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

Human Vaccines & Immunotherapeutics

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/khvi20

Progress in Guillain–Barré syndrome


immunotherapy—A narrative review of new
strategies in recent years

Jiajia Yao, Rumeng Zhou, Yue Liu & Zuneng Lu

To cite this article: Jiajia Yao, Rumeng Zhou, Yue Liu & Zuneng Lu (2023) Progress
in Guillain–Barré syndrome immunotherapy—A narrative review of new strategies
in recent years, Human Vaccines & Immunotherapeutics, 19:2, 2215153, DOI:
10.1080/21645515.2023.2215153

To link to this article: https://doi.org/10.1080/21645515.2023.2215153

© 2023 The Author(s). Published with


license by Taylor & Francis Group, LLC.

Published online: 06 Jun 2023.

Submit your article to this journal

Article views: 3124

View related articles

View Crossmark data

Citing articles: 1 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=khvi20
HUMAN VACCINES & IMMUNOTHERAPEUTICS
2023, VOL. 19, NO. 2, 2215153
https://doi.org/10.1080/21645515.2023.2215153

REVIEW

Progress in Guillain–Barré syndrome immunotherapy—A narrative review of new


strategies in recent years
Jiajia Yao, Rumeng Zhou, Yue Liu, and Zuneng Lu
Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China

ABSTRACT ARTICLE HISTORY


Guillain – Barré syndrome (GBS) is an immune-mediated neuropathy, the pathology of which is not clear. Received 12 March 2023
Both cellular and humoral immunity are involved in the occurrence of the disease, and molecular mimicry Revised 3 May 2023
is currently the most widely recognized pathogenesis. Intravenous immunoglobulin (IVIg) and plasma Accepted 15 May 2023
exchange (PE) have been proven to be effective in improving the prognosis of patients with GBS, but KEYWORDS
there has been no progress in the treatment of the disease or strategies to improve the prognosis. New Guillain–Barré syndrome;
treatment strategies for GBS are mostly immunotherapies, including treatment against antibodies, treatment; complement
complement pathways, immune cells and cytokines. Some of the new strategies are being investigated pathway; immunotherapy
in clinical trials, but none of them have been approved for the treatment of GBS. Here, we summarized
the current therapies for GBS, and new immunotherapies for GBS according to pathogenesis.

Introduction
Campylobacter jejuni. This suggests that the pathogenic
Guillain–Barré syndrome (GBS) is an immune-mediated neu­ mechanism of AMAN may be the cross-reaction of homolo­
ropathy that is the most common cause of acute flaccid paraly­ gous epitopes between bacterial lipo-oligosaccharides and per­
sis and affects approximately 100,000 people per year ipheral motor axon gangliosides.18–21
worldwide. Antecedent events are often found 4 weeks before Miller-Fisher syndrome (MFS) is a variation of GBS, and
clinical syndromes appear in GBS patients, such as surgery or the clinical features of MFS are facial muscle weakness and
infection and Campylobacter jejuni, Haemophilus influenzae, ataxia. Most MFS patients have anti-GQ1b antibodies, imply­
cytomegalovirus, Zika virus and Japanese encephalitis virus are ing a potential role for ganglioside antibodies in disease patho­
widely discussed.1–7 genesis or as reliable diagnostic biomarkers.22–24 Anti-GQ1b
Depending on the different sites of the immune response, antibodies have been proven to activate complement at the
GBS is generally divided into demyelinating and axonal sub­ neuromuscular junction in vitro, and complement activation is
types. Acute inflammatory demyelinating polyradiculoneuropa­ thought to be the primary pathogenic mechanism of MFS.25
thy (AIDP) is characterized by the demyelination of peripheral Inflammatory cells and inflammatory factors play an impor­
nerves and infiltration of inflammatory cells, with subsequent tant role in the pathogenesis of GBS. Macrophages play a dual
axonal damage. Antibodies can be detected on Schwann cells, role in the pathogenesis of GBS. Proinflammatory macrophages
and AIDP is the most common subtype in Western countries.8,9 (M1) and anti-inflammatory macrophages (M2) play a decisive
Antigenic epitopes of bacteria and viruses are presented to role in the initiation and development of GBS and EAN. M1
T cells by activated macrophages, causing the cross reactivity macrophages can promote the destruction of the blood-nerve
of T cells. Activated T cells promote the release of cytokines and barrier, induce the production of cytokines and chemokines,
free radicals, disrupt the blood nerve barrier and damage mye­ promote Th1 polarization, and eventually lead to demyelination
lin, ultimately leading to acute demyelination syndrome.10 of peripheral nerves. M2 macrophages play a protective role in
Experimental autoimmune neuritis (EAN) is an animal model the course of disease; they can promote T-cell apoptosis, remove
of AIDP that uses myelin epitopes P0 or P2 as major antigens to myelin and axon fragments, inhibit inflammation, and promote
induce T-cell-mediated neuritis.1,11 the regeneration of axons and myelin.26,27
Acute motor axonal neuropathy (AMAN) is the second T-cell subtypes are also involved in the pathogenesis of
most common subtype of GBS. AMAN presents as primary GBS. The imbalance between Th1 and Th2 responses contri­
axonal injury with antibody and membrane attack complex butes to the pathogenesis of GBS.28–30 Th1 responses are
(MAC) deposition in nodes of Ranvier without obvious thought to provoke disease by activating and recruiting macro­
inflammatory cell infiltration or demyelination.1,12–14 phages to sites in peripheral nerves, subsequently leading to
Antibodies related to AMAN include anti-GM1 and anti- nerve damage induced by the direct action of macrophages or
GD1a antibodies.14–17 In AMAN patients related to by toxic and inflammatory substances released in situ. On the
Campylobacter jejuni, the gangliosides of the peripheral nerves other hand, the Th2 response acts as a suppressor and regu­
are similar in structure to the lipo-oligosaccharides of lator of the Th1 pathway and therefore may have the resolving

CONTACT Zuneng Lu lzn196480@126.com Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
© 2023 The Author(s). Published with license by Taylor & Francis Group, LLC.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way. The
terms on which this article has been published allow the posting of the Accepted Manuscript in a repository by the author(s) or with their consent.
2 J. YAO ET AL.

Table 1. New therapies used for treatment of GBS in recent years.


Species Therapy Type Pathway or approach Year/author
Rat PR-957 Original article T cell and cytokine regulation 2017/Liu H 67
Rabbit IdeS Original article Cleaved antibodies 2017/Wang Y 51
Rat Fingolimod Original article Inflammatory cells (macrophages and T cells) 2017/Ambrosius B 74
Rat cyclo-DPAKKR Original article p75NTR pathway 2017/Gonsalvez DG 77
Rat 4-AP Original article Not clear 2017/Moriguchi K 78
Human Eculizumab Clinical trial Complement pathway 2018/Misawa S 54
Rat Decitabine (DAC) Original article Treg cell induction 2018/Fagone P 59
Rat 2-deoxy-D glucose (2-DG) Original article glycolytic pathway 2018/Liu RT 60
Rat Bifidobacterium Original article T cell regulation 2018/Shi P 62
Rat ZSTK474 Original article T cell and cytokine regulation 2018/Chen X 69
Rat Vasoactive intestinal peptide (VIP) Original article Inflammatory cells regulation 2018/Jiao H 73
Rat Dimethyl fumarate(DMF) Original article Inflammatory cells (macrophages and T cells) 2019/Pitarokoili K 58
Rat TLCK Original article Thrombin-PAR1 pathway 2019/Shavit-Stein E 75
Human Second course of IVIg Retrospective study Immune regulation 2020/Verboon C 42
Mouse Fasudil Original article Inflammatory cells (macrophages and T cells) 2020/Zhao Y 61
Human Second course of IVIg Clinical trial Immune regulation 2021/Walgaard C 43
Mouse Ginkgolide Original article T cell and cytokine regulation 2021/Li C 68
Rat Ginsenoside Rd Original article Monocyte modulation 2021/Ren K 72
Rat Nanoparticles (NPs) Original article Inflammatory cells regulation 2022/Elahi E 71
Rat Bifidobacterium Original article PD-1 signaling 2023/Shi P 63

effect observed in the recovery phase of GBS and EAN.31–33 treatment/therapy, experimental autoimmune neuritis, Miller-
Recently, IL-17 and Th17 cells have been found to play an Fisher syndrome, etc. Clinical trials that were not completed
important role in many immune diseases, and Treg cells have were excluded. Titles and abstracts of papers were screened by
a suppressive effect on inflammation.34 In the acute phase of reviewers to identify whether they met the criteria of the
the clinical course of GBS, the number and ratio of CD4+CD25 review. All references were examined for background informa­
+ T cells decrease, but this decrease is reversible.35 Another tion and potentially relevant articles.
study found that regulatory T cells (Tregs) from GBS patients
and Tregs from healthy controls showed equal expression of
FoxP-3 mRNA, and their ability to suppress the proliferation Results
and cytokine secretion of CD4+ effector T cells was unim­
paired in GBS patients.36 Furthermore, adoptive infusion of Classical therapies for GBS
autologous CD4+CD25+ Treg cells can reduce inflammatory Intravenous immunoglobulin (IVIg) and plasma exchange
cell infiltration of the sciatic nerve in EAN rats.37 (PE) have been proven to be effective in improving the prog­
Cytokines are small active proteins secreted by immune nosis of patients with GBS and should be applied before the
cells and some nonimmune cells that have several functions, occurrence of irreversible axonal injury as soon as possible.1
such as regulating cell growth and differentiation, modulating These two therapies have equal effects.1,39 Patients undergo
the immune response, and participating in the inflammatory five sessions of PE with 50 ml/kg plasma each session, admi­
response. Proinflammatory cytokines such as interleukin (IL)- nistered over 1–2 weeks. The typical dose for IVIg is 0.4 g/kg
1β, IL-6, IL-12, tumor necrosis factor (TNF)-α, interferon body weight per day for five consecutive days.39–41 The
(IFN)-γ, etc., damage myelin by recruiting effector cells to mechanism by which IVIg and PE are effective for GBS is
peripheral nerves and promoting the in situ release of toxic still unclear but mostly involves removing the antibodies and
substances. Anti-inflammatory cytokines, such as IL-4 and IL- MAC in plasma and regulating immune cells.
10, inhibit disease progression or promote myelin repair by A single course of IVIg is effective for GBS patients, but
exerting anti-inflammatory effects.38 some patients still have poor outcomes. A possible cause for this
Along with more studies on the pathogenesis of GBS, new may be the rapid consumption of immunoglobulin, and
strategies targeting at different points in the treatment of the a second course of IVIg may benefit these patients.
disease have emerged. In this narrative review, we summarize A retrospective study in 2020 and a prospective study in 2021
the new approaches of classical therapies and new strategies in both showed that patients with poor outcomes after the first
both animal models and clinical practice to identify potential course of IVIg could not benefit from the repeated use of IVIg,
therapies for GBS patients. and research suggested that other immunomodulatory treat­
ments should be used for GBS patients with poor prognosis.42,43
Although IVIg and PE are the most effective therapies for
Materials and methods
GBS, it has been proven that neither IVIg nor PE can prevent
Articles published between 2017 and 2022 were included in the disease course or nerve damage.6 PE requires specific equip­
our research, and clinical trials, original articles, retrospective ment, and hypocalcemia, thrombosis, pneumothorax, compli­
studies and systematic reviews in MEDLINE via PubMed cations from central venous access and allergic reactions are the
interface and Cochrane Library were all carefully examined. main complications. Moreover, PE should be avoided in
Both animal experiments and human studies were included. patients with severe autonomic dysfunction.44 IVIg is more
Key words for searching included Guillain – Barré syndrome, likely to be completed than PE but is much more expensive,
HUMAN VACCINES & IMMUNOTHERAPEUTICS 3

and liver dysfunction and thromboembolic events are rare but adverse events. However, because the outcome indicators did
severe adverse events of IVIg.1 More research on classical not meet expectations, the researchers suggested that further
treatments is needed for patients with mild disease and patients large-scale prospective studies were needed to prove the effect
whose treatment starts more than 2 weeks after disease onset, of eculizumab.54 The 2020 Cochrane Database of Systematic
and new strategies are also needed for patients with poor out­ Reviews also pointed out that the current level of evidence for
comes after treatment with PE or IVIg. eculizumab in the treatment of GBS is low.46
Neither oral nor intravenous use of glucocorticoids in GBS Previous studies showed that C5 inhibition could mitigate
can accelerate recovery, nor can they affect the long-term nerve injuries, but Rhona McGonigal determined that the early
outcome of patients.45 The combination of glucocorticoids stage of complement activation could also cause immune cell
and IVIg does not have a better effect than IVIg alone,46,47 recruitment. C1q is the first complement cascade molecule in
but in the early phase of the disease, the combination of the classical pathway. Two animal models were used to evalu­
glucocorticoids and IVIg has a better effect in GBS patients.47 ate the efficacy of the anti-C1q antibody (M1). Studies have
shown that anti-C1q treatment reduces axonal injury, and
improves respiratory function in mouse models.55 ANX005 is
New strategies in GBS treatment
a humanized immunoglobulin G4 (IgG4) recombinant anti­
Therapies for antibodies body against C1q that blocks the initiation of the classical
Neonatal Fc receptor (FcRn) can adjust the concentration of complement cascade. Inhibition of C1q can be used in acute
endogenous IgG. Inhibiting FcRn can reduce the level of IgG immune-mediated diseases such as GBS, and the pharmacoki­
in the body, and it has been proven effective in animal experi­ netics and pharmacology are currently under study.56 ANX005
ments. Anti-ganglioside antibody levels in mice lacking FcRn has not been used to treat in GBS patients or animal models,
are significantly reduced. The use of FcRn inhibitors signifi­ and it may be a promising treatment option.
cantly reduces the antibody levels in mice, reducing nerve
damage and clinical symptoms. This suggests that FcRn inhi­ Therapies inhibiting inflammatory cells and inflammatory
bitors can be used to treat GBS in the future.48 factors
Immunoglobulin G-degrading enzyme of Streptococcus A study by Ranran Han et al. found that dimethyl fumarate
pyogenes (IdeS) is secreted by Streptococcus pyogenes and can (DMF) improved the demyelination and inflammatory cell
cleave IgG antibodies into F(ab’)2 and Fc fragments, thereby infiltration of the sciatic nerve when used in the treatment of
inhibiting the killing of S. pyogenes by the immune response of EAN rats. DMF reduces the level of M1 macrophages and
hosts.49 Ryo Takahashi found that IdeS efficiently cleaved IgG increases the level of M2 macrophages in the spleen and sciatic
and blocked complement activation in vitro.50 A further study nerve. In the sciatic nerve, DMF treatment increases the level of
showed that IdeS could reduce complement deposition in the nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and its
spinal nerve heel and significantly facilitate the clinical recov­ target gene hemooxygenase-1 (HO-1), which can promote the
ery process in the rabbit model of AMAN, and axonal degen­ transfer of macrophages to M2-type polarization. In addition,
eration of the anterior spinal nerve root was significantly DMF also improves the inflammatory environment of the
reduced in IdeS-treated rabbits.51 spleen of EAN rats, characterized by the downregulation of
IFN-γ, TNF-α, IL-6 and IL-17 messenger RNA (mRNA) and
Therapies for the complement pathway upregulation of IL-4 and IL-10 mRNA levels.57 Another study
Anti-GQ1b antibodies bind and destroy neuromuscular junc­ showed that DMF regulated T-cell proliferation and differentia­
tions, causing muscle paralysis. This damage activates comple­ tion through different regions and layers of the small intestine,58
ment and ultimately leads to the deposition of membrane indicating that DMF protects EAN rats from nerve damage
attack complex (MAC) C5b-9. Susan K. Halstead and collea­ through multiple mechanisms.
gues conducted a study to block the role of C5b-9 in auto­ A preventive and therapeutic study of decitabine (DAC) in
immune peripheral neuropathy using eculizumab to treat EAN suggests that DAC can increase the number of thymic
MFS. Studies have shown that the application of eculizumab Tregs and reduce the production of proinflammatory cyto­
in MFS mice can effectively prevent respiratory failure and kines, thereby improving the clinical symptoms of EAN.59
neurological symptoms.52 Furthermore, they conducted Ru-Tao Liu’s research found that 2-deoxy-D glucose
a randomized trial to investigate the effect of eculizumab in (2-DG) inhibits the initiation and development of EAN at
GBS patients. The clinical trial included 28 patients diagnosed the same time by inhibiting the glycolytic pathway, simulta­
with GBS on the basis of a functioning score greater than 2 neously inhibiting the differentiation of Th1 and Th17 cells
points, and 8 subjects were finally recruited. Four weeks after and enhancing the development of Treg cells. The enhance­
recruitment, 2 out of 2 patients received placebo, and 2 out of 5 ment of the glycolysis pathway can promote the pathogenesis
patients received eculizumab and had decreased functioning of EAN, and inhibiting glycolysis may be a new treatment for
scores of more than one point. The results indicated the need GBS.60
for further studies on eculizumab.53 A prospective study was A study of fasudil for EAN found that Th1 cell and Th17
carried out on the application of eculizumab in GBS patients. cell proportions were decreased in mice treated with fasudil,
The study included patients with a GBS disability score of 3–5. the proportion of Th2 cells was similar, and the proportion of
After 4 weeks of treatment, the proportion of patients in the Treg cells in splenocytes was increased. Fasudil has a good
eculizumab and placebo groups who were able to walk inde­ therapeutic effect on EAN by attenuating Th1/Th17 cells and
pendently was 61% and 45%, respectively, but both groups had promoting Treg activation and M2 macrophage polarization.61
4 J. YAO ET AL.

Shi Peng et al. found that the level of Bifidobacterium in GBS low-dose fingolimod could reduce circulating peripheral blood
patients was significantly lower than that in healthy controls T cells and infiltrating T cells and macrophages in the sciatic
(HCs), and the concentration of Bifidobacterium was negatively nerve, and fewer apoptotic Schwann cells were found in rats
correlated with Th2 and Th17 subgroups. Treatment with treated with fingolimod at disease nadir.74
Bifidobacterium significantly reduced the levels of Th2 and
Th17 cells and increased the level of Treg cells in EAN rats. Therapies targeting other mechanisms
This finding suggested that Bifidobacterium could alleviate GBS Efrat Shavit-Stein found that thrombin and its protease-
by regulating Th17 and Treg cells.62 Further study showed that, activated receptor 1 (PAR1) participate in the development of
the expression level of programmed death (PD)-1 increased in EAN. N-Tosyl-Lys-chloromethylketone (TLCK) and N-alpha 2
EAN rats treated with Bifidobacterium. The function of
naphthalenesulfonylglycyl 4 amidino-phenylalaninepiperidide
Bifidobacterium in regulating T cells was partly blocked by
(NAPAP) were used as PAR1 inhibitors to block the thrombin-
inhibiting the expression of PD-1, suggesting that
PAR1 pathway. In vitro studies showed that both TLCK and
Bifidobacterium alleviates GBS in a partial way through PD-1.63
NAPAP could inhibit the thrombin-PAR1 pathway in rat sciatic
The immunoproteasome plays a critical role in homeostasis
nerve damage. In vivo studies showed that TLCK- or NAPAP-
and immunity, especially in processing antigens for presenta­
treated rats had improved clinical scores, and TLCK treatment
tion on major histocompatibility complex (MHC) class
could prevent structural damage to the node of Ranvier in the
I molecules to CD8+ T lymphocytes.64–66 The subunit of the
sciatic nerve.75
immunoproteasome, low-MW polypeptide (LMP) 7, has an
Neurotrophic factors are essential for the development and
important role in cytokine production. PR-957 is a highly
damage repair of the peripheral nervous system. The p75
selective inhibitor of LMP7, and previous studies found that
neurotrophic receptor (p75NTR) is fundamental to peripheral
PR-957 can regulate the differentiation of Th cells and reduce
nerve growth.76 Brain-derived neurotrophic factor (BDNF)
the amount of proinflammatory cytokines. PR-957 reduced the
promotes peripheral nerve myelination via the p75NTR path­
proportion of Th17 cells in the sciatic nerve and spleen in EAN
way. David G. Gonsalvez used the cyclic pentapeptide cyclo-
rats, decreased the expression of IL-6 and IL-23, and down­
[DPro-Ala-Lys-Lys-Arg] (cyclo-DPAKKR), a structural
regulated STAT3 phosphorylation, thereby reducing the sever­
mimetic of BDNF to investigate the therapeutic effect in
ity and duration of EAN.67
EAN rats. The study found that cyclo-DPAKKR administra­
Chunrong Li et al. found that ginkgolide can decrease the
tion limited the extent of inflammatory demyelination and
score of EAN mice and delay the peak of disease. It can also
axonal damage by inhibiting the p75NTR pathway, and the
downregulate the proportion of Th17 cells in the spleen of
therapeutic effect of cyclo-DPAKKR was abrogated.77
EAN mice and reduce the levels of IFN-γ and IL-12 in GBS
Kota Moriguchi found that 4-aminopyridine (4-AP) could
patients, suggesting that ginkgolides have potential therapeutic
reduce the clinical scores of EAN rats and improve the electro­
effects in GBS patients and the EAN model.68
physiological properties, but the histological assessment
The class I phosphatidylinositol 3-kinase inhibitor
revealed no significant difference between the 4-AP and con­
2-(2-difluoromethylbenzimidazol-1-yl)-4,6-dimorpholino-
trol groups.78 The mechanism of 4-AP in the treatment of
1,3,5-triazine (ZSTK474) was also proven to be effective in
EAN is not clear, and further studies are needed to investigate
alleviating the inflammatory response of sciatic nerves. The
the therapeutic effect of 4-AP.
author reported that ZSTK474 could reduce the number of
Th1/Th17 cells and levels of proinflammatory cytokines
through the PI3K/AKT/mTORC1 pathway, suggesting
Discussion
a possible anti-inflammatory therapy for GBS.69
Nanoparticles (NPs) can bind with monocytes and remove In 1916, Guillain, Barré and Strohl reported two cases of acute
monocytes from the lesion site to the spleen.70 Ehsan Elahi flaccid paralysis with high cerebrospinal fluid (CSF) protein
et al. used drug-free poly-lactic coglycolic acid (PLGA)-based levels and normal cell counts, which is now known as GBS. In
NPs to modulate the inflammatory cells of EAN rats and found the past century, we have made great progress in understanding
that regardless of different treatment approaches, NPs showed the clinical variations, pathology and treatment of GBS.79,80
good effects in modulating inflammatory cells in circulation Until the 1970s, there was no specific and effective treat­
system, helping to reduce disease severity.71 ment for GBS. In the late 1970s, PE was first used to treat
Kaixi Ren et al. found that ginsenoside Rd (GSRd) could GBS.81 In 1984, two clinical trials showed that PE was efficient
also protect against nerve damage in EAN rats by modulating in GBS patients.82,83 A year later, one larger study confirmed
monocyte conversion and increasing resolution-phase macro­ the results and found that PE could benefit patients who were
phage infiltration, showing that GSRd may be useful for GBS unable to walk independently, especially when treatment was
patients.72 started within 2 weeks of disease onset.84 PE was the first
Furthermore, Hong Jiao et al. found that vasoactive intest­ treatment proven to be effective for GBS, but the treatment
inal peptide (VIP) can inhibit neural inflammation, reduce also carries risk for patients with autonomic dysfunctions,
inflammatory cytokine levels and improve body functions in which are common in GBS patients.
EAN rats.73 Ten years after the first report of PE, immunoglobulin
Björn Ambrosius et al. used fingolimod as an immunomo­ was first reported in the treatment of GBS.85 A randomized
dulator for the treatment of EAN rats. A study showed that clinical trial (RCT) in 1992 compared the efficiency of PE
HUMAN VACCINES & IMMUNOTHERAPEUTICS 5

and IVIg and found that IVIg was an effective alternative 2. Jacobs BC, Rothbarth PH, van der Meché FG, Herbrink P,
to PE; patients treated with IVIg showed more improve­ Schmitz PIM, de Klerk MA, van Doorn PA. The spectrum of
ment after 4 weeks than those with PE.86 Currently, clinical antecedent infections in Guillain-Barré syndrome: a case-control
study. Neurology. 1998;51(4):1110–15. doi:10.1212/WNL.51.4.
trials have shown that PE and IVIg have equal effects.1,39 1110.
IVIg has become the most popular therapy in most coun­ 3. Cao-Lormeau VM, Blake A, Mons S, Lastère S, Roche C,
tries, unless the costs limit its use. Vanhomwegen J, Dub T, Baudouin L, Teissier A, Larre P, et al.
The treatment of GBS is still challenging for neurologists. Guillain-Barré syndrome outbreak associated with Zika virus
Traditional therapies such as PE and IVIg are effective, but the infection in French Polynesia: a case-control study. Lancet.
2016;387(10027):1531–9. doi:10.1016/S0140-6736(16)00562-6.
high price of immunoglobulin and lack of specific equipment and
4. Islam Z, Jacobs BC, van Belkum A, Mohammad QD, Islam MB,
PE specialists limit their use. New immunotherapies start by Herbrink P, Diorditsa S, Luby SP, Talukder KA, Endtz HP. Axonal
clearing antibodies, reducing membrane attack complexes and variant of Guillain-Barre syndrome associated with
regulating the immune response, and targeting other pathways Campylobacter infection in Bangladesh. Neurology. 2010;74
are also considered to be effective in EAN models or clinical trials. (7):581–7. doi:10.1212/WNL.0b013e3181cff735.
5. Hao Y, Wang W, Jacobs BC, Qiao B, Chen M, Liu D, Feng X,
New therapies are aimed at the pathology of GBS, which means
Wang Y. Antecedent infections in Guillain-Barré syndrome:
they are more specific. Most of the new therapies are drugs that a single-center, prospective study. Ann Clin Transl Neurol.
are easy to produce, transport and store, and the use of new 2019;6(12):2510–7. doi:10.1002/acn3.50946.
therapies does not require specific equipment. The combination 6. Leung J, Sejvar JJ, Soares J, Lanzieri TM. Guillain-Barré syndrome
of new therapies with classical therapies could help to improve the and antecedent cytomegalovirus infection, USA 2009-2015.
prognosis in patients with poor outcomes after treatment with Neurol Sci. 2020;41(4):885–91. doi:10.1007/s10072-019-04156-z.
7. Yamana M, Kuwahara M, Fukumoto Y, Yoshikawa K, Takada K,
classical therapies alone. Immunotherapies are still the most Kusunoki S. Guillain-Barré syndrome and related diseases after
popular methods for GBS treatment. Further clinical and basic influenza virus infection. Neurol Neuroimmunol Neuroinflamm.
studies are needed to prove the role of these methods in the 2019;6(4):e575. doi:10.1212/NXI.0000000000000575.
treatment of GBS and provide more possibilities for treatment. 8. Asbury AK, Arnason BG, Adams RD. The inflammatory lesion in
New therapies also have limitations. Most therapies have only idiopathic polyneuritis. Its role in pathogenesis. Medicine
(Baltimore). 1969;48(3):173–215. doi:10.1097/00005792-196905
been verified in animal models, and only eculizumab and IdeS are 000-00001.
undergoing clinical trials. The Cochrane library also reviewed the 9. Wanschitz J, Maier H, Lassmann H, Budka H, Berger T. Distinct
therapeutic effect of methods that have been used in RCTs and time pattern of complement activation and cytotoxic T cell
found that IFNβ-1a, CSF filtration and the Chinese herb tripter­ response in Guillain-Barré syndrome. Brain. 2003;126(Pt
ygium polyglycoside all had a very low level of certainty based on 9):2034–42. doi:10.1093/brain/awg207.
10. Ebrahim Soltani Z, Rahmani F, Rezaei N. Autoimmunity and
the evidence.46 Further studies should focus on the translation of
cytokines in Guillain-Barré syndrome revisited: review of patho­
basic research to clinical trials. Nonetheless, it will take a long time mechanisms with an eye on therapeutic options. Eur Cytokine
for new strategies to be applied in clinical practice. Netw. 2019;30(1):1–14. doi:10.1684/ecn.2019.0424.
In conclusion, IVIg and PE are still the most effective thera­ 11. Zhang HL, Zheng XY, Zhu J. Th1/Th2/Th17/Treg cytokines in
pies for GBS. Additionally, immunotherapies are still the most Guillain-Barré syndrome and experimental autoimmune neuritis.
popular methods for GBS treatment (Table 1). Further clinical Cytokine Growth Factor Rev. 2013;24(5):443–53. doi:10.1016/j.
cytogfr.2013.05.005.
and basic studies are needed to prove the role of these methods 12. Griffin JW, Li CY, Macko C, Ho TW, Hsieh ST, Xue P, Wang FA,
in the treatment of GBS and provide more possibilities for Cornblath DR, McKhann GM, Asbury AK. Early nodal changes in the
treatment. acute motor axonal neuropathy pattern of the Guillain-Barré syn­
drome. J Neurocytol. 1996;25(1):33–51. doi:10.1007/BF02284784.
13. Hafer-Macko C, Hsieh ST, Li CY, Sheikh K, Cornblath DR, Li CY,
Author contributions McKhann GM, Asbury AK, Griffin JW. Acute motor axonal neu­
ropathy: an antibody-mediated attack on axolemma. Ann Neurol.
JY and ZN provided the idea for the study, RZ, YL, JY collected the 1996;40(4):635–44. doi:10.1002/ana.410400414.
clinical data, JY analyzed the data and wrote the manuscript. All authors 14. Kuwabara S, Yuki N. Axonal Guillain-Barré syndrome: concepts
read and approved the final version of the manuscript and agreed with its and controversies. Lancet Neurol. 2013;12(12):1180–8. doi:10.
submission for publication. 1016/S1474-4422(13)70215-1.
15. Willison HJ, Jacobs BC, van Doorn PA. Guillain-Barré syndrome.
Lancet. 2016;388(10045):717–27. doi:10.1016/S0140-6736(16)
Disclosure statement 00339-1.
16. Lopez PH, Zhang G, Bianchet MA, Schnaar RL, Sheikh KA.
No potential conflict of interest was reported by the author(s).
Structural requirements of anti-GD1a antibodies determine their
target specificity. Brain. 2008;131(Pt 7):1926–39. doi:10.1093/
brain/awn074.
Funding
17. Willison HJ, Yuki N. Peripheral neuropathies and anti-glycolipid
The author(s) reported there is no funding associated with the work antibodies. Brain. 2002;125(Pt 12):2591–625. doi:10.1093/brain/
featured in this article. awf272.
18. Koga M, Takahashi M, Masuda M, Hirata K, Yuki N.
Campylobacter gene polymorphism as a determinant of clinical
References features of Guillain-Barré syndrome. Neurology. 2005;65
(9):1376–81. doi:10.1212/01.wnl.0000176914.70893.14.
1. Shahrizaila N, Lehmann HC, Kuwabara S. Guillain-Barré syn­ 19. Godschalk PC, Kuijf ML, Li J, St. Michael F, Ang CW, Jacobs BC,
drome. Lancet. 2021;397(10280):1214–28. doi:10.1016/S0140- Karwaski M-F, Brochu D, Moterassed A, Endtz HP, et al. Structural
6736(21)00517-1. characterization of Campylobacter jejuni lipooligosaccharide outer
6 J. YAO ET AL.

cores associated with Guillain-Barre and Miller Fisher syndromes. 37. Wang FJ, Cui D, Qian WD. Therapeutic effect of CD4+CD25+
Infect Immun. 2007;75(3):1245–54. doi:10.1128/IAI.00872-06. regulatory T cells amplified in vitro on experimental autoimmune
20. Susuki K, Yuki N, Schafer DP, Hirata K, Zhang G, Funakoshi K, neuritis in rats. Cell Physiol Biochem. 2018;47(1):390–402. doi:10.
Rasband MN. Dysfunction of nodes of Ranvier: a mechanism for 1159/000489919.
anti-ganglioside antibody-mediated neuropathies. Exp Neurol. 38. Lu MO, Zhu J. The role of cytokines in Guillain-Barré syndrome.
2012;233(1):534–42. doi:10.1016/j.expneurol.2011.11.039. J Neurol. 2011;258(4):533–48. doi:10.1007/s00415-010-5836-5.
21. Willison HJ. The translation of the pathological findings described 39. Hughes RA, Swan AV, van Doorn PA. Intravenous immunoglo­
in humans to experimental models of acute motor axonal bulin for Guillain-Barré syndrome. Cochrane Database Syst Rev.
neuropathy. J Peripher Nerv Syst. 2012;17(Suppl 3):3–8. doi:10. 2014;2014(9):Cd002063. doi:10.1002/14651858.CD002063.pub6.
1111/j.1529-8027.2012.00423.x. 40. Hughes RA, Swan AV, Raphaël JC, Annane D, van Koningsveld R,
22. Mori M, Kuwabara S, Fukutake T, Yuki N, Hattori T. Clinical van Doorn PA. Immunotherapy for Guillain-Barré syndrome:
features and prognosis of Miller Fisher syndrome. Neurology. a systematic review. Brain. 2007;130(Pt 9):2245–57. doi:10.1093/
2001;56(8):1104–6. doi:10.1212/WNL.56.8.1104. brain/awm004.
23. Chiba A, Kusunoki S, Shimizu T, Kanazawa I. Serum IgG antibody 41. Chevret S, Hughes RA, Annane D. Plasma exchange for Guillain-
to ganglioside GQ1b is a possible marker of Miller Fisher Barré syndrome. Cochrane Database Syst Rev. 2017;2(2):
syndrome. Ann Neurol. 1992;31(6):677–9. doi:10.1002/ana. Cd001798. doi:10.1002/14651858.CD001798.pub3.
410310619. 42. Verboon C, van den Berg B, Cornblath DR, Venema E,
24. Mori M, Kuwabara S. Fisher syndrome. Curr Treat Options Gorson KC, Lunn MP, Lingsma H, Van den Bergh P, Harbo T,
Neurol. 2011;13(1):71–8. doi:10.1007/s11940-010-0103-8. Bateman K, et al. Original research: second IVIg course in
25. Jacobs BC, O’Hanlon GM, Bullens RW, Veitch J, Plomp JJ, Guillain-Barré syndrome with poor prognosis: the non-
Willison HJ. Immunoglobulins inhibit pathophysiological effects randomised ISID study. J Neurol Neurosurg Psychiatry. 2020;91
of anti-GQ1b-positive sera at motor nerve terminals through inhi­ (2):113–21. doi:10.1136/jnnp-2019-321496.
bition of antibody binding. Brain. 2003;126(Pt 10):2220–34. 43. Walgaard C, Jacobs BC, Lingsma HF, Steyerberg EW, van den
doi:10.1093/brain/awg235. Berg B, Doets AY, Leonhard SE, Verboon C, Huizinga R,
26. Shen D, Chu F, Lang Y, Geng Y, Zheng X, Zhu J, Liu K. Beneficial Drenthen J, et al. Second intravenous immunoglobulin dose in
or harmful role of macrophages in Guillain-Barré syndrome and patients with Guillain-Barré syndrome with poor prognosis
experimental autoimmune neuritis. Mediators Inflamm. (SID-GBS): a double-blind, randomised, placebo-controlled
2018;2018:4286364. doi:10.1155/2018/4286364. trial. Lancet Neurol. 2021;20(4):275–83. doi:10.1016/S1474-
27. Koike H, Katsuno M. Macrophages and autoantibodies in demye­ 4422(20)30494-4.
linating diseases. Cells. 2021;10(4):844. doi:10.3390/cells10040844. 44. Schröder A, Linker RA, Gold R. Plasmapheresis for neurological
28. Nyati KK, Prasad KN, Rizwan A, Verma A, Paliwal VK. TH1 and disorders. Expert Rev Neurother. 2009;9(9):1331–9. doi:10.1586/
TH2 response to campylobacter jejuni antigen in Guillain-Barre ern.09.81.
syndrome. Arch Neurol. 2011;68(4):445–52. doi:10.1001/arch 45. Hughes RA, Brassington R, Gunn AA, van Doorn PA.
neurol.2011.51. Corticosteroids for Guillain-Barré syndrome. Cochrane Database
29. Zhu J, Bai XF, Mix E, Link H. Cytokine dichotomy in peripheral Syst Rev. 2016;10(10):Cd001446. doi:10.1002/14651858.
nervous system influences the outcome of experimental allergic CD001446.pub5.
neuritis: dynamics of mRNA expression for IL-1 beta, IL-6, IL-10, 46. Doets AY, Hughes RA, Brassington R, Hadden RD, Pritchard J.
IL-12, TNF-alpha, TNF-beta, and cytolysin. Clin Immunol Pharmacological treatment other than corticosteroids, intravenous
Immunopathol. 1997;84(1):85–94. doi:10.1006/clin.1997.4356. immunoglobulin and plasma exchange for Guillain-Barré syn­
30. Yeghiazaryan NS, Zara F, Capovilla G, Brigati G, Falsaperla R, drome. Cochrane Database Syst Rev. 2020;1(1):Cd008630. doi:10.
Striano P. Pyridoxine-dependent epilepsy: an under-recognised 1002/14651858.CD008630.pub5.
cause of intractable seizures. J Paediatr Child Health. 2012;48(3): 47. van Koningsveld R, Schmitz PI, Meché FG, Visser LH, Meulstee J,
E113–5. doi:10.1111/j.1440-1754.2010.01866.x. van Doorn PA. Effect of methylprednisolone when added to stan­
31. Créange A, Bélec L, Clair B, Degos JD, Raphaël JC, Gherardi RK. dard treatment with intravenous immunoglobulin for Guillain-
Circulating transforming growth factor beta 1 (TGF-beta1) in Barré syndrome: randomised trial. Lancet. 2004;363(9404):192–6.
Guillain-Barré syndrome: decreased concentrations in the early doi:10.1016/S0140-6736(03)15324-X.
course and increase with motor function. J Neurol Neurosurg 48. Zhang G, Lin J, Ghauri S, Sheikh KA. Modulation of IgG-FcRn
Psychiatry. 1998;64(2):162–5. doi:10.1136/jnnp.64.2.162. interactions to overcome antibody-mediated inhibition of nerve
32. Dahle C, Ekerfelt C, Vrethem M, Samuelsson M, Ernerudh J. regeneration. Acta Neuropathol. 2017;134(2):321–4. doi:10.1007/
T helper type 2 like cytokine responses to peptides from P0 and s00401-017-1730-x.
P2 myelin proteins during the recovery phase of Guillain-Barré 49. von Pawel-Rammingen U, Johansson BP, Björck L. IdeS, a novel
syndrome. J Neurol Sci. 1997;153(1):54–60. doi:10.1016/S0022- streptococcal cysteine proteinase with unique specificity for
510X(97)00178-0. immunoglobulin G. Embo J. 2002;21(7):1607–15. doi:10.1093/
33. Deretzi G, Zou LP, Pelidou SH, Nennesmo I, Levi M, Wahren B, emboj/21.7.1607.
Mix E, Zhu J. Nasal administration of recombinant rat IL-4 ame­ 50. Takahashi R, Yuki N. Streptococcal IdeS: therapeutic potential for
liorates ongoing experimental autoimmune neuritis and inhibits Guillain-Barré syndrome. Sci Rep. 2015;5:10809. doi:10.1038/
demyelination. J Autoimmun. 1999;12(2):81–9. doi:10.1006/jaut. srep10809.
1998.0259. 51. Wang Y, Shi Q, Lv H, Hu M, Wang W, Wang Q, Qiao B,
34. Wu X, Wang J, Liu K, Zhu J, Zhang HL. Are Th17 cells and their Zhang G, Lv Z, Kjellman C, et al. IgG-degrading enzyme of
cytokines a therapeutic target in Guillain-Barré syndrome? Expert Streptococcus pyogenes (IdeS) prevents disease progression
Opin Ther Targets. 2016;20(2):209–22. doi:10.1517/14728222. and facilitates improvement in a rabbit model of Guillain-
2016.1086751. Barré syndrome. Exp Neurol. 2017;291:134–40. doi:10.1016/j.
35. Harness J, McCombe PA. Increased levels of activated T-cells and expneurol.2017.02.010.
reduced levels of CD4/CD25+ cells in peripheral blood of Guillain- 52. Halstead SK, Zitman FM, Humphreys PD, Greenshields K,
Barré syndrome patients compared to controls. J Clin Neurosci. Verschuuren JJ, Jacobs BC, Rother RP, Plomp JJ, Willison HJ.
2008;15(9):1031–5. doi:10.1016/j.jocn.2007.09.016. Eculizumab prevents anti-ganglioside antibody-mediated neuro­
36. Chi LJ, Wang HB, Zhang Y, Wang WZ. Abnormality of circulating pathy in a murine model. Brain. 2008;131(5):1197–208. doi:10.
CD4(+)CD25(+) regulatory T cell in patients with Guillain-Barré 1093/brain/awm316.
syndrome. J Neuroimmunol. 2007;192(1–2):206–14. doi:10.1016/j. 53. Davidson AI, Halstead SK, Goodfellow JA, Chavada G, Mallik A,
jneuroim.2007.09.034. Overell J, Lunn MP, McConnachie A, van Doorn P, Willison HJ.
HUMAN VACCINES & IMMUNOTHERAPEUTICS 7

Inhibition of complement in Guillain-Barré syndrome: the ICA- productio. Faseb J. 2017;31(4):1756–66. doi:10.1096/fj.
GBS study. J Peripher Nerv Syst. 2017;22(1):4–12. doi:10.1111/jns. 201601147R.
12194. 68. Li C, Liu S, Aerqin Q, Shen D, Wu X, Liu K. The therapeutic effects
54. Misawa S, Kuwabara S, Sato Y, Yamaguchi N, Nagashima K, of ginkgolides in Guillain-Barré syndrome and experimental auto­
Katayama K, Sekiguchi Y, Iwai Y, Amino H, Suichi T, et al. immune neuritis. J Clin Neurosci. 2021;87:44–9. doi:10.1016/j.
Safety and efficacy of eculizumab in Guillain-Barré syndrome: jocn.2021.02.016.
a multicentre, double-blind, randomised phase 2 trial. Lancet 69. Chen X, Guo Y, Han R, Liu H, Ding Y, Shi Y, Kong D, Ma X.
Neurol. 2018;17(6):519–29. doi:10.1016/S1474-4422(18)30114-5. Class I PI3K inhibitor ZSTK474 attenuates experimental auto­
55. McGonigal R, Cunningham ME, Yao D, Barrie JA, immune neuritis by decreasing the frequency of Th1/Th17
Sankaranarayanan S, Fewou SN, Furukawa K, Yednock TA, cells and reducing the production of proinflammatory
Willison HJ. C1q-targeted inhibition of the classical complement cytokines. Cell Immunol. 2018;329:41–9. doi:10.1016/j.cel
pathway prevents injury in a novel mouse model of acute motor limm.2018.04.011.
axonal neuropathy. Acta Neuropathol Commun. 2016;4:23. doi:10. 70. Saito E, Kuo R, Pearson RM, Gohel N, Cheung B, King NJC,
1186/s40478-016-0291-x. Miller SD, Shea LD. Designing drug-free biodegradable nanopar­
56. Lansita JA, Mease KM, Qiu H, Yednock T, Sankaranarayanan S, ticles to modulate inflammatory monocytes and neutrophils for
Kramer S. Nonclinical development of ANX005: a humanized ameliorating inflammation. J Control Release. 2019;300:185–96.
anti-C1q antibody for treatment of autoimmune and neurodegen­ doi:10.1016/j.jconrel.2019.02.025.
erative diseases. Int J Toxicol. 2017;36(6):449–62. doi:10.1177/ 71. Elahi E, Ali ME, Zimmermann J, Getts DR, Müller M,
1091581817740873. Lamprecht A. Immune modifying effect of drug free biodegradable
57. Han R, Xiao J, Zhai H, Hao J. Dimethyl fumarate attenuates experi­ nanoparticles on disease course of experimental autoimmune
mental autoimmune neuritis through the nuclear factor neuritis. Pharmaceutics. 2022;14(11):2410. doi:10.3390/
erythroid-derived 2-related factor 2/hemoxygenase-1 pathway by pharmaceutics14112410.
altering the balance of M1/M2 macrophages. J Neuroinflammation. 72. Ren K, Li S, Ding J, Zhao S, Liang S, Cao X, Su C, Guo J. Ginsenoside
2016;13(1):97. doi:10.1186/s12974-016-0559-x. Rd attenuates mouse experimental autoimmune neuritis by modulat­
58. Pitarokoili K, Bachir H, Sgodzai M, Grüter T, Haupeltshofer S, ing monocyte subsets conversion. Biomed Pharmacother.
Duscha A, Pedreiturria X, Motte J, Gold R. Induction of regulatory 2021;138:111489. doi:10.1016/j.biopha.2021.111489.
properties in the intestinal immune system by dimethyl fumarate 73. Jiao H, Ren H. The effects of vasoactive intestinal peptide in the rat
in Lewis rat experimental autoimmune neuritis. Front Immunol. model of experimental autoimmune neuritis and the implications
2019;10:2132. doi:10.3389/fimmu.2019.02132. for treatment of acute inflammatory demyelinating polyradiculo­
59. Fagone P, Mazzon E, Chikovani T, Saraceno A, Mammana S, neuropathy or Guillain-Barré syndrome. Drug Des Devel Ther.
Colletti G, Mangano K, Bramanti P, Nicoletti F. Decitabine induces 2018;12:3817–24. doi:10.2147/DDDT.S175331.
regulatory T cells, inhibits the production of IFN-gamma and IL-17 74. Ambrosius B, Pitarokoili K, Schrewe L, Pedreiturria X, Motte J,
and exerts preventive and therapeutic efficacy in rodent experimental Gold R. Fingolimod attenuates experimental autoimmune neuritis
autoimmune neuritis. J Neuroimmunol. 2018;321:41–8. doi:10.1016/j. and contributes to Schwann cell-mediated axonal protection.
jneuroim.2018.05.013. J Neuroinflammation. 2017;14(1):92. doi:10.1186/s12974-017-
60. Liu RT, Zhang M, Yang CL, Zhang P, Zhang N, Du T, Ge M-R, 0864-z.
Yue L-T, Li X-L, Li H, et al. Enhanced glycolysis contributes to the 75. Shavit-Stein E, Aronovich R, Sylantiev C, Gera O, Gofrit SG,
pathogenesis of experimental autoimmune neuritis. Chapman J, Dori A. Blocking thrombin significantly ameliorates
J Neuroinflammation. 2018;15(1):51. doi:10.1186/s12974-018- experimental autoimmune neuritis. Front Neurol. 2018;9:1139.
1095-7. doi:10.3389/fneur.2018.01139.
61. Zhao Y, Liu B, Wang Y, Xiao B. Effect of fasudil on experi­ 76. Lee KF, Li E, Huber LJ, Landis SC, Sharpe AH, Chao MV,
mental autoimmune neuritis and its mechanisms of action. Jaenisch R. Targeted mutation of the gene encoding the low
Braz J Med Biol Res. 2020;53(1):e8669. doi:10.1590/1414- affinity NGF receptor p75 leads to deficits in the peripheral sen­
431x20198669. sory nervous system. Cell. 1992;69(5):737–49. doi:10.1016/0092-
62. Shi P, Qu H, Nian D, Chen Y, Liu X, Li Q, Li Q, Wang C, Ye M, 8674(92)90286-L.
Ma B. Treatment of Guillain-Barré syndrome with 77. Gonsalvez DG, Tran G, Fletcher JL, Hughes RA, Hodgkinson S,
Bifidobacterium infantis through regulation of T helper cells sub­ Wood RJ, Yoo SW, De Silva M, Agnes WW, McLean C, et al. A
sets. Int Immunopharmacol. 2018;61:290–6. doi:10.1016/j.intimp. brain-derived neurotrophic factor-based p75(NTR) peptide
2018.06.015. mimetic ameliorates experimental autoimmune neuritis induced
63. Shi P, Nian D, Qu H, Ye M, Wang C, Li L, Li Q. B.Infantis axonal pathology and demyelination. eNeuro. 2017;4(3):
enhances immunotherapy for Guillain-Barre syndrome through ENEURO.0142–17.2017. doi:10.1523/ENEURO.0142-17.2017.
regulating PD-1. BMC Neurol. 2023;23(1):48. doi:10.1186/s12883- 78. Moriguchi K, Miyamoto K, Kusunoki S. 4-aminopyridine amelio­
022-03046-w. rates experimental autoimmune neuritis in Lewis rats.
64. Kimura H, Caturegli P, Takahashi M, Suzuki K. New insights into J Neuroimmunol. 2017;305:72–4. doi:10.1016/j.jneuroim.2017.01.
the function of the immunoproteasome in immune and nonim­ 023.
mune cells. J Immunol Res. 2015;2015:541984. doi:10.1155/2015/ 79. Goodfellow JA, Willison HJ. Guillain-Barré syndrome: a century
541984. of progress. Nat Rev Neurol. 2016;12(12):723–31. doi:10.1038/
65. Kloetzel PM, Ossendorp F. Proteasome and peptidase function in nrneurol.2016.172.
MHC-class-I-mediated antigen presentation. Curr Opin 80. Uncini A. Guillain-Barré syndrome: what have we learnt during
Immunol. 2004;16(1):76–81. doi:10.1016/j.coi.2003.11.004. one century? A personal historical perspective. Rev Neurol (Paris).
66. Basler M, Youhnovski N, Van Den Broek M, Przybylski M, 2016;172(10):632–44. doi:10.1016/j.neurol.2016.08.006.
Groettrup M. Immunoproteasomes down-regulate presentation 81. Brettle RP, Gross M, Legg NJ, Lockwood M, Pallis C. Treatment of
of a subdominant T cell epitope from lymphocytic choriomenin­ acute polyneuropathy by plasma exchange. Lancet. 1978;2
gitis virus. J Immunol. 2004;173(6):3925–34. doi:10.4049/jimmu (8099):1100. doi:10.1016/S0140-6736(78)91837-8.
nol.173.6.3925. 82. Greenwood RJ, Newsom-Davis J, Hughes RA, Gordon NS,
67. Liu H, Wan C, Ding Y, Han R, He Y, Xiao J, Hao J. PR-957, Millac P, Newsom-Davis J, Aslan S, Chadwick DW, Mclellan DL,
a selective inhibitor of immunoproteasome subunit low-MW poly­ Stott RB, et al. Controlled trial of plasma exchange in acute
peptide 7, attenuates experimental autoimmune neuritis by sup­ inflammatory polyradiculoneuropathy. Lancet. 1984;1
pressing T(h)17-cell differentiation and regulating cytokine (8382):877–9. doi:10.1016/S0140-6736(84)91341-2.
8 J. YAO ET AL.

83. Osterman PO, Fagius G, Pirskanen G, Fagius J, Pihlstedt P, 85. Kleyweg RP, van der Meché FG, Meulstee J. Treatment of Guillain-
Sidén Å, Säfwenberg J. Beneficial effects of plasma exchange in Barré syndrome with high-dose gammaglobulin. Neurology.
acute inflammatory polyradiculoneuropathy. Lancet. 1984;2 1988;38(10):1639–41. doi:10.1212/WNL.38.10.1639.
(8415):1296–9. doi:10.1016/S0140-6736(84)90819-5. 86. van der Meché FG, Schmitz PI. A randomized trial comparing
84. The Guillain-Barré syndrome Study Group. Plasmapheresis and intravenous immune globulin and plasma exchange in Guillain-
acute Guillain-Barré syndrome. Neurology. 1985;35(8):1096–104. Barré syndrome. Dutch Guillain-Barré study group. N Engl J Med.
doi:10.1212/WNL.35.8.1096. 1992;326(17):1123–9. doi:10.1056/NEJM199204233261705.

You might also like