Sie sind auf Seite 1von 12

Update on the Clinical Use and

Mechanisms of Action of IVIG


in the Treatment of Autoimmune
Neurological Disorders
By Marinos C. Dalakas, M.D.

Supported by an unrestricted educational grant from Talecris Biotherapeutics

About the Author


Marinos C. Dalakas, M.D., is currently Professor and Chair of Neuro-
muscular Diseases at Imperial College, London, UK; he also maintains
the position of Professor of Neurology at Jefferson Medical College of
Thomas Jefferson University in Pennsylvania. Prior to coming to Jef-
ferson, Dr. Dalakas spent more than 20 years with the Neuromuscular
Diseases Section at the National Institute of Neurological Disorders and
Stroke (NINDS) in Bethesda, Maryland. He joined the NINDS staff in
1981 and became Chief of the Neuromuscular Diseases Section in 1989.
Dr. Dalakas research is focused on exploring the immune, viral,
and genetic basis of various neuromuscular, demyelinating and neu-
ronal disorders affecting muscle, nerves, or brain, and applying new
therapeutic interventions via target-oriented control therapeutic clin-
ical trials. He is the author or coauthor of more than 500 scientific pa-
pers or chapters in peer-reviewed journals. He has also edited 11
books and monographs and currently serves on the editorial board
of several journals, including Brain, Neurology, BMC Neurology, and
Neuromuscular Disorders. Dr. Dalakas also holds a patent on an im-
munotoxin for the treatment of focal movement disorders.
Board certified in neurology and psychiatry, Dr. Dalakas is a Fellow
of the American Academy of Neurology, as well as a member of the
American Neurological Association, the European Neurological Soci-
ety and the World Muscle Society. He is also a Diplomate of the Amer-
ican Board of Psychiatry and Neurology. He received a doctor of
medicine degree in 1972 from the Medical School of the National Uni-
versity of Athens, Greece. He then completed a neurology residency
at the University of Medicine and Dentistry of New Jersey, followed
by a fellowship in neuromuscular diseases and a staff fellowship in
neurovirology and neuroimmunology at NINDS.
Dr. Dalakas is the recipient of a number of honors and awards, in-
cluding the U.S. Public Health Service Special Recognition Award,
NIH Directors Award, and many others.

1
This NHIA monograph article is co-sponsored by the Na- is providing promise in treating various neuroinflammatory or even
tional Community Pharmacists Association (NCPA), neurodegenerative disorders.1-3,5 The success and enthusiasm in
which is accredited by the Accreditation Council for Phar- safely and effectively treating such immunologically diverse disor-
macy Education (ACPE) as a provider of continuing phar- ders, however, has led clinicians to use the drug more liberally even
macy education. NCPA has assigned 2.0 contact hours (0.2 CEUs) of in diseases where the data is weak and not evidence-based, causing
continuing education credits to this article. Eligibility to receive con- logistical problems regarding supply, drug reimbursement and long-
tinuing education credits for this article begins on August 8, 2010 and term safety.6,7 Because all indications for IVIg in neurology (except
expires August 8, 2013. The universal activity number for this program for CIDP for one IVIg product) are still off-label, these issues have
is 207-999-10-045-H01-P. Activity Type: Knowledge-based. generated considerable skepticism and scrutiny from insurance car-
riers, health care organizations, and government agencies.
This continuing nursing education activity was
approved by the Virginia Nurses Association General Issues Regarding Mechanisms of Action of
Continuing Education Approval Committee, IVIg as Relates to Autoimmune Neurologic Disorders
an accredited approver by the American Nurses Credentialing There is overwhelming evidence that IVIg has multiple actions,
Centers Commission on Accreditation. which often operate in concert with each other. For each neuro-
The CNE approval number for this activity is 08-05-102S. Eligi- muscular disorder, however, there appears to be a predominant
bility to receive continuing nursing education credits for this mechanism dictated by the underlying immunopathogenetic
article begins on August 5, 2010 and expires August 31, 2012. cause of the respective disorder.6 Among the main mechanisms
of action of IVIg,8 those relevant to its efficacy in autoimmune
This continuing education article is intended for pharmacists, neuromuscular disorders include the following:
nurses, and other alternate-site infusion professionals. Effect on autoantibodies. The IgG molecules within the IVIg con-
tain antibodies with a wide range of idiotypic and anti-idiotypic
Continuing Education Objectives specificities, which may neutralize pathogenic autoantibodies
After reading this monograph, the participant should be able to: and prevent their interaction with the autoantigen. This effect
List autoimmune neurologic disorders for which IVIG has has been shown experimentally, when extracted F(ab')2 frag-
been found effective ments of IVIg bound to and neutralized known autoantibodies,
Explain the pathophysiology of IVIG in the treatment of such as anti-DNA, anti-AChR, anti-thyroglobulin, anti-GM1 and
autoimmune neuromuscular disorders other.9-12 The idiotypic/anti-idiotypic effect has been also shown
Describe treatment considerations for administration of in patients with acute Guillain-Barre Syndrome (GBS) whose
IgG therapy in autoimmune neurological disorders serum contains various IgG glycolipid antibodies. In an in vitro
nerve-muscle preparation, the F(ab')2 portion of IVIg neutralized
Continuing education credit is free to NHIA members and avail- the blocking effect exerted by the serum of acute GBS patients
able to non-members for a processing fee. To apply for Nursing on the quantal release. 13,14 The effects of IVIg on autoantibodies
or Pharmacy continuing education, go to www.nhia.org/
SeptOct10Supplement and follow the online instructions. may be relevant in explaining the effect of IVIg in antibody-me-
diated autoimmune neuromuscular diseases, such as myasthenia
Introduction gravis (MG), Lambert-Eaton myasthenic syndrome, stiff-person
During the last two decades the use of intravenous immunoglobulin syndrome, and the antibody-mediated demyelinating neu-
(IVIg) has dramatically changed the way we approach the treatment ropathies (GBS, chronic inflammatory polyneuropathy [CIDP],
of autoimmune neuromuscular disorders and dominated the im- and multifocal motor neuropathy [MMN]).
munotherapeutic drugs. Its relative safety for long-term therapy and Inhibition of complement binding and prevention of mem-
undisputable efficacy based on class I evidence derived from ran- branolytic attack complex (MAC) formation. The effect of IVIg
domized trials, have had a major impact in the quality of life of pa- on complement binding has been demonstrated in vitro, in an-
tients with a number of these disorders, some of which were imal models, and in patients who received IVIg. In early stud-
previously unresponsive, or insufficiently responsive, to available im- ies, IVIg was shown to prevent death in guinea pigs from the
munotherapies.1-3 In some disorders, IVIg is as effective as plasma complement-dependent Forssman shock by inhibiting the up-
exchange or steroids but safer and easier to administer; in others, it take of complement C3 and C4 fragments to the endothelial
is superior to all existing drugs; and in still others like Multifocal cells.15 In patients with dermatomyositis, which is a comple-
Motor Neuropathy, it is the only effective therapy.1,3,4 Based on con- ment-dependent microangiopathy mediated by activation of
trolled clinical trials, IVIg has been effective in various acute and C3 and deposition of MAC on the endomysial capillaries, 6, 15-17
chronic demyelinating neuropathies, neuromuscular transmission IVIg is not only clinically effective as discussed later,18 but it
defects, inflammatory myopathies and stiff-person syndrome and also inhibits complement uptake and intercepts the formation
1
and deposition of MAC on the endomysial capillaries19 . Post- is probably pathogenetically relevant. Whether such an effect
IVIg, but not post-placebo serum, inhibits the uptake of C3b is a downstream event related to an upstream effect on the
and C4b fragments by sensitized in vitro targets, probably due primary immune process of the disease, remains unclear.
to formation of covalent or noncovalent complexes between
C3 and specific receptor sites within the infused IgG mole- Status of IVIg in Autoimmune Neuromuscular Dis-
cules.19 Such an inhibition limits the available C3 molecules for orders: Evidence from Controlled Clinical Trials
further incorporation into the C5 convertase assembly, In this section, the clinical efficacy of IVIg based on clinical trials
thereby preventing the formation and in situ deposition of and practical issues unique to each disorder will be discussed.
MAC, as confirmed in the repeated muscle biopsy specimens Guillain-Barre Syndrome (GBS)
of patients with dermatomyositis treated with IVIg 2,6,18-20 The GBS is an acute demyelinating polyneuropathy that peaks within
effect of IVIg on the complement, as proven in dermatomyosi- two weeks of onset and causes severe weakness or paralysis of
tis, is directly relevant to GBS, CIDP and MG, where the com- the limbs and respiratory muscles. Although the target antigen
plement pathway is activated21 and complement fragments is still unknown, humoral and cellular immune mechanisms are
are fixed in the targeted tissues.1,6 Indeed, IVIg has been implicated, as evidenced by activation of complement and dep-
shown to protect the anti-ganglioside antibody-mediated cy- osition of membranolytic attack complex on the myelin sheath,
totoxicity implicated in the pathogenesis of GBS and other au- the presence of circulating antiganglioside or glycolipid antibod-
toimmune neuropathies by displacing complement C3 ies, an increase of T-cell activation products and cytokines, and
fragments fixed on the sciatic nerve. 22 invasion of the myelin sheath by sensitized macrophages.35-39
Modulation or blockade of Fc receptors on Macrophages. The Up to about a decade ago, during the first week of the illness
IgG molecules bind through their Fc region to Fc receptors on the recommended therapy for patients with Guillain-Barr syn-
macrophages and via intracellular signaling mediate inflamma- drome who have severe disease and require assistance to walk
tion or immune effector functions.8,23 The ratio of expression has been plasmapheresis. This practice has been based on con-
of the inhibitory and activation FcR receptors determines the trolled clinical trials which showed that plasmapheresis hastens
final immune response; overexpression of FcR I and FcR III recovery in as many as 52% of patients, compared to 38% of pa-
favor activation, whereas overexpression of FcR II infers inhi- tients receiving sham apheresis.40,41 Since then, this practice has
bition of phagocytosis and interception of antibody-dependent changed based on at least two controlled randomized trials which
cell-mediated cytotoxicity.8,23 IVIg upregulates the inhibitory have shown that IVIg has comparable effects. The first random-
FcR II receptors and modulates the FcR II/ FcR III ratio on ized trial concluded that up to 52.7% of 74 patients receiving IVIg,
macrophages.23 In GBS and CIDP, a blockade of the Fc receptors compared to 34% of 73 patients undergoing plasmapheresis, had
on the macrophages could inhibit the macrophage-mediated functional improvement of one grade or more after four weeks.42
phagocytosis of antigen-bearing target cells, and might inter- This conclusion was strengthened with a second larger trial which
cept the macrophage-mediated demyelination20, 24, 25. An in- compared, in parallel, the efficacy of IVIg therapy alone (5-day reg-
crease in the number of monocytes bearing the FcR II imen of 0.4 gm/Kg/d), plasmapheresis alone, and plasmapheresis
inhibitory receptors and an increase in the FcR II/ FcR III ratio followed by IVIg therapy. After 4 weeks of therapy and 48 weeks
on monocytes has been noted one week after IVIg in GBS and of follow-up, no statistically significant difference was seen be-
CIDP patients who started to improve, suggesting that such in- tween the three treatments, in outcome measures including time
hibitory signaling may be clinically relevant.26 to unaided walking and discontinuation of ventilation.43 The study
Suppression of pathogenic cytokines and other immunoreg- confirmed that although each treatment is beneficial, combining
ulatory molecules. In vitro and in vivo studies have shown that IVIg with PE produces no incremental response. A pilot study of
IVIg causes a dose-dependent downregulation of tissue ex- patients with the GBS, that combined IVIg and 500 mg intra-
pression or reduction in the circulating levels of cytokines and venous methylprednisone showed that the combination was bet-
adhesion molecules, such as ILI, TNF-a, IL1b, TGF-b and TGF-b ter than IVIg therapy alone.44 However a controlled, randomized
mRNA, MHC-I, ICAM-I on the endothelial cells and lymphocyte trial showed no benefit of steroids.45 In atypical Guillain-Barr syn-
function-associated antigen-1 (LFA-1) on activated T-cells.28-31 drome, such as Miller-Fisher variant,46 acute dysautonomia or
The latter has been convincingly shown in the repeated muscle other variants, IVIg appears to be efficacious but controlled stud-
biopsies of patients with dermatomyositis who improved after ies have not been conducted.
IVIg therapy18 and on the lymphocytes of GBS patients one Need for a second IVIg infusion. A common clinical dilemma in
week after infusion with IVIg.26 Because upregulation of cy- managing patients with Guillain-Barr syndrome is whether a
tokines and adhesion molecules is critical in almost all of the second IVIg infusion may add more benefit when three weeks
autoimmune neuromuscular diseases in which IVIg appears to after the first infusion, improvement has either not occurred or
be effective, 4,24 its downregulatory effect on these molecules is deemed inadequate. There is a clinical challenge to try a sec-
2
ond IVIg infusion, but the available data are not adequate to sup- A randomized, controlled, crossover study compared a six-week
port such practice.6 The reported improvement of a few patients course of oral prednisolone (tapered from 60 to 10 mg/d) with a
following a second infusion three to four weeks later,48 requires two-day course of IVIg 1.0 g/kg/d for the treatment of CIDP.62
confirmation with a controlled study to assess whether the re- Treatment was switched after a four-week washout period. Both
ported benefit is due to IVIg rather than due to the natural treatments produced significant improvements in disability after
course of the disease. After almost 10 years of trying to conduct two weeks, although there was slightly more improvement with
such a study, funding became possible only recently and a con- IVIg in the two treatment periods. Also observed were improve-
trolled study is expected to begin in 2010 in the Netherlands. ments favoring IVIg in the time to walk 10 meters after two weeks
Early relapses. In some patients with GBS, early relapse may and disability grade after six weeks. Intravenous immunoglobulin
occur after the initial beneficial response either to plasmaphere- was also equal to plasma exchange (PE) in a single-blind, con-
sis or to IVIg therapy. Based on the two large controlled trials, trolled, crossover study of CIDP patients assigned to a 6-week
it is clear that relapses occur almost equally in patients treated course of PE or IVIg 0.2-0.4 g/kg given weekly.63 Similar improve-
first with plasmapheresis and those treated first with IVIg ther- ments in neurological disability occurred with IVIg compared with
apy.42,43,49 Another retrospective study of 54 patients also con- placebo in a parallel controlled study.64
firmed that there are no increased relapses in patients treated The position of IVIg in the hierarchy of CIDP treatment was in-
with IVIg as opposed to plasmapheresis.50 Changing from one vestigated in a three-year placebo-controlled study of IVIg in
treatment to another is not recommended; instead, staying with treatment-nave patients.65 Patients received an infusion of IVIg
the chosen treatments supplemented with supportive care is 1.0 g/kg/d or placebo on days 1, 2, and 21. Differences in muscle
most appropriate. An associated medical condition appears to strength favoring IVIg were seen as early as day 10, and this
correlate best with an increased risk of relapses, while earlier trend increased over time. At day 42, muscle strength had im-
treatment onset is associated with a lesser chance for relapse.50 proved significantly more with IVIg than with placebo (P = .006),
Markers of response to therapy with IVIg: Recently, the phar- and functional performance was also significantly better (P =
macokinetics of IVIg in relation to clinical outcome, were exam- .019). These findings support IVIg as first-line therapy in the early
ined in 174 patients with GBS treated with IVIg.55 It was found inflammatory phase of CIDP, a strategy that could ameliorate
that the increase in serum IgG (DeltaIgG) two weeks after IVIg the significant axonal degeneration that typically accompanies
treatment varied considerably between patients (mean, 7.8g/L; disease progression.
standard deviation, 5.6g/L). Patients with a low DeltaIgG recov- The IVIg-triggered improvement generally begins after a mean
ered significantly more slowly, and fewer reached the ability to period of nine days and reaches maximum improvement after
walk unaided at six months (log-rank p < 0.001). In multivariate three to four weeks. Repeated treatments every three- to six-
analysis adjusted for other known prognostic factors, a low week intervals, usually with 1g/kg, are needed to maintain re-
DeltaIgG was independently associated with poor outcome (p = sponse. It should be noted that sometimes a number of patients
0.022). It seems that patients with a small increase in serum IgG who initially responded to IVIg may show a less consistent re-
level after IVIg may have a poor outcome; if confirmed, this is sponse in subsequent infusions; such patients should be identi-
important because such patients may benefit from a higher fied early to supplement their therapy with other agents. In
dosage or second course of IVIg early in the disease course. general, CIDP may be more difficult to treat if axonal changes
have become extensive, regardless of the regimen used.
Chronic Inflammatory Demyelinating Polyneuropathy (CIDP) The most important study in the use of IVIg, not only in CIDP
Chronic inflammatory demyelinating polyneuropathy (CIDP) is a dis- but in neurology in general, was the ICE trial.69 This was the
tinct acquired demyelinating polyneuropathy characterized by the largest trial ever conducted involving 117 CIDP patients random-
slow onset (over weeks to months) of weakness, areflexia, and im- ized to IVIg (Gammunex) or Placebo given every three weeks for
paired sensation. The immunopathologic alternations in CIDP, al- 24 weeks (primary end point). Patients who showed an improve-
though fragmentary, are considered similar to Guillain-Barr ment at 24 weeks were re-randomized in a blinded 24-week ex-
syndrome.37,59 Molecular mimicry, antiglycolipid antibodies, T cell sen- tension. The study confirmed that 54% of those receiving
sitization, myelin invasion by activated macrophages and activation Gammunex compared to 21% receiving placebo, improved their
of complement are the main immunopathological features of the dis- disability scores and maintained it up to 24 weeks (p=0,0002).
ease. 37,59-61 However, unlike GBS, which is a monophasic disease, CIDP Results were similar in the extension phase for another 24
requires long-term therapy to maintain improvement. Steroids have weeks. A strong and positive effect on quality of life was also
long been considered the first choice for treating CIDP, hence its des- noted70; some electrophysiologic measurements also im-
ignation as a steroid-responsive neuropathy. Evidence from con- proved.71 The study not only confirmed the short-term efficacy
trolled studies, however, has shown that plasmapheresis and IVIg are of IVIg in CIDP but also established the long-term benefit and
also effective and their efficacy is similar or even superior to steroids.6 safety of maintainance therapy. The study led to the first FDA-
3
approved indication of this IVIg product in CIDP, and the first for much as 1 g/m2 body surface area, may be helpful.88,89 The long-
any neurological disease. term maintenance therapy with IVIg was investigated in 11 MMN
Predictors of response and new molecular markers: An associa- patients, followed for four to eight years.86,87 Patients initially re-
tion analysis with single nucleotide polymorphisms (SNPs) and ceived IVIg 0.4 g/kg/d for 5 days followed by one 0.4-g/kg infusion
haplotype studies was recently performed in 100 Japanese pa- every week for one year and, as needed, in subsequent years with
tients categorized as responders or nonresponders to IVIg ther- a mean dose of 7-48 g/wk. Muscle strength improved significantly
apy.74 Two separate SNPs, corresponding to TAG-1 (transient within three weeks of IVIg treatment but declined slightly and sig-
axonal glycoprotein 1) and CLEC10A (C-type lectin domain family nificantly during the follow-up period. Interestingly, electrophys-
10, member A), showed strong significant differences between iologic changes consistent with improvement (remyelination or
responders and nonresponders. Haplotype analysis of a series of reinnervation) and worsening (demyelination or axon loss) oc-
expanded SNPs, from TAG-1 or CLEC10A, showed that only TAG-1 curred simultaneously in different nerves. Conduction block also
included a significant haplotype within 1 linkage disequilibrium disappeared in some nerve segments but appeared in others.
block, which accommodates IVIg responsiveness. This very inter- These results contrast with those form Vucic et al who were able
esting study is the first to show that TAG-1 is a crucial molecule in- to maintain response when the monthly infusions of IVIg were
volved in IV immunoglobulin responsiveness in Japanese patients kept high up to 2 gram/kg monthly (instead of less than 1 gram/kg
with CIDP and opens the way to explore whether responsiveness as in the Dutch study). Although the optimal maintenance dose
to IVIg is dependent on genetic determinants. remains still empirical, the latter study suggests that IVIg, at the
Another marker recently explored in CIDP was the expression full maintenance dose, has the potential to arrest disease progres-
of Inhibitory FcRIIB on B cells.75 The FcRIIB on B cells trans- sion and prevent axonal degeneration.90 Patients with amy-
duce inhibitory signals on B cells and prevent their transforma- otrophic lateral sclerosis (ALS), which sometimes resembles
tion into IgG-producing plasma cells; as a result, mice lacking MMN, do not respond to IVIg. In a study of nine patients with ALS,
FcRIIB develop autoimmune diseases. Patients with CIDP were IVIg treatment failed to change the course of the disease.91
found to have lower FcRIIB on nave B cells and failed to upreg-
ulate or maintain FcRIIB as the disease progressed.75 Of inter- Paraproteinemic Demyelinating Polyneuropathies
est, FcRIIB protein expression was upregulated on monocytes Demyelinating polyneuropathies associated with IgG or IgA mono-
and B cells after clinically effective IVIg therapy, suggesting that clonal gammopathies respond to IVIg therapy like patients with
the effect of IVIg on FcRIIB may be a factor predicting those CIDP. Patients with IgM monoclonal gammopathy and demyelinat-
patients more likely to respond to IVIg. These results are ar- ing polyneuropathy (IgM-DP), however, form a distinct subset.
guably very preliminary but of potential significance, if confirmed More than 50% of these patients have antibodies against myelin-as-
in a large number of patients studied prospectively. sociated glycoprotein (MAG) and sphingoglycolipids and comprises
a more uniform group, which is clinically characterized by a predom-
Multifocal Motor Neuropathy inantly sensory ataxic or a sensorimotor neuropathy. Because pa-
Multifocal motor neuropathy (MMN) presents with a slow onset tients with anti-MAG neuropathies respond poorly to therapies, a
weakness and muscular atrophy, usually in the distal upper extrem- double-blind controlled study with IVIg was performed.92 The study
ities, with areflexia and preserved sensation. The disease is char- was prompted after the improvement observed in two patients
acterized by conduction block of the motor axons and, in many treated with IVIg in an open-labeled fashion.93
patients, by the presence of antibodies to the GM1 ganglioside. Un- Eleven patients were randomized to IVIg or placebo, given
like CIDP, MMN does not respond to steroids or plasmapheresis. monthly for three months in a double-blind study; after a washout
However, based on controlled trials and several open-label studies, period, they crossed over to the alternate therapy. The trial
MMN responds remarkably well to IVIg therapy.78-86 The improve- showed modest, but not significantly different, benefits. After IVIg,
ment usually begins after seven to 10 days, as in CIDP, and pre- strength improved in only two of 11 patients and declined after
dictably lasts for four to six weeks, at which time a new infusion is placebo; in one other patient, only the sensory scores improved.92
required either with 2 gm/kg or 1 gm/kg. We have noticed that even Antibody titers to MAG/SGPG or gangliosides did not change ap-
chronic cases of MMN with already significant loss of motor axons preciably. A second trial conducted in Europe, also showed modest
can respond to a certain degree. As symptoms diminish, the elec- benefits which were statistically significant only in the secondary
trophysiologic conduction block may resolve.85 end-points.94 Because elevated IgM may cause high serum viscosity
Therapy with IVIg is currently the treatment of choice in MMN. which can be further increased with IVIg as discussed below, cau-
However, some patients may not respond adequately after a pe- tion is needed when IVIg is infused in such patients to avoid trig-
riod of time, probably because of further progression of the un- gering thromboembolic events. Because the disease responds to
derlying disease and axonal degeneration.86,87 In these Rituximab, based on a controlled study89, Rituximab is becoming
circumstances, Rituximab or intravenous cyclophosphamide, as the treatment of choice for the IgM-anti-MAG neuropathy.
4
Myasthenia Gravis the 3.5 units cited as clinically significant and used to calculate
Myasthenia gravis (MG) is characterized by fluctuating weakness the sample size. Third, it was statistically significant (by more
or fatigability of the extraocular, bulbar, respiratory, and limb than 4.1 points) only in a small number of patients with severe
muscles. Diplopia, dysphagia, and dysathria are common. Myas- MG, suggesting that the study lost power because of including
thenia is the prototypic autoimmune disease mediated by path- patients with less severe disease. Fourth, the definition of wors-
ogenic autoantibodies against the acetylcholine receptor (AChR) ening weakness was rather subjective.
and possibly against muscle specific kinase (MuSK).96,97 Two additional randomized controlled studies 104,105 have ad-
Patients with MG respond fairly well to the available therapies, dressed the efficacy of IVIg for the treatment of moderate to se-
such as anticholinesterases, thymectomy, steroids, azathioprine, vere but stable MG, one comparing IVIg to plasma exchange and
cyclosporine and plasmapheresis. The need for another effec- the other to placebo. None showed a significant difference
tive immunomodulating therapy without long-term side effects From these data, along with other published series and our own
has prompted experimentation with high-dose IVIg. observations, several conclusions can be drawn, as recently dis-
As reviewed previously,98,99 IVIg, is promising for disease ex- cussed98,99. First, the majority of patients with MG exacerbations
acerbations, but the evidence is weak regarding the chronic can respond to IVIg therapy. The improvement is seen early, begin-
management of the disease or as a steroid-sparing agent. 98,100 ning after a mean period of three to 10 days lasting for a mean pe-
The first randomized study compared the efficacy and tolerance riod of 30 days. Second, IVIg appears to be as effective as
of two doses of IVIg to plasma exchange in MG exacerbations101. plasmapheresis, but for myasthenic crises plasmapheresis is supe-
A total of 87 patients were randomized to receive either three rior. Third, the role of IVIg in the chronic management of MG has
courses of PE or IVIg 0.4 g/kg/d for three to five days. The study not yet been established. The numbers of confounding factors in-
demonstrated similar efficacy, measured by variation in myas- volved in deciding the best therapy of MG at a given state of the
thenic muscular score, in both the PE and IVIg groups; the anti- disease demand carefully designed studies, clear objectives, accu-
AChR antibody titers fell by about two thirds in both groups rate assessment of efficacy, and precise control of the concomitant
among. Interestingly, the three-day IVIg regimen was slightly su- immunosuppressive drugs. At present and until further controlled
perior to the five-day regimen, although methodologic deficien- trials are conducted, IVIg may be justified in lieu of plasmapheresis:
cies limit a rigorous comparison. A second large controlled study a) for acutely worsening disease to prevent or minimize impending
conducted also by the same group, randomized 173 patients to bulbar or respiratory failure; b) prepare a weak patient for thymec-
1g/kg IVIg for one day or to 2gm/kg for two days.102 At day 15, tomy and c) as adjuvant to immunosuppressive therapies and peri-
the myasthenic scores improved equally in both groups conclud- odically (every one to three months) to minimize the long-term side
ing that IVIg is efficacious in MG but that there is no difference effects or stabilize a patient until immunosuppressants, such as aza-
between 2 g/kg versus 1.0 g/kg. thioprine or cyclosporin, become effective.2
A recent trial compared IVIg to placebo in MG patients who
had worsening weakness defined as increasing symptoms or Lambert-Eaton Myasthenic Syndrome
signs severe enough (as judged by both the patient and the Patients with LEMS have a presynaptic neuromuscular junction
physician) to warrant a change in therapy103. Fifty-one AChR-pos- defect that results in proximal muscle weakness, oculomotor
itive patients were randomized to receive either IVIg 2g/kg or signs and autonomic dysfunction. The symptoms are caused by
the equivalent volume of dextrose 5% over two days. The main antibodies against presynaptic voltage-gated calcium channels
endpoint was the change in Quantitative MG scores (QMGS) resulting in decreased acetylcholine release. Patients respond
from baseline (day 0) to day 14. On day 14, the mean change in to diaminopyridine, steroids and azathioprine. Even though IVIg
QMGS was -2.5 in the IVIg group and -0.9 in the placebo group is rarely needed, a controlled study has been conducted compar-
(p = 0.047). On day 28, these values were -3 in the IVIg group and ing IVIg to placebo. The IVIg-randomized patients showed a sta-
-1.2 in the placebo group (p = 0.055). For the mild MG cases, the tistically significant increase in muscle strength compared to
mean change in QMGS on day 14 was similar in the two groups: placebo, which peaked at two to four weeks and declined by
-0.7 in the IVIg group and -1.1 in the placebo group. The only sig- eight weeks. The effectiveness of IVIg was associated with a sta-
nificant difference was noted for the moderate to severe MG tistically significant reduction of antibodies against voltage-
group where these values were -4.1 in the IVIg group and -0.7 in gated calcium channel.106 IVIg is useful in difficult cases of LEMS,
the placebo group (p = 0.01); that treatment effect was main- especially when steroids and azathioprine are not very effective
tained at day 28. This study, has several limitations. First, the ef- or cause significant side effects.
fect was modest, as patients improved by 2,54 QMG units
compared to 0,89 in the placebo; considering that individual Inflammatory Myopathies
QMG scores usually vary up to 2.6 units, this effect might not The main subsets in this group include: Dermatomyositis,
have been clinically meaningful. Second, the effect did not reach Polymyositis and Inclusion Body Myositis.
5
Dermatomyositis is an acquired myopathy causing proximal patients with IBM in a study designed similarly to the previously
muscle weakness and a violaceous rash on the face and extrem- mentioned dermatomyositis trial.107,111 In the IVIg-treated group,
ities. Early deposition of membranolytic attack complex (MAC) there was an increase in muscle scores compared to placebo,
on the endomysial capillaries leads to capillary destruction, mus- but the differences were not statistically significant, perhaps be-
cle ischemia, and inflammation.16,17 The disease responds to cause the study lacked sufficient power.111 Nonetheless, signifi-
steroids but often becomes steroid-resistant. Azathioprine, cant regional differences occurred in IVIg-treated patients,
methotrexate, or cyclosporine offer modest benefit but rarely especially in the swallowing function which improved signifi-
cause a remission. Administration of IVIg to patients with der- cantly in patients receiving IVIg compared with those receiving
matomyositis has proven effective. In a double-blind, placebo- placebo (P < .05). The mild benefits noted in certain muscle
controlled study, 15 patients with treatment-resistant groups, prompted a larger study aimed to investigate the poten-
dermatomyositis received IVIg 2.0 g/kg or placebo once a month tial synergistic effect of IVIg with prednisone.112 Thirty-six pa-
for three months, with the option of crossing over to the alter- tients were randomized to IVIg 2.0 g/kg or placebo once a month
native therapy for three more months.18 At the end of the first for three months. Before infusions, all patients received pred-
three-month treatment phase, IVIg-treated patients experienced nisone (tapered from 60 mg/d). After 3 months of treatment,
a significant improvement in MRC (Medical Research Council) there was no significant difference in muscle strength score, be-
scores (P < .018) and neuromuscular symptoms (P < .035) com- tween the IVIg-plus-prednisone group and the placebo-plus-
pared with those receiving placebo. Marked improvements were prednisone group. A third trial showed similar results.113 In spite
also noted in the active violaceous rash or the chronic scaly erup- of these negative trials, a few patients may show transient signs
tions.18 In subsequent open-label infusions, the benefit of IVIg of improvement which, although minor and difficult to capture
has been documented in a large number of patients treated by with the methods used, can be at times clinically significant for
us or under our supervision, and by several investigators the patients activities and lifestyles, at least for a period of time.
throughout the world.107,108 Repeat open muscle biopsies on pa- When life-threatening dysphagia is apparent, IVIg is a treatment
tients who clinically improved have shown marked improvement option in view of the positive results noted above.114
in the muscle cytoarchitecture including muscle fiber diameter,
revascularization with increased number of capillaries per fiber, Stiff Person Syndrome (SPS)
and reduction of inflammation and connective tissue.18 Im- Stiff Person Syndrome (SPS) is a disabling central nervous sys-
munopathologically, it was shown that IVIg inhibits the deposi- tem disorder characterized by muscle rigidity, episodic muscle
tion of MAC on the endomysial capillaries by intercepting the spasms, and high titers of antibodies against glutamic acid de-
incorporation of C3 into the C5 convertase assembly, downreg- carboxylase (GAD65), the rate-limiting enzyme for synthesis of
ulates the expression of the intercellular adhesion molecule gamma-aminobutyric acid (GABA). Various other autoantibodies
(ICAM-I) on the endomysial capillaries and the major histocom- (e.g., anti-amphiphysin) are also present.115
patibility complex class I (MHC-I) antigen on muscle fibers, and Drugs that enhance GABA neurotransmission, such as di-
reduces the expression of TGF-b1 protein and mRNA in connec- azepam, provide only mild to modest relief of clinical symptoms115;
tive tissue. 19,107,18,28. IVIg also modified certain immunoregulatory however, treatment with IVIg confers substantial benefit. This im-
and structural genes in the muscles of DM patients who re- provement was documented in a placebo-controlled crossover
sponded to IVIg therapy, as determined by gene array studies109. study in 16 patients administered IVIg 2.0 g/kg or placebo once a
Although a controlled study was never performed, IVIg is also month for three months.112 Efficacy was assessed with the use of
effective in patients with polymyositis. Because both polymyosi- an objective distribution-of-stiffness index and heightened-sensi-
tis and dermatomyositis initially responds to steroids, IVIg ther- tivity scale. Among patients initially treated with IVIg, stiffness
apy is best reserved as second line add-on therapy in patients scores decreased significantly (P = .02) and heightened-sensitivity
who are not adequately controlled with combination of steroids scores declined markedly, but they rebounded during placebo ad-
and methotrexate or azathioprine, and for patients who are im- ministration; the opposite pattern occurred among those treated
munodeficient or in whom steroids are contraindicated.110 with placebo first. Eleven patients who received IVIg were able to
Inclusion-body myositis (IBM) is the most common ac- walk more easily or without assistance and to perform work-re-
quired inflammatory myopathy in patients above the age of 50 lated or household tasks; their frequency of falls decreased. The
years. Immunopathologically, IBM is identical to polymyositis, study convincingly showed that IVIg is effective as supplementary
characterized by sensitized, antigen-driven cytotoxic T cells that therapy in patients with SPS.110
invade MHC-I expressing muscle fibers.16,17 What makes IBM
unique however is the presence of vacuolated fibers which con- Treatment Considerations
tain amyloid deposits. The disease is resistant to most immuno- The therapeutic dose of IVIg is empirically set at 2 g/kg. Although
suppressive medications. The efficacy of IVIg was tested in 19 past practice has been to divide the total dose for infusion into
6
five daily doses of 400 mg/kg each, the preference now is to di- noted in children with Guillain-Barr syndrome.56 The new ICE
vide the total dose into two daily doses of 1 g/kg each especially trial in CIDP was also conducted with a two-day infusion with ex-
in younger patients who have normal renal and cardiovascular cellent safety and tolerance profile.
function.1 In our experience, the two-day infusion is not associ- In general, adverse reactions to IVIg therapy are usually minor
ated with more adverse reactions than the five-day infusion, and and occur in no more than 10% of the patients with neurological
it is preferable except for patients with major cardiovascular disorders (see the box below for a brief list). Mild to moderate
risks. In children with Kawasaki syndrome, a single, large, 2 g/kg headache, which responds to nonsteroidal anti-inflammatory
dose of IVIg given in a 10-hour infusion was more effective than medications, is common. Chills, myalgia, or chest discomfort
four daily infusions of 400 mg/kg each.124 Similar experience was may develop in the first hour of the infusion and usually re-

Adverse Reactions to IVIg Therapy


Serum Viscosity and Thromboembolic Events. IVIg therapy deficiency is common (prevalence about 1:1000), but asympto-
causes an increase in serum viscosity, up to 0.5 centipoise, and matic. About 29% of these individuals have anti-IgA antibodies,
in patients with high-normal or slightly elevated serum viscos- which do not necessarily predict the development of adverse re-
ity, such as those with cryoglobulinemia, hypercholesterolemia, actions.138 Theoretically, when these patients receive IVIg, the
or hypergammaglobulinemia, the viscosity level increases even small amount of IgA present with the IVIg may lead to anaphy-
further.126 Serum viscosity greater than 2.5 cp (normal, 1.2-1.8 lactic reaction due to formation of macromolecular complexes
cp) increases the risk of thromboembolic events and may be between the infused IgA and circulating anti-IgA antibodies. The
one of the factors responsible for rare cases of stroke, pul- reaction is rare and we have not seen it, nor has it been reported
monary embolism or myocardial infarction noted after IVIg to our knowledge, in neurological patients; it may occur in pa-
treatment.127-131,126,132 Patients with recent deep vein thrombosis, tients with common variable immunodeficiency.139
or immobilized patients who are at higher risk of having a sub- Renal Tubular Necrosis. Acute renal tubular necrosis, mostly
clinical thrombosis, may be more prone for developing a throm- reversible, occurs rarely with IVIg therapy in patients who have
boembolic event after IVIg. In such patients, prudence in pre-existing kidney disease and volume depletion, especially
justifying the use of IVIg, screening the legs with an ultrasound the elderly and those with diabetes or poor hydration.140 It is
for subclinical clots, and very slow infusion rate are recom- usually reversible, but rare fatalities have been noted.141,142
mended. Whether low-dose heparin or anti-platelet agents can Serum creatinine may rise one to 10 days after the infusion but
prevent thromboembolic events in such patients is uncertain. returns to baseline within two to 60 days of discontinuation.142
A reversible cerebral vasospasm has also been noted.133 This complication has often, but not exclusively, been associ-
Migraine Headache. In patients with a history of migraine, ated with the high concentration of sucrose in some propri-
IVIg therapy may trigger a migraine attack, which can be pre- etary IVIg products.141 Osmotically induced tubular injury and
vented by pretreatment with propranolol.134 The occurrence vacuolization are the common histopathological findings on
of aseptic meningitis, discussed below, is also high in migrain- renal biopsy.142 Identical osmotic tubular nephrosis is caused
ous patients.135 IVIg therapy has been associated with a stroke by intravenous solutions containing a concentration of hyper-
in a young woman with a history of migraine.130 tonic sucrose similar to that of IVIg.141 Diluting the IVIg prepa-
Aseptic Meningitis. Aseptic meningitis develops in some pa- ration, slowing the rate of infusion or selecting products with
tients treated with IVIg. Its occurrence is unrelated to the pro- low osmolality minimizes the risk. In patients with pre-existing
prietary product, the rate of infusion, or the underlying kidney disease, if there is no alternate therapy to IVIg treat-
disease.135-137 Prophylaxis with intravenous steroids is often in- ment, close monitoring of creatinine and BUN are essential.
effective. The symptoms respond to strong analgesia and Spurious Results on Serological Tests. After IVIg therapy, the
subside in 24 to 48 hours. Additional diagnostic testing is erythrocyte sedimentation rate increases sixfold or more,143
rarely necessary.136,137 probably as the result of enhanced rouleux formation and re-
Skin Reactions. Skin reactions to IVIg therapy, although rare, can duced surface area caused by the infused gamma globulin.1
develop two to five days after the infusions and may last up to This increase can persist for two to three weeks, and should
30 days. They include urticaria, lichenoid cutaneous lesions, pru- not be considered a sign of a developing vasculitis. We have
ritus of the palms, and petechiae of the extremities.1,136, Skin re- also observed hyponatremia, with sodium concentrations as
actions, associated with various lots of IVIg, have occurred in low as 130 mg/l (normal, 135-145 mg/l), after IVIg therapy, but
seven of the 120 patients we had initially treated.1,136, not after placebo use.143 Hyponatremia appears to be related
Severe Anaphylactic Reaction. A severe anaphylactic reaction to the assay method used to measure Na+ because additional
has been reported in patients with an absence or severe defi- dilution of the sample is required owing to the high serum
ciency of IgA who also have anti-IgA antibodies. Selective IgA protein concentration that follows IVIg infusion.
7
sponds to stopping the infusion for 30 minutes and resuming it 17. Dalakas MC, Hohlfeld R. Polymyositis and dermatomyositis. Lancet. 2003; 362,
at a slower rate. Post-infusion fatigue, fever, or nausea may 971-982.
occur and last for up to 24 hours. The cause of these reactions 18. Dalakas MC, Illa I, Dambrosia JM, et al. A controlled trial of high-dose intra-
is unclear, but activation of complement by aggregated im- venous immunoglobulin infusions as treatment for dermatomyositis. N Engl J
munoglobulin molecules or various stabilizing agents in the IVIg Med. 1993; 329, 1993-2000
preparation have been implicated.1,125 A slow rate of infusion is 19. Basta M, Dalakas MC. High-dose intravenous immunoglobulin exerts its bene-
advisable in patients with a compromised cardiovascular system ficial effect in patients with dermatomyositis by blocking endomysial deposition
or congestive heart failure to avoid rapid fluid overload. of activated complement fragments. J Clin Invest.1994; 94, 1729-35.
20. Dalakas MC. Mechanism of action of intravenous immunoglobulin and thera-
References peutic considerations in the Treatment of autoimmune neurologic diseases.
1. Dalakas MC. Intravenous immunoglobulin in the treatment of autoimmune neu- Neurology. 1998; 51(Suppl), S2-S8.
romuscular diseases: present status and practical therapeutic guidelines. Muscle 21. Basta M, Illa I, Dalakas MC. Increased in vitro uptake of the complement C3b in
Nerve.1999; 22, 1479-1497. the serum of patients with Guillain Barre syndrome, myasthenia gravis and der-
2. Dalakas MC. Intravenous immunoglobulin in autoimmune neuromuscular dis- matomyositis. J Neuroimmunol. 1996; 71, 227-229.
eases. JAMA. 2004;291:2367-2375 22. Zhang G, Lopez PH, Li CY, Mehta NR, Griffin JW, Schnaar RL, Sheikh KA. Anti-
3. Dalakas MC, Spath PJ (Eds) Intravenous Immunoglobulins in the Third Millen- ganglioside antibody-mediated neuronal cytotoxicity and its protection by in-
nium. Lancaster, UK. Parthenon Publishing. 2004 travenous immunoglobulin: implications for immune neuropathies. Brain 2004
4. Stangel M, Hartung H-P, Marx P, Gold R. Intravenous immunoglobulin treat- May;127:1085-100.
ment of neurological autoimmune diseases. J Neurol Sci.1998; 153, 203-214 23. Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG medi-
5. Dalakas MC. Clinical use of IVIg in Neurology. In Lazarus AH, Semple JW eds Im- ated through the inhibitory Fc receptor. Science.2001; 291, 484-486.
munoglobulin Therapy. Bethesda MD AABB Press, In press 2010 24. Dalakas MC. Intravenous immunoglobulin therapy for neurological diseases.
6. Dalakas MC. The use of intravenous immunoglobulin in the treatment of au- Ann Intern Med. 1997; 126, 721-730.
toimmune Neurological Disorders: Evidence-based indications and safety pro- 25. Dalakas MC. Mechanisms of action of IVIg and therapeutic considerations in
file. Pharmacol Therapeutics 2004;102:177-193. the treatment of acute and chronic demyelinating neuropathies. Neurol-
7. Gold R, Stangel M, Dalakas MC. Drug Insight: Use of intravenous immunoglob- ogy.2002; 59(Suppl 6), S13-21.
ulin in neurology: Practical issues and therapeutic considerations. Nature Clin 26. Creange A, Gregson NA, Hughes RA. Intravenous immunoglobulin modulates
Pract Neurol. 2007; 3: 36-44 lymphocyte CD54 and monocyte FCgammaRII expression in patients with
8. Kazatchkine MD, Kaveri SV. Immunomodulation of autoimmune and inflam- chronic inflammatory neuropathies. J Neuroimmunol. 2003; 135, 91-95.
matory diseases with intravenous immune globulin. N Engl J Med. 2001; 345, 27. Svenson M, Hansen MB, Bendtzen K. Binding of cytokines to pharmaceutically
747-755. prepared human immunoglobulin. J Clin Invest.1993; 92, 2533-9.
9. Dietrich G, Kazatchkine MD. Normal immunoglobulin G (IgG) for therapeutic 28. Amemiya K, Semino-Mora C, Granger RP, Dalakas MC. .Downregulation of TGF-
use (intravenous Ig) contains anti-idiotypic specificities against an immunodom- b1 mRNA and protein in the muscles of patients with inflammatory myopathies
inant, disease-associated, cross-reactive idiotype of human anti-thyroglobulin after treatment with high-dose intravenous immunoglobulin. Clin Im-
autoantibodies. J Clin Invest. 1990; 85, 620-9. munol.2000; 94, 99-104.
10. Kaveri S, Prasad N, Vassilev T, et al. Modulation of autoimmune responses by 29. Rigal D, Vermot-Desroches C, Heitz S, et al. Effect of IVIg in peripheral blood B, NK,
intravenous immunoglobulin. Mult Scler.1997; 3, 121-128. and T cell subpopulations in women with recurrent spontaneous abortions: specific
11. Kazatchkine MD, Dietrich G, Hurez, V,et al.V region-mediated selection of autore- effect on LFA-1 and CD56 molecules. Clin Immunol Immunopathol. 1994; 71, 309-14.
active repertoires by intravenous immunoglobulin. Immunol Rev .1994;139, 79-107. 30. Abe Y, Horiuchi A, Miyake M, Kimura S. Anti-cytokine nature of natural human
12. Malik U. Oleksowicz L, Latov N, Cardo LJ. Intravenous y-globulin inhibits binding immunoglobulin: one possible mechanism of the clinical effect of intravenous
of anti-GMI to its target antigen. Ann Neurol. 1996; 39,136-139. immunoglobulin therapy. Immunol Rev . 1994;139, 5-19.
13. Buchwald B, Ahangari R, Weishaupt A, Toyka KV. Intravenous immunoglobulins 31. Ankrust P, Muller F, Svenson M, et al. Administration of intravenous im-
neutralize blocking antibodies in Guillain-Barr syndrome. Ann Neurol. 2002; 51, munoglobulin (IVIg) in vivo downregulatory effects on the IL-1 system. Clin Exp
673-680. Immuno.1999;l 115, 136-143.
14. Dalakas MC. Blockade of blocking antibodies in Guillain-Barr syndromes: un- 32. Gabriel CM, Gregson NA, Redford EJ, et al. Human immunoglobulin ameliorates
blocking the mystery of action of intravenous immunoglobulin. Ann Neurol. rat experimental allergic neuritis. Brain. 1997; 120, 1533-1540.
2002;51, 667-669. 33. van Engelson BGM, Miller DJ, Pavelko KD, et al. romotion of remyelination by
15. Basta M, Kirshbom P, Frank MM, Fries LF. Mechanisms of therapeutic effect polyclonal immunoglobulin in Theilers virus-induced demyelination and in mul-
of high-dose intravenous immunoglobulin. J Clin Invest.1989; 84, 1974-1981. tiple sclerosis. J Neurol Neuros Psych.1994; (Suppl) 57 Suppl, 65-68.
16. Dalakas MC. Polymyositis, dermatomyositis and inclusion-body myositis. N Engl 34. Sekul EA, Cupler EJ, Dalakas MC. Aseptic meningitis associated with high-dose
J Med. 1991; 325, 1487-1498. intravenous immunoglobulin therapy: frequency and risk factors. Ann Intern
Med.1994;121, 259-62.

8
35. Hahn AF. Guillain-Barr syndrome. Lancet. 1998; 352, 635-641. 55. Kuitwaard K, de Gelder J, Tio-Gillen AP,et al. Pharmacokinetics of intravenous
36. Hartung HP, Pollard JD, Harvey GK, Toyka KV. Immunopathogenesis and treat- immunoglobulin and outcome in Guillain-Barr syndrome. Ann Neurol.
ment of the Guillain-Barr syndrome Part II. Muscle Nerve.1995; 18, 154-164. 2009;66(5):597-603.
37. Gold R, Dalakas MC, Toyka KV. Immunotherapy in autoimmune neuromuscular 56. Kanra G, Uzon A, Vajsar J, et al. Intravenous immunoglobulin treatment in chil-
disorders. Lancet Neurology.2003; 2, 22-32. dren with Guillain-Barr syndrome. Europ J of Ped Neurol.1997; 1, 7-12.
38. Hafer-Macko C, Hsieh ST, Li CY, et al. Acute motor axonal neuropathy: an anti- 57. Reisin RC, Pociecha J, Rodriguez E, et al. Severe Guillain-Barr syndrome in child-
body-mediated attack on axolemma. Ann Neurol. 1996; 40, 635-644. hood treated with human immune globulin. Pediatr Neurol.1996; 14, 308-312.
39. Hafer-Macko CE, Sheikh KA, Li CY, et al. Immune attack on the Schwann cell surface 58. Kornberg AJ. Clinical experience with intravenous immunoglobulin for treat-
in acute inflammatory demyelinating polyneuropathy. Ann Neurol.1996; 39, 625-635. ment of pediatric Guillain-Barr syndrome and chronic inflammatory demyeli-
40. French Cooperative Group on Plasma Exchange in Guillain-Barr Syndrome. Ef- nating polyradiculoneuropathy. Dalakas MC, Spath PJ (eds). Intravenous
ficiency of plasma exchange in Guillain-Barr syndrome: role of replacement Immunoglobulin in the third millennium. UK. Parthenon Publishers. 2004.
fluids. Ann Neurol .1987; 22, 753-761. 59. Kieseier BC, Dalakas MC, Hartung HP. Immune mechanisms in chronic inflam-
41. Guillain-Barr Study Group. Plasmapheresis and acute Guillain-Barr syndrome. matory demyelinating neuropathy. Neurology .2002; 59(Suppl 6), S7-12.
Neurology.1985; 35, 1096-1104. 60. Yuki N. . Infectious origins of, and molecular mimicry in, Guillain-Barr and Fisher
42. van der Meche FGA, Schmitz PIM, and the Dutch Guillain-Barr Study Group. A syndromes. Lancet.2001; Infect Dis 1, 29-37.
randomized trial comparing intravenous immune globulin and plasma exchange 61. Weiss MD, Luciano CA, Semino-Mora C, et al. Molecular mimicry in chronic in-
in Guillain-Barr syndrome. N Engl J Med.1992; 326, 1123-1129. flammatory demyelinating polyneuropathy and melanoma. Neurology.1998;
43. Plasma Exchange/Sandoglobulin Guillain-Barr Syndrome Trial Group. Random- 51:1738-1741.
ized trial of plasma exchange, intravenous immunoglobulin, and plasma ex- 62. Hughes R, Bensa S, Willison H, et al. Randomized controlled trial of intravenous
change followed by intravenous immunoglobulin in Guillain-Barr syndrome. immunoglobulin versus oral prednisolone in chronic inflammatory demyelinat-
Lancet.1997; 349, 225-230. ing polyradiculoneuropathy. Ann Neurol.2001;50, 195-201.
44. Dutch Guillain-Barr Syndrome Study Group. Treatment of Guillain-Barr syn- 63. Dyck PJ, Litchy WJ, Kratz KM, et al. A plasma exchange versus immune globulin
drome with high-dose immune globulins combined with methylprednisolone: infusion trial in chronic inflammatory demyelinating polyradiculoneuropathy.
a pilot study. Ann Neurol 1994; 35, 749-752. Ann Neurol.1994; 36, 838-845.
45. van Koningsveld R, Schmitz PIM, van der Mech FG, et al.. Effect of methylpred- 64. Hahn AF, Bolton CF, Zochodne D, Feasby TE. Intravenous immunoglobulin
nisolone when added to standard treatment with intravenous immunoglobulin treatment in chronic inflammatory demyelinating polyneuropathy. A double-
for Guillain-Barr syndrome: randomized trial. Lancet.2004; 363, 192-196. blind, placebo-controlled, cross-over study. Brain.1996; 119, 1067-1077.
46. Arakawa Y, Yoshimura M, Kobayashi S, Ichihashi K. The use of intravenous im- 65. Mendell JR, Barohn RJ, Freimer ML, et al. Randomized controlled trial of IVIg
munoglobulin in Miller-Fisher syndrome. Brain Dev. 1993; 15, 231-233. in untreated chronic inflammatory demyelinating polyradiculoneuropathy. Neu-
47. Raphael JC, Chevret S, Harboun M, Jars-Guincestre MC. Intravenous immune glob- rology.2001; 56, 445-449.
ulins in patients with Guillain-Barr syndrome and contraindications to plasma ex- 66. Dalakas MC. . Advances in chronic inflammatory demyelinating polyneuropathy:
change: 3 days versus 6 days. J Neurol Neurosurg Psychiatry.2001; 71:235-238. disease variants and inflammatory response mediators and modifiers. Curr Opin
48. Farcas P, Avnun L, Frisher S,et al. . Efficacy of repeated intravenous immunoglob- Neurol.1999; 12, 403-409.
ulin in severe unresponsive Guillain-Barr syndrome. Lancet.1997; 350, 1747. 67. Saperstein DS, Katz JS, Amato AA, Barohn RJ. Clinical spectrum of chronic ac-
49. Visser LH, van der Meche FG, Meulstee J, van Doorn PA. Risk factors for treat- quired demyelinating polyneuropathies. Muscle Nerve.2001; 24, 311-324.
ment related clinical fluctuations in Guillain-Barr syndrome. Dutch Guillain- 68. Sharma KR, Cross J, Farronay O,et al. Demyelinating neuropathy in diabetes
Barr study group. J Neurol Neurosurg Psychiatry.1998; 64, 242-244. mellitus. Arch Neurol.2002; 59, 758-765.
50. Romano JG, Rotta FT, Potter P,et al. Relapses in the Guillain-Barr syndrome 69. Hughes RA, Donofrio P, Bril V, et al. Intravenous immune globulin (10% caprylate-
after treatment with intravenous immune globulin or plasma exchange. Muscle chromatography purified) for the treatment of chronic inflammatory demyelinat-
Nerve.1998; 21, 1327-1330. ing polyradiculoneuropathy (ICE study): a randomised placebo-controlled trial.
51. Ho TW, Hsieh ST, Nachamkin I, et al. Motor nerve terminal degeneration pro- Lancet Neurol. 2008;7(2):136-44. Erratum in: Lancet Neurol. 2008;7(9):771.
vides a potential mechanism for rapid recovery in acute motor axonal neuropa- 70. Merkies IS, Bril V, Dalakas MC, et al. Health-related quality-of-life improvements in
thy after Campylobacter infection. Neurology.1997; 48, 695-700. CIDP with immune globulin IV 10%: the ICE Study. Neurology. 2009;72(15):1337-44.
52. Kuwabara S, Asahina M, Koga M, et al. Two patterns of clinical recovery in Guil- 71. Bril V, Katzberg H, Donofrio P, et al. Electrophysiology in chronic inflammatory
lain-Barre syndrome with IgG anti-GM1 antibody. Neurology .1998;51, 1656-1660. demyelinating polyneuropathy with IGIV. Muscle Nerve. 2009;39(4):448-55.
53. Rees JH, Gregson NA, Hughes RAC. Antiganglioside antibodies in Guillain Barr 72. McCrone P, Chisholm D, Knapp M, et al. Cost-utility Analysis of Intravenous Im-
syndrome and their relationship to Campylobacter jejuni infection. Ann Neurol munoglobulin and Polyradiculoneuropathies (CIDP). Eur J Neurol.2003; 10, 687-694.
1995; 38, 809-816. 73. Vermeulen M, van Doorn PA, Brand A, et al. Intravenous immunoglobulin treat-
54. van Koningsveld R, Steyerberg EW, Hughes RA, Swan AV, van Doorn PA, Jacobs BC. ment in patients with chronic inflammatory demyelinating polyneuropathy: a
A clinical prognostic scoring system for Guillain-Barr syndrome. Lancet Neurol. 2007 double blind, placebo controlled study. J Neurol Neurosurg Psychiatry. 1993;56,
Jul;6(7):589-94. 36-39.

9
74. Iijima M, Tomita M, Morozumi S, et al. Single nucleotide polymorphism of TAG- 92. Dalakas MC, Quarles RH, Farrer RG, et al. A controlled study of intravenous im-
1 influences IVIg responsiveness of Japanese patients with CIDP. Neurology. munoglobulin in demyelinating neuropathy with IgM gammopathy. Ann Neurol.
2009;73(17):1348-52. 1996; 40, 792-795.
75. Tackenberg B, Jelcic I, Baerenwaldt A, et al. Impaired inhibitory Fcgamma re- 93. Cook D, Dalakas MC, Galdi A, et al. High-dose intravenous immunoglobulin in
ceptor IIB expression on B cells in chronic inflammatory demyelinating polyneu- the treatment of demyelinating neuropathy associated with monoclonal gam-
ropathy. Proc Natl Acad Sci U S A. 2009;106(12):4788-92. mopathy. Neurology .1990;40, 212-214.
76. Nevo V, Pestronk A, Kornberg AJ, et al. Childhood chronic inflammatory de- 94. Comi G, Roveri L, Swan A, et al. A randomized controlled trial of intravenous
myelinating neuropathies: clinical course and long-term follow-up. Neurology. immunoglobulin in IgM paraprotein associated demyelinating neuropathy. J
1996; 47, 98-102. Neurol.2002; 249, 1370-1377.
77. Simmons Z, Wald JJ, Albers JW. Chronic inflammatory demyelinating 95. Hughes RA, Dalakas MC, Cornblath DR,et al. Clinical applications of intravenous
polyradiculoneuropathy in children: II. Long-term follow-up, with comparison immunoglobulins in neurology. Clin Exp Immunol. 2009;158 Suppl 1:34-42
to adults. Muscle Nerve. 1997; 20, 1569-1575. 96. Drachman DB. Myasthenia gravis. N Engl J Med.1994; 330, 1797-1810.
78. Azulay JP, Blin O, Pouget J, et al. Intravenous immunoglobulin treatment in pa- 97. Hoch W, McConville J, Helms S, et al. Auto-antibodies to the receptor tyrosine
tients with motor neuron syndromes associated with anti-GM1 antibodies. Neu- kinase MuSK in patients with myasthenia gravis without acetylcholine receptor
rology.1994; 44, 429-432. antibodies. Nat Med.2001; 7, 365-368.
79. Bouche P, Moulonguet A, Ben Younes-Chennoufi A, et al. Multifocal motor neu- 98. Dalakas MC. Experience with IVIg in the treatment of patients with myasthenia
ropathy with conduction block: a study of 24 patients. J Neurol Neurosurg Psy- gravis. Neurolog.1997;y 48S, 64-69.
chiatry. 1995; 59, 38-44. 99. Dalakas MC: IVIg Therapy in Myasthenia Gravis and Mechanism of Immunosup-
80. Chaudhry V, Corse AM, Cornblath DR,et al. Multifocal motor neuropathy: re- pression, In Christados P (Editor) Myasthenia Gravis. Disease Mechanisms and
sponse to human immune globulin. Ann Neurol. 1993; 33, 237-42. Immune Interventions. pp 89-102, 2010 Linus Publications Deer Park, N.Y
81. Leger JM, Younes-Chennoufi AB, Chassande B, et al. Human immunoglobulin 100. Gajdos P, Chevret S, Toyka K. Intravenous immunoglobulin for myasthenia
treatment of multifocal motor neuropathy and polyneuropathy associated with gravis. Cochrane Database Syst Rev. 2008;(1):CD002277.
monoclonal gammopathy. J Neurol Neurosurg Psychiatry. 1994; 57S, 46-49. 101. Gajdos PH, Chevret S, Clair B, et al. Clinical trial of plasma exchange and high-
82. Nobile-Orazio E, Meucci N, Barbieri S, et al. High-dose immunoglobulin therapy dose intravenous immunoglobulin in myasthenia gravis. Ann Neurol.1997; 41,
in multifocal motor neuropathy. Neurology. 1993; 43, 537-544. 789-796.
83. Van den Berg LH, Franssen H, Wokke JHJ. The long term effect of intravenous 102. Gadjos P. Intravenous immunoglobulin for myasthenia gravis. In Dalakas MC,
immunoglobulin treatment in multifocal motor neuropathy. Brain. 1998; 121, Spath PJ (Eds) Intravenous Immunoglobulins in the Third Millennium. (203-205)
421-428. Lancaster, UK. Parthenon Publishing. 2004
84. Van den Berg LH, Kerkhoff H, Oey PL, et al. Treatment of multifocal motor neu- 103. Zinman L, Ng E, Bril V.IV immunoglobulin in patients with myasthenia gravis: a
ropathy with high dose intravenous immunoglobulins: a double blind, placebo randomized controlled trial. Neurology. 2007;68(11):837-41.
controlled study. J Neurol Neurosurg Psychiatry. 1995; 59, 248-252. 104. Ronager J, Ravnborg M, Hermansen I, Vorstrup S. Immunoglobulin treatment
85. Federico P, Zochodne DW, Hahn AF,et al. Multifocal motor neuropathy im- versus plasma exchange in patients with chronicmoderate to severe myasthe-
proved by IVIg: randomized, double-blind, placebo-controlled study. Neurology. nia gravis. Artificial Organs 2001;25(12):96773.
2000; 55, 1256-1262. 105. Wolf HM, Eibl MM. Immunomodulatory effect of immunoglobulins. Clin Exp
86. Van den Berg-Vos RM, Franssen H, Wokke JH, Van den Berg LH. Multifocal motor Rheumatol. 1996;14 Suppl 15:S17-25.
neuropathy: long-term clinical and electrophysiological assessment of intra- 106. Bain PG, Motomura M, Newsom-Davis J, et al. Effects of intravenous im-
venous immunoglobulin maintenance treatment. Brain.2002; 125, 1875-1886. munoglobulin (IVIg) treatment on muscle weakness and calcium channel au-
87. Terenghi F, Cappellari A, Bersano A, et al. How long is IVIg effective in multifocal toantibodies in the Lambert-Eaton myasthenic syndrome. Neurology. 1996; 47,
motor neuropathy? Neurology.2004; 62, 666-668. 678-683.
88. Elliott JL, Pestronk A. Progression of multifocal motor demyelinating neuropa- 107. Dalakas MC. Controlled studies with high-dose intravenous immunoglobulin in
thy during apparently successful treatment with human immunoglobulin. Neu- the treatment of dermatomyositis, inclusion body myositis and polymyositis.
rology.1994; 44, 967-968. Neurology.1998; 51(Suppl), S37-S45.
89. Dalakas M.C., Rakocevic G., Salajegheh M., et al. Placebo-controlled trial of rit- 108. Mastaglia FL, Phillips BA, Zilko PJ. Immunoglobulin therapy in inflammatory
uximab in IgM anti-myelin- associated glycoprotein antibody demyelinating neu- myopathies. J Neurol Neurosurg Psychiatry.1998; 65, 107-110.
ropathy Ann Neurol. 2009; 65(3):286-93 109. Raju R, Dalakas MC. Gene expression profile in the muscles of patients with in-
90. Vucic S, Black KR, Chong PS, Cros D. Multifocal motor neuropathy: decrease in flammatory myopathies: effect of therapy with IVIg and biologic validation of
conduction blocks and reinnervation with long-term IVIg. Neurology. clinical relevant genes. Brain 2005;128:1887-1896.
2004;63(7):1264-9 110. Dalakas MC. IVIg in patients with anti-GAD antibody associated neurological dis-
91. Dalakas MC, Stein DP, Otero C, et al. Effect of high-dose intravenous im- eases and patients with inflammatory myopathies: effects on clinicopathologic
munoglobulin on amyotrophic lateral sclerosis and multifocal motor neuropa- features and immunoregulatory genes. Clin Review in Allergy & Immunol.
thy. Arch Neuro. 1994;l 51, 861-864. 2005;29:255-269.

10
111. Dalakas MC, Sonies B, Dambrosia J, et al. Treatment of inclusion body, myositis 128. Schiff RJ. Transmission of viral infections through intravenous immune globulin.
with IVIg: a double-blind, placebo-control study. Neurology.1997; 48, 712-716. N Engl J Med.1994; 331, 1649-50.
112. Dalakas MC, Fujii M, Li M,et al. High-dose intravenous immune globulin for stiff- 129. Silbert PL, Knezevic WV, Bridge DT. Cerebral infarction complicating intravenous
person syndrome. N Engl J Med .2001;345, 1870-1876. immunoglobulin therapy for polyneuritis cranialis. Neurology.1992; 42, 257-8.
113. Walter MC, Lochmuller H, Toepfer M, et al. High-dose immunoglobulin therapy 130. Steg RE, Lefkowitz DM. Cerebral infarction following intravenous immunoglob-
in sporadic inclusion body myositis: a double-blind, placebo-controlled study. J ulin therapy for myasthenia gravis. Neurology.1994; 44, 1180-1181.
Neurol. 2000; 247:22-28. 131. Woodruff RK, Griff AP, Firkin FL, Smith IL. Fatal thrombic events during treat-
114. Cherin P, Pelletier S, Teixeira A, et al. Intravenous immunoglobulin for dysphagia ment of autoimmune thrombocytopenia with intravenous immunoglobulin in
of inclusion body myositis. Neurology.2002; 58, 326. elderly patients. Lancet.1986; 2, 217-218.
115. Dalakas MC, Fujii M, Li M, McElroy B. The clinical spectrum of anti-GAD antibody- 132. Dalakas MC, Clark WM. Strokes, thromboembolic events, and IVIg: rare inci-
positive patients with stiff-person syndrome. Neurology.2000; 55, 1531-1535. dents blemish an excellent safety record. Neurology.2003; 60, 1736-1737.
116. Dalakas MC. Inhibiting leucocyte recruitment to the brain by IVIg. Is it relevant 133. Voltz RV, Rosen F, Yousry T, et al. Reversible encephalopathy with cerebral va-
to the treatment of demyelinating CNS disorders? (Editorial) Brain sospasm in a GBS patient treated with IV immunoglobulin. Neurology.1996; 46,
2004;127:2569-2571. 250-251.
117. Achiron A, Kishner I, Dolev M, et al. Effect of intravenous immunoglobulin treat- 134. Constantinescu CS, Chang AP, McClusky LF. Recurrent migraine and intra-
ment on pregnancy and postpartum-related relapses in multiple sclerosis. J venous immune globulin therapy. N Engl J Med.1993; 329, 583-4.
Neurol. 2004;251(9):1133-7. 135. Sekul EA, Cupler EJ, Dalakas MC. Aseptic meningitis associated with high-dose
118. Stangel M, Gold R. Intravenous Immunoglobulins in MS. Int MS J. 2005;12(1):5-10, 4. intravenous immunoglobulin therapy: frequency and risk factors. Ann Intern
119. Dalakas MC. Role of IVIg in autoimmune, neuroinflammatory and neurodegen- Med. 1994;121(4):259-62.
erative disorders of the central nervous system: present and future prospects. 136. Dalakas MC. Meningitis and skin reactions after Intravenous Immunoglobulin
J Neurol. 2006;253 Suppl 5:v25-v32. therapy. Ann Intern Med.1997; 127, 1130.
120. Dodel RC, Du Y, Depboylu C, et al. Intravenous immunoglobulins containing an- 137. Dalakas MC. Aseptic meningitis and intravenous immunoglobulin. Ann Intern
tibodies against beta-amyloid for the treatment of Alzheimer's disease. J Neurol Med.1995; 122, 316-7.
Neurosurg Psychiatry. 2004;75(10):1472-4. 138. Burks AW, Sampson HA, Buckley RH. Anaphylactic reactions after gamma glob-
121. Dalakas MC. Post-polio Syndrome 12 years later: How it all started. Ann NY Acad ulin administration in patients with hypogammaglobulinemia. N Engl J Med.
Sci .1995;.753:11-18 1986;314, 560-564.
122. Gonzalez H, Sunnerhagen KS, Sjberg I, et al. Intravenous immunoglobulin for 139. Bjrkander J, Hammarstrm L, Smith E, et al. Immunoglobulin prophylaxis in
post-polio syndrome: a randomised controlled trial. Lancet Neurol. patients with antibody deficiency syndromes and anti-IgA antibodies. J Clin Im-
2006;5(6):493-500. munol. 1987;7, 8-15.
123. Kurlander RJ, Hall J. Comparison of intravenous gammaglobulin and a mono- 140. Ellie E, Combe C, Ferrer X. High-dose intravenous immune globulin and acute
clonal anti-Fc receptor antibody as inhibitors of immune clearance in vivo in renal failure. N Engl J Med. 1992; 327, 1032-1033.
mice. J Clin Invest .1986;77, 2010-2018. 141. Ahsan N, Palmer BF, Wheeler D, et al. Intravenous immunoglobulin-induced os-
124. Newburger JW, Takahashi M, Berser AS, et al. A single intravenous infusion of motic nephrosis. Arch Intern Med . 1994;154, 1985-1987.
gamma globulin as compared with four infusions in the treatment of acute 142. Ahsan N. Intravenous immunoglobulin induced-nephropathy: a complication
Kawasaki syndrome. N Engl J Med. 1991; 324, 1633-9. of IVIg therapy. J Nephrol. 1998; 11, 157-161.
125. Dwyer JM. Manipulating the immune system with immune globulin. . N Engl J 143. Koffman BM, Dalakas MC. . Effect of high-dose intravenous immunoglobulin
Med. 1992; 326,107-16. on serum chemistry, hematology and lymphocyte subpopulations: assessments
126. Dalakas MC. . High-dose intravenous immunoglobulin and serum viscosity: Risk based on controlled treatment trials in patients with neurological diseases.
of precipitating thromboembolic events. Neurology.1994; 44, 223-226. Muscle Nerve. 1997; 20, 1102-1107.
127. Hague RA, Eden DB, Yap PL, et al. Hyperviscosity in HIV infected children: a po-
tential hazard during intravenous immunoglobulin therapy. Blut. 1990; 61, 66-67.

This publication is a supplement to INFUSION (ISSN 1080-3858), the bimonthly journal of the National Home Infusion Association,
100 Daingerfield Rd., Alexandria, VA 22314 (703-549-3740). Originally published in September/October 2010 (Vol. 16, Number 5).
Copyright 2010 by NHIA.

Das könnte Ihnen auch gefallen