment of anti-MAG polyneuropathy. Treatment options have been limited to a temporizing effect of intravenous immunoglobulins in some patients and a more sustained effect of rituximab but in only 30 to 55 percent of patients. An increase in our knowledge concerning genetic mutations, particularly the MYD88L265P mutation, led to the development of novel targeted treatment options such as BTK inhibitors. Similarly, due to the increasing knowledge of the pathophysiology of anti-MAG polyneuropathy, new treatment options are emerging. Since anti-MAG polyneuropathy is a rare disease with diverse symptomatology, large trials with good outcome measures are a challenge.
... Subsequently Briani et al. reported on two drug-naive patients with anti-MAG antibody neuropathy and CLL who were treated with obinutuzumab and chlorambucil as first-line therapy, with significant clinical and neurophysiological improvement. However, both patients required hospitalization for severe pneumonia [92]. ...
Article
Full-text available
Autoimmune diseases of the peripheral nervous system have so far been treated mainly with exogenous high-dose intravenous immunoglobulins (IVIg), that act through several mechanisms, including neutralization of pathogenic autoantibodies, modulation of lymphocyte activity, interference with antigen presentation, and interaction with Fc receptors, cytokines, and the complement system. Other therapeutic strategies have recently been developed, in part to address the increasing shortage of IVIg, prime among which is the use of B cell depleting monoclonal antibodies, or small molecule inhibitors targeting the B-cell specific kinases. Rituximab, a chimeric monoclonal antibody against CD20 + B lymphocytes, is currently the most used, especially in anti-MAG antibody neuropathy and autoimmune neuropathies with antibodies to nodal/paranodal antigens that are unresponsive to IVIg. After several reports of its efficacy in chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), rituximab is currently under investigation in three Phase 2 trials in CIDP. In addition, the possible role of complement activation in the pathogenesis of chronic autoimmune neuropathies has brought into consideration drugs that can block the complement cascade, such as eculizumab, a monoclonal antibody already assessed in acute polyradiculoneuropathies, and approved for myasthenia gravis. Preliminary data on eculizumab in multifocal motor neuropathy have been published, but randomized controlled studies are pending. Moreover, the neonatal Fc receptor, that recycles IgGs by preventing their lysosome degradation, is an important and attractive pharmacological target. Antibodies against FcRn, which reduce circulating IgG (both pathogenic and non-pathogenic) have been developed. The FcRn blocker efgartigimod, a humanized IgG1-derived Fc fragment, which competitively inhibits the FcRn, has recently been approved for the treatment of myasthenia gravis and is currently under investigation in CIDP. In addition, the anti-human FcRn monoclonal antibody rozanolixizumab is currently being assessed in phase 2 trials in CIDP. However, none of the abovementioned monoclonal antibodies is currently approved for treatment of any immune-mediated neuropathies. While more specific and individualized therapies are being developed, the possibility of combined treatments targeting different pathogenic mechanisms deserves consideration as well.
... Obinutuzumab, an anti-CD20 antibody that targets a different epitope than the other anti-CD20 mAbs, as depicted in Fig. 4, is also resistant (in contrast to rituximab) to internalization by B cells through FcRIIbeta, thereby achieving more effective depletion in theory [5]. Obinutuzumab was tried in 2 anti-MAG-positive patients unresponsive to rituximab and was ineffective in our hands [180]; however, others found some benefit [181]. In MMN, rituximab can be effective in anecdotal series of patients insufficiently responding to IVIg. ...
Article
Full-text available
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antigen-presenting cells facilitating antibody production but also as sensors, coordinators, and regulators of the immune response. In particular, B cells can regulate the T cell activation process through their participation in antigen presentation, production of proinflammatory cytokines (bystander activation or suppression), and contribution to ectopic lymphoid aggregates. Such an important interplay between B and T cells makes therapeutic depletion of B cells an attractive treatment strategy. The last decade, anti-B cell therapies using monoclonal antibodies against B cell surface molecules have evolved into a rational approach for successfully treating autoimmune neurological disorders, even when T cells seem to be the main effector cells. The paper summarizes basic aspects of B cell biology, discusses the roles of B cells in neurological autoimmunities, and highlights how the currently available or under development anti-B cell therapeutics exert their action in the wide spectrum and immunologically diverse neurological disorders. The efficacy of the various anti-B cell therapies and practical issues on induction and maintenance therapy is specifically detailed for the treatment of patients with multiple sclerosis, neuromyelitis-spectrum disorders, autoimmune encephalitis and hyperexcitability CNS disorders, autoimmune neuropathies, myasthenia gravis, and inflammatory myopathies. The success of anti-B cell therapies in inducing long-term remission in IgG4 neuroautoimmunities is also highlighted pointing out potential biomarkers for follow-up infusions.
... However, given the high unmet medical need, over the last 3 decades, many different immunotherapies have been used for the management of anti-MAG neuropathy including IV immunoglobulins (IVIg), therapeutic plasma exchange, chemotherapeutic drugs, and various biologic drugs such as rituximab and obinutuzumab. 8,10,13,14 The significance of the anti-MAG antibody titers or levels as predictive of response to therapy is controversial. Although there is considerable evidence for the pathogenicity of anti-MAG IgM autoantibodies, the association of reduced serum levels of anti-MAG IgM autoantibodies and clinical improvement of neuropathic symptoms is less clear based on the available literature and reviews. ...
Article
Full-text available
Background and Objectives The objective of the retrospective analysis was to test the hypothesis that changes in serum anti-myelin-associated glycoprotein (MAG) autoantibodies are associated with clinical response to immunotherapy in patients with anti-MAG neuropathy. Methods As of January 29, 2020, we used anti-myelin-associated glycoprotein-related search strings in the Medline database to identify studies that provided information on anti-MAG immunoglobulin M (IgM) autoantibodies and clinical outcomes during immunotherapies. The relative change in anti-MAG IgM titers, paraprotein levels, or total IgM was determined before, during, or posttreatment, and the patients were assigned to “responder,” “nonresponder,”’ or “acute deteriorating” category depending on their clinical response to treatment. The studies were qualified as “supportive” or “not supportive” depending on the percentage of patients exhibiting an association between relative change of anti-MAG antibody titers or levels and change in clinical outcomes. Results Fifty studies with 410 patients with anti-MAG neuropathy were included in the analysis. Forty studies with 303 patients supported the hypothesis that a “responder” patient had a relative reduction of anti-MAG antibody titers or levels that is associated with clinical improvements and “nonresponder” patients exhibited no significant change in anti-MAG IgM antibodies. Six studies with 93 patients partly supported, and 4 studies with 26 patients did not support the hypothesis. Discussion The retrospective analysis confirmed the hypothesis that a relative reduction in serum anti-MAG IgM antibodies is associated with a clinical response to immunotherapies; a sustained reduction of at least 50% compared with pretreatment titers or levels could be a valuable indicator for therapeutic response.
... • pathological studies of sural nerves show deposits of IgM and complement in myelin sheets, suggesting the need for complement activation in the demyelination process [19]; • IgM recognize NCAM (Neural Cell Adhesion Molecules) and are seen in correspondence of MAG in demyelinated areas [13]; in skin biopsies of the same patients there is a concurrent localization of IgM, C3d complement, and MAG in the dermal myelinated fibers, leading to the loss of nerve fibers [20]; • feline nerves injected with the serum of patients with anti-MAG/SGPG IgM supplemented with additional complement, develop complement-mediated demyelination and conduction block within 2-9 days [21]; • systemic transfusion of chickens with anti-MAG IgM produces segmental demyelination with IgM deposits on external myelin sheets and consequent widening of myelin lamellae as observed in human pathology [22]; • cats immunized with purified SGPG develop an ataxic neuropathy with the involvement of dorsal root ganglia, similar to anti-MAG antibody neuropathy [23]; • patients with anti-MAG antibody neuropathy respond to immunomodulant therapies, especially monoclonal antibodies (i.e., rituximab, obinutuzumab, ibrutinib) [24][25][26], and therapy response seems to correlate with the reduction of anti-MAG antibodies titers [27][28][29]. ...
Article
Full-text available
Despite the introduction of non-invasive techniques in the study of peripheral neuropathies, sural nerve biopsy remains the gold standard for the diagnosis of several neuropathies, including vasculitic neuropathy and neurolymphomatosis. Besides its diagnostic role, sural nerve biopsy has helped to shed light on the pathogenic mechanisms of different neuropathies. In the present review, we discuss how pathological findings helped understand the mechanisms of polyneuropathies complicating hematological diseases.
Article
Background and purpose The aim of this study was to determine the prevalence of anti‐myelin‐associated glycoprotein (MAG) neuropathy and the current status of such patients in Japan. Methods We conducted a nationwide survey in 2021 using established epidemiological methods. Questionnaires were sent to all neurology and pediatric neurology departments throughout Japan to identify patients with anti‐MAG neuropathy. An initial questionnaire was used to determine the number of patients, with a second one used to collect detailed clinical information. Results The estimated number of patients with anti‐MAG neuropathy was 353, with a prevalence of 0.28 per 100,000 and an incidence of 0.05 per 100,000. The detailed clinical profiles of 133 patients were available. The median (range) age of onset was 67 (30–87) years, with a prominent peak in the age range 66–70 years, and the male‐to‐female ratio was 3.6. Most patients had distal sensory‐predominant polyneuropathy, and neuropathic pain (50%), or sensory ataxia (42%), while 18% had Waldenström's macroglobulinemia or multiple myeloma. Intravenous immunoglobulin was the most frequently used treatment (65%), but the response rate was <50%, whereas rituximab was given in 32% of patients, and 64% of these showed improvement. At the last visit, 27% of patients could not walk independently. Conclusions This study on anti‐MAG neuropathy provides updated insights into the epidemiology of this disease, clinical profiles, and treatment approaches in Japan. Rituximab therapy, used for only one‐third of the patients, demonstrated efficacy. During the final visit, a quarter of the patients were unable to walk independently. Further studies are warranted to determine the optimal management of this rare and intractable disorder.
Article
OBJECTIVE Coexistence of polyneuropathy and gammopathy is a common but potentially challenging situation in clinical practice. This article reviews the clinical, electrophysiologic, and hematologic phenotypes of the paraproteinemic neuropathies and the diagnostic and treatment strategies for each. LATEST DEVELOPMENTS Advances in our understanding of the underlying pathophysiology of various paraproteinemic neuropathies and their corresponding phenotypes have identified potential new therapeutic targets. Therapeutic strategies to diminish anti–myelin-associated glycoprotein (MAG) IgM antibodies have shown partial and inconsistent efficacy; however, antigen-specific immune therapy is being investigated as a novel treatment to remove the presumably pathogenic anti-MAG antibody. Advances in genetic and cell signaling studies have resulted in the approval of Bruton tyrosine kinase inhibitors for Waldenström macroglobulinemia. Monoclonal antibodies are being investigated for the treatment of light chain amyloidosis. ESSENTIAL POINTS Early recognition and treatment of underlying plasma cell disorders improves clinical outcomes in patients with paraproteinemic neuropathy. Despite significant progress, our knowledge regarding underlying mechanisms for paraproteinemic neuropathy is still limited. Clinicians’ awareness of clinical phenotypes, electrophysiologic hallmarks, and hematologic findings of the different paraproteinemic neuropathies is crucial to promptly identify and treat patients and to avert misdiagnosis. Multidisciplinary collaboration among specialists, including neurologists and hematologists, is paramount for the optimal treatment of these patients with overlapping conditions.
Article
Full-text available
Background and Objectives Neuropathy with antibodies to myelin-associated glycoprotein (MAG) is the most common paraproteinemic IgM neuropathy. Recently, the mutational profile of the MYD88 and CXCR4 genes has been included in the diagnostic workup of IgM monoclonal gammopathies. The objective of our study was to assess the prevalence of MYD88 L265P and CXCR4 S338X gene variants in patients with anti-MAG antibody neuropathy. Secondary aims were to evaluate possible correlations between the mutational profile and neuropathy severity, antibody titers, and treatment response. Methods Seventy-five patients (47 men, mean age at molecular analysis 70.8 ± 10.2 years; mean disease duration 5.1 ± 4.9 years) with anti-MAG antibody neuropathy were recruited. Among them, 38 (50.7%) had IgM monoclonal gammopathy of undetermined significance, 29 (38.7%) Waldenstrom macroglobulinemia (WM), and 8 (10.6%) chronic lymphocytic leukemia/marginal zone lymphoma/hairy cell leukemia variant. Molecular analysis was performed on DNA from the bone marrow mononuclear cells in 55 of 75 patients and from peripheral mononuclear cells in 18 of 75 patients. Forty-five patients were treated with rituximab, 6 with ibrutinib, 2 with obinutuzumab-chlorambucil, and 3 with venetoclax-based therapy. All the patients were assessed with the Inflammatory Neuropathy Cause and Treatment (INCAT) Disability Scale, INCAT Sensory Sum Score, and MRC Sum Score at baseline and follow-up. We considered as responders, patients who improved by at least 1 point in 2 clinical scales. Results Fifty patients (66.7%) carried the MYD88 L265P variant, with a higher frequency in WM and naive patients (77.2% vs 33.3%, p = 0.0012). No patients harbored the CXCR4 S338X variant. There were no significant differences in hematologic data (IgM levels, M protein, and anti-MAG antibody titers), neuropathy severity, or response to rituximab in MYD88 -altered and MYD88 wild-type patients. Nine of 11 (81.8%) patients treated with novel targeted drug, according to the MYD88 status, responded to treatments. Discussion MYD88 L265P variant has a high prevalence (66.7%) in anti-MAG antibody neuropathy representing a potential effective mutational target for Bruton tyrosine kinase inhibitors. MYD88 L265P variant, however, does not seem to be a prognostic factor of neuropathy severity or response to rituximab. In patients not responding or becoming refractory to rituximab, a tailored therapy with new effective target therapies should be considered.
Article
Full-text available
Anti-MAG demyelinating neuropathy is difficult to treat. All immunotherapies have failed except for rituximab, a chimeric B-cell-depleting monoclonal antibody against CD20, that helps up to 40% of patients based on 2 controlled and several uncontrolled series.¹⁻³ Because the majority of these patients are left disabled, stronger anti-B-cell agents might be promising.
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
CD20 is an important target for the treatment of B-cell malignancies, including non-Hodgkin lymphoma as well as autoimmune disorders. B-cell depletion therapy using monoclonal antibodies against CD20, such as rituximab, has revolutionized the treatment of these disorders, greatly improving overall survival in patients. Here, we report the development of GA101 as the first Fc-engineered, type II humanized IgG1 antibody against CD20. Relative to rituximab, GA101 has increased direct and immune effector cell-mediated cytotoxicity and exhibits superior activity in cellular assays and whole blood B-cell depletion assays. In human lymphoma xenograft models, GA101 exhibits superior antitumor activity, resulting in the induction of complete tumor remission and increased overall survival. In nonhuman primates, GA101 demonstrates superior B cell-depleting activity in lymphoid tissue, including in lymph nodes and spleen. Taken together, these results provide compelling evidence for the development of GA101 as a promising new therapy for the treatment of B-cell disorders.
Article
Full-text available
The study compared chlorambucil alone with chlorambucil in combination with plasma exchange in patients with polyneuropathy associated with monoclonal IgM. Forty four patients were prospectively randomly assigned, in a comparative open trial, to receive either 0.1 mg/kg/day chlorambucil orally, for 12 months or chlorambucil associated with 15 courses of plasma exchange, during the first four months of treatment. They were evaluated by a neuropathy disability score and nerve conduction studies. No difference was found between the two treatment groups. The average neuropathy disability score improved by 2.1 points from baseline (21.0 to 18.9) in the chlorambucil group and by 1.8 points (20.4 to 18.6) in the chlorambucil + plasma exchange group (P = 0.70). The mean motor nerve conduction velocity decreased from 20.0 to 18.2 m/s in the chlorambucil group and increased from 20.5 to 22.5 m/s in the chlorambucil + plasma exchange group (P = 0.51). A slight improvement of the sensory component of the neuropathy disability score (from 10.5 to 8.3) was noted in both groups (P = 0.01). At the end of the study and according to self evaluation, 15 patients--eight from the chlorambucil group and seven from the chlorambucil + plasma exchange group--reported clinical improvement, whereas 15--eight from the chlorambucil group and seven from the chlorambucil + plasma exchange group--reported clinical worsening. Neuropathy remained stable in the others. Thus plasma exchange seemed to confer no additional benefit in the treatment of polyneuropathy associated with monoclonal IgM.
Article
No adequate immunotherapy has so far been shown to be effective in anti-myelin-associated glycoprotein (MAG) antibody neuropathy.1 Rituximab was assessed in two randomised controlled trials, both including only patients with monoclonal gammopathy of undetermined significance (MGUS), with Waldenstrom’s macroglobulinaemia (WM) being an exclusion criterion.2 The aim of our multicentre retrospective study was to assess whether the response to rituximab in patients with anti-MAG antibody neuropathy would differ according to the associated haematological condition. We reviewed the clinical and laboratory features of 33 patients (21 men, mean age at time of therapy: 64.6±10.3, range: 41–87, mean neuropathy duration at time of therapy: 4.67 years±4.82, range: 0.5–17) with anti-MAG antibody neuropathy who underwent therapy with rituximab single agent in five Italian neurological centres. Twenty-five (73.5%) patients had Immunoglobulin M (IgM) MGUS and 8/33 had WM, according to bone marrow biopsy. The mean age was significantly different in the two groups (62.5±10.3 years in MGUS vs 70.5±7.9 years in WM, p=0.04). The mean neuropathy duration was 4.56±4.8 years in MGUS versus 5.6±5.1 years in WM (p=0.62, Mann-Whitney U test). Anti-MAG antibodies were assayed by ELISA (Buhlmann Laboratories AG) (28 patients) or Western blot (5 patients). The median antibodies titre was 51 200 (range: 7500–800 000) in MGUS and 56 000 (range: 11 126–600 000) in patients with WM. Patients with low (<10 000 Buhlmann Titer Units) titre were included for clinical and neurophysiological features consistent with anti-MAG neuropathy. Antibody titres were not retrievable in two patients. Twenty patients were therapy-naive, while 13 …
Article
Leukemia is one of the leading journals in hematology and oncology. It is published monthly and covers all aspects of the research and treatment of leukemia and allied diseases. Studies of normal hemopoiesis are covered because of their comparative relevance.
Article
Introduction: Nerve conduction study of the dorsal sural nerve (DSN) has been reported to be a sensitive method for early detection of peripheral neuropathies. However, normal reference values are scarce and vary greatly among the different studies. Methods: A comprehensive neurophysiological study, including nerve conduction velocity (NCV) and sensory nerve action potential (SNAP) recording, was performed in 294 healthy subjects (21-86 years) with no evidence of neuropathy. Results: The amplitude of the DSN SNAP ranged from 2.50 to 15.90 μV, and NCV ranged from 28.9 to 52.8 m/s. A significant age-related decrease in DSN SNAP amplitude and NCV was observed. The mean ratio of sural NCV to DSN NCV was 1.33 ± 0.19, and the mean ratio of sural nerve SNAP amplitude to DSN SNAP amplitude was 3.17 ± 1.33. Conclusion: These normative data of the DSN might be used as reference values for the study of this very distal peripheral nerve.
Article
Serum monoclonal anti-myelin-associated glycoprotein antibodies may be pathogenic in some people with immunoglobulin M (IgM) paraprotein and demyelinating neuropathy. Immunotherapies aimed at reducing the level of these antibodies might be expected to be beneficial. This is an update of a review first published in 2003 and previously updated in 2006. To assess the effects of immunotherapy for IgM anti-myelin-associated glycoprotein paraprotein-associated demyelinating peripheral neuropathy. We searched the Cochrane Neuromuscular Disease Group Specialized Register 6 June 2011), CENTRAL (2011, Issue 2), MEDLINE (January 1966 to May 2011) and EMBASE (January 1980 to May 2011) for controlled trials. We also checked bibliographies and contacted authors and experts in the field. We included randomised or quasi-randomised controlled trials involving participants of any age treated with any type of immunotherapy for anti-myelin-associated glycoprotein antibody-associated demyelinating peripheral neuropathy with monoclonal gammopathy of undetermined significance and of any severity.Our primary outcome measure was change in the Neuropathy Impairment Scale or Modified Rankin Scale at six months after randomisation. Secondary outcome measures were: Neuropathy Impairment Scale or the Modified Rankin Score at 12 months after randomisation; 10-metre walk time, subjective clinical scores and electrophysiological parameters at six and 12 months after randomisation; IgM paraprotein levels and anti-myelin-associated glycoprotein antibody titres at six months after randomisation; and adverse effects of treatments. The two authors independently selected studies. Two authors independently assessed the risk of bias in included studies. We identified seven eligible trials (182 participants), which tested intravenous immunoglobulin, alfa interferon alfa-2a, plasma exchange, cyclophosphamide and steroids, and rituximab. Only two trials, of intravenous immunoglobulin (with 33 participants, including 20 with antibodies against myelin-associated glycoprotein), had comparable interventions and outcomes, but both were short-term trials.There were no clinical or statistically significant benefits of the treatments used on the outcomes predefined for this review, but not all the predefined outcomes were used in every included trial. Intravenous immunoglobulin showed a statistical benefit in terms of improvement in Modified Rankin Scale at two weeks and 10-metre walk time at four weeks. Cyclophosphamide failed to show any benefit in the trial's primary outcome, and showed a barely significant benefit in the primary outcome specified here, but some toxic adverse events were identified. A trial of rituximab was of poor methodological quality with a high risk of bias and a further larger study is awaited. Serious adverse events were few in the other trials. There is inadequate reliable evidence from trials of immunotherapies in anti-myelin-associated glycoprotein paraproteinaemic neuropathy to form an evidence base supporting any particular immunotherapy treatment. There is very low quality evidence of benefit from rituximab. Large well designed randomised trials of at least six to 12 months duration are required to assess existing or novel therapies, preferably employing unified, consistent, well designed, responsive and valid outcome measures.