ArticlePDF AvailableLiterature Review

Abstract and Figures

Purpose of review: After the connection between AS03-adjuvanted pandemic H1N1 vaccine Pandemrix and narcolepsy was recognized in 2010, research on narcolepsy has been more intensive than ever before. The purpose of this review is to provide the reader with current concepts and recent findings on the Pandemrix-associated narcolepsy. Recent findings: After the Pandemrix vaccination campaign in 2009-2010, the risk of narcolepsy was increased 5- to 14-fold in children and adolescents and 2- to 7-fold in adults. According to observational studies, the risk of narcolepsy was elevated for 2 years after the Pandemrix vaccination. Some confounding factors and potential diagnostic biases may influence the observed narcolepsy risk in some studies, but it is unlikely that they would explain the clearly increased incidence in all the countries where Pandemrix was used. An increased risk of narcolepsy after natural H1N1 infection was reported from China, where pandemic influenza vaccination was not used. There is more and more evidence that narcolepsy is an autoimmune disease. All Pandemrix-associated narcolepsy cases have been positive for HLA class II DQB1*06:02 and novel predisposing genetic factors directly linking to the immune system have been identified. Even though recent studies have identified autoantibodies against multiple neuronal structures and other host proteins and peptides, no specific autoantigens that would explain the disease mechanism in narcolepsy have been identified thus far. There was a marked increase in the incidence of narcolepsy after Pandemrix vaccination, especially in adolescents, but also in young adults and younger children. All vaccine-related cases were of narcolepsy type 1 characterized by hypocretin deficiency in the central nervous system. The disease phenotype and the severity of symptoms varied considerably in children and adolescents suffering from Pandemrix-associated narcolepsy, but they were indistinguishable from the symptoms of idiopathic narcolepsy. Narcolepsy type 1 is most likely an autoimmune disease, but the mechanisms have remained elusive.
This content is subject to copyright. Terms and conditions apply.
SLEEP (M THORPY AND M BILLIARD, SECTION EDITORS)
Narcolepsy Associated with Pandemrix Vaccine
Tomi Sarkanen
1,2
&Anniina Alakuijala
2,3
&Ilkka Julkunen
4
&Markku Partinen
2,5
#Springer Science+Business Media, LLC, part of Springer Nature 2018
Abstract
Purpose of Review After the connection between AS03-adjuvanted pandemic H1N1 vaccine Pandemrix and narcolepsy was
recognized in2010, research on narcolepsy has been more intensive than ever before. The purpose of this review is to provide the
reader with current concepts and recent findings on the Pandemrix-associated narcolepsy.
Recent Findings After the Pandemrix vaccination campaign in 20092010, the risk of narcolepsy was increased 5- to 14-fold in
children and adolescents and 2- to 7-fold in adults. According to observational studies, the risk of narcolepsy was elevated for
2 years after the Pandemrix vaccination. Some confounding factors and potential diagnostic biases may influence the observed
narcolepsyrisk in some studies, but it is unlikely that they would explain the clearly increased incidence in all the countries where
Pandemrix was used. An increased risk of narcolepsy after natural H1N1 infection was reported from China, where pandemic
influenza vaccination was not used. There is more and more evidence that narcolepsy is an autoimmune disease. All Pandemrix-
associated narcolepsy cases have been positive for HLA class II DQB1*06:02 and novel predisposing genetic factors directly
linking to the immune system have been identified. Even though recent studies have identified autoantibodies against multiple
neuronal structures and other host proteins and peptides, no specific autoantigens that would explain the disease mechanism in
narcolepsy have been identified thus far.
Summary There was a marked increase in the incidence of narcolepsy after Pandemrix vaccination, especially in adolescents,but
also in young adults and younger children. All vaccine-related cases were of narcolepsy type 1 characterized by hypocretin
deficiency in the central nervous system. The disease phenotype and the severity of symptoms varied considerably in children
and adolescents suffering from Pandemrix-associated narcolepsy, but they were indistinguishable from the symptoms of idio-
pathic narcolepsy. Narcolepsy type 1 is most likely an autoimmune disease, but the mechanisms have remained elusive.
Keywords Narcolepsy .H1N1 vaccination .Va c c i n es .Pandemrix .Hypocretin .Orexin
Introduction
Narcolepsy is a chronic hypersomnia syndrome characterized
by excessive daytime sleepiness, disturbed sleep pattern, and
REM sleep parasomnias such as hypnagogic hallucinations
and sleep paralyzes. In the most recent (3rd version)
International Classification of Sleep Disorders (ICSD-3), the
disease is divided into two different subcategories, narcolepsy
type 1 and type 2 (Table 1)[1]. Narcolepsy type 1 (NT1) is
likely an immune-mediated disease caused by the destruction
of hypocretin-producing neurons in the lateral hypothalamus
resulting in hypocretin deficiency in the central nervous
This article is part of the Topical Collection on Sleep
*Markku Partinen
markpart@me.com; markku.partinen@helsinki.fi
Tomi Sarkanen
tsarkane@gmail.com
Anniina Alakuijala
anniina.alakuijala@hus.fi
Ilkka Julkunen
ilkka.julkunen@utu.fi
1
Department of Neurology, Tampere University Hospital,
Tampere, Finland
2
Department of Clinical Neurosciences, University of Helsinki,
Helsinki, Finland
3
HUS Medical Imaging Center, Department of Clinical
Neurophysiology, Helsinki University Central Hospital,
Helsinki, Finland
4
Institute of Biomedicine, University of Turku, Turku, Finland
5
Helsinki Sleep Clinic, Vitalmed Research Center, Helsinki, Finland
Current Neurology and Neuroscience Reports (2018) 18:43
https://doi.org/10.1007/s11910-018-0851-5
system (CNS). Over 98% of the NT1 patients have a predis-
posing genetic background, positivity for HLA DQB1*06:02
allele. Cataplexy, sudden loss of muscle tone triggered by
emotions, is a common symptom in these patients. In narco-
lepsy type 2 (NT2), hypocretin levels are normal and cata-
plexy is absent. The etiology of NT2 is presently not known.
Narcolepsy is a rare disease. A remarkable increase in the
incidence of NT1 was seen after the H1N1 vaccination cam-
paign in the countries where the Pandemrix vaccine was used
[2••,3]. Pandemrix-related narcolepsy with a clearly recog-
nized environmental trigger increased global research activi-
ties on the pathophysiological mechanism of narcolepsy but
crucial observations explaining the underlying disease mech-
anisms have so far not been identified.
EpidemiologyHow Big Was the Risk
of Pandemrix-Related Narcolepsy?
The most recent pandemic, called swine flu,was caused by
a new reassortant H1N1-type influenza A virus, which ap-
peared first in Mexico and the USA in March 2009. The num-
ber of laboratory-confirmed cases increased rapidly and the
new pandemic virus spread to many other countries via people
traveling. By June 2009, the WHO had already declared that a
new pandemic had started. The previous H1N1 pandemic in
19181919 (Spanish flu) was a devastating disease with a
high mortality rate resulting in 50 to even 100 million deaths
worldwide. In 2009, an early observational study from
Mexico indicated that ca. 6.5% of 900 hospitalized H1N1
infection patients were critically ill and, of those, 41% died
[4]. The mortality seemed to be high especially in children,
young adults, and pregnant women, in contrast to the seasonal
influenza epidemics. Therefore, initially, the first influenza
pandemic of the twenty-first century appeared to be very se-
vere and there was a substantial need for the rapid develop-
ment of an efficient pandemic vaccine.
Eight different pandemic vaccines were used in Europe,
with a coverage of at least 46 million people. Five of these
vaccines had no adjuvant, while two vaccines included MF59
as an adjuvant and one vaccine, namely Pandemrix, had AS03
as an adjuvant. Adjuvants increase the immunogenicity of
antigens, thus allowing the use of lower amounts of immuno-
gens in vaccines for efficient induction of protective immuni-
ty. Pandemrix was the most widely used vaccine in Europe
with more than 30.5 million administered doses [5]. Another
AS03-adjuvanted vaccine, Arepanrix, was used particularly in
Canada. Globally, over 90 million doses of AS03-adjuvanted
H1N1 vaccines were given in different countries [6]. In the
USA, over 90 million doses of pandemic H1N1 vaccines were
administered in 20092010, but they were all non-adjuvant
vaccines.
The first signals of the increased number of narcolepsy
cases after H1N1 vaccination campaigns came from Finland,
Sweden, and France [7••,8,9]. Increased incidence of narco-
lepsy was later found in Norway, the UK, Ireland, and
Germany. The vaccination coverage with Pandemrix was high
in all of these countries [1013]. The risk was first noted in
children and adolescents, and later also in young adults
[1316]. The time period for increased risk is still unclear.
Finnish and Swedish epidemiological (observational) studies
have reported that the time window of an increased risk ex-
tends to 2 years after the vaccination with Pandemrix [15,17].
The reported relative risk of narcolepsy during the first year
after the vaccination varied from 2 to 25 in children and
Table 1 Diagnostic criteria of types 1 and 2 narcolepsy (ICSD-3) [1]
NARCOLEPSY TYPE 1
Criteria A and B must be met
A. The patient has daily periods of irrepressible need to sleep or daytime
lapses into sleep occurring for at least three months.
1
B. The presence of one or both of the following:
1. Cataplexy (as defined under Essential Features) and ameansleep
latency of 8 minutes and two or more sleep onset REM periods
(SOREMPs) on an MSLT performed according to standard techniques.
A SOREMP (within 15 minutes of sleep onset) on the preceding
nocturnal polysomnogram may replace one of the SOREMPs on the
MSLT.
2
2. CSF hypocretin-1 concentration, measured by immunoreactivity, is
either 110 pg/mL or <1/3 of mean values obtained in normal subjects
with the same standardized assay.
NARCOLEPSY TYPE 2
Criteria A-E must be met
A. The patient has daily periods of irrepressible need to sleep or daytime
lapses into sleep occurring for at least three months.
B. A mean sleep latency of 8 minutes and two or more sleep onset
REM periods (SOREMPs) are found on a MSLT performed according
to standard techniques. A SOREMP (within 15 minutes of sleep onset)
on the preceding nocturnal polysomnogram may replace one of the
SOREMPs on the MSLT.
C. Cataplexy is absent.
3
D. Either CSF hypocretin-1 concentration has not been measured or
CSF hypocretin-1 concentration measured by immunoreactivity is
either >110 pg/mL or > 1/3 of mean values obtained in normal subjects
with the same standardized assay.
4
E. The hypersomnolence and/or MSLT findings are not better explained
by other causes, such as insufficient sleep, obstructive sleep apnea,
delayed sleep phase disorder, or the effect of medication or substances
or their withdrawal.
ICSD-3 International Classification of Sleep Disorders, 3rd edition
1
In young children, narcolepsy may sometimes present as excessively
long night sleep or as resumption of previously discontinued daytime
napping
2
If narcolepsy type I is strongly suspected clinically but the MSLT criteria
of B1 are not met, a possible strategy is to repeat the MSLT
3
If cataplexy develops later, then the disorder should be reclassified as
narcolepsy type 1
4
If the CSF Hcrt-1 concentration is tested at a later stage and found to be
either 110 pg/mL or < 1/3 of mean values obtained in normal subjects
with the same assay, then the disorder should be reclassified as narcolepsy
type 1
43 Page 2 of 10 Curr Neurol Neurosci Rep (2018) 18:43
adolescents, and 2 to 9 in adults. In a recent meta-analysis, we
found a 5- to 14-fold increased risk of Pandemrix-related nar-
colepsy in children and adolescents, and a 2- to 7-fold in-
creased risk in adults [2••].
The magnitude of observed risk is partly dependent on the
analytical methods and the criteria concerning how the narco-
lepsy cases were identified. For example, in the Swedish reg-
istry study, where there was no case ascertainment and no
medical record review [15], the risk was lower than in a pre-
vious cohort study [8]. Nonetheless, this study also demon-
strated an increase in the incidence of narcolepsy in young
adults during the second post-vaccination year. It is worth
noting that the 2-year risk window is based on epidemiologi-
cal data. The biologic risk window is not known. In the first
series of patients with Pandemrix-related narcolepsy, the me-
dian delay fromtime of vaccination to onset of narcolepsy was
42 days (0 to 242 days) [18••]. However, it is important to note
that a maximal biologic length of a temporal relationship be-
tween the onset of narcolepsy and a specific immunological
trigger, such as a viral infection or a vaccination, has not been
defined.
Controversies in Epidemiological StudiesIs
the Association True?
The main limitation in the observational studies is that they are
unable to prove a direct causal link between the trigger(s) and
the disease. They are also prone to various biases such as
ascertainment and recall bias and other confounding factors.
It has been speculated that a natural H1N1 influenza infection
per se would have been an important confounding factor con-
tributing to an increased incidence of narcolepsy [1921]. A
3-fold increased risk was reported in Beijing and Shanghai
areas, where pandemic H1N1 vaccines were not used, during
the post-pandemic period [22,23,24]. These findings sug-
gest a temporal association between the H1N1 epidemic peak
and a peak in new narcolepsy cases 3 to 6 months after the
influenza epidemic. However, these results have not been rep-
licated in the neighboring countries or in any other popula-
tions [2••,25]. The vaccination campaign and H1N1 influenza
epidemic occurred almost simultaneously, and, for example,
in Norway and Finland, less than 10% of the population had
been vaccinated prior to the peak of the pandemic [26].
Another study proposed that more than 50% of the vaccinated
individuals in Norway could already have been infected with
the H1N1 virus prior to vaccinations [27]. Conversely, practi-
cally, no serological evidence of natural 2009 pandemic H1N1
virus infection was found in Finnish narcolepsy patients [28].
The manufacturer of Pandemrix, GlaxoSmithKline, has criti-
cized the latter study for its low specificity and the long inter-
val between the vaccination or infection and collection of the
serum samples [29].
Diagnostic bias may be involved if vaccinated subjects
were more likely to be diagnosed as narcolepsy cases than
unvaccinated subjects. There is a possibility that primary
health care practitioners referred vaccinated subjects to special
sleep clinics more often than unvaccinated children/adoles-
cents. However, this seems unlikely, since narcolepsy, wheth-
er associated with vaccine or not, is usually an incapacitating
disease clearly affecting the quality of life. The unequivocal
diagnostic criteria also reduce the risk of diagnostic ascertain-
ment bias.
There is also a possibility for recall bias. This could be
caused by a memory bias of the correct onset time of narco-
lepsy symptoms and their relation to the vaccination time.
Memory bias could happen involuntarily but also knowingly
after the increased media and public awareness of the connec-
tion between H1N1 vaccination and narcolepsy in the hope of
a monetary reimbursement. However, many observational
studies used the first health care contact as an index date and
the results were similar between different study groups and
countries. Proven association of Pandemrix vaccination and
narcolepsy in the countries where media attention and aware-
ness of the association were lower (England, France, Ireland,
and Germany) also makes the recall bias more unlikely. Some
controversies and unsolved issues are summarized in Table 2.
PathophysiologyWhat Is Narcolepsy
and Why Did H1N1 Vaccination Trigger It?
The hallmark of NT1 is hypocretin deficiency. Hypocretin, also
called orexin, is a neuropeptide independently discovered by
two separate research groups, hence the two different names
[33,34]. Soon after these findings, the lack of a functional
hypocretin system as an etiological factor in narcolepsy in
humans was recognized in immunohistochemical studies [35].
All Pandemrix-related, verified, and true narcolepsy cases
have been of NT1. Although some people may have devel-
oped excessive daytime sleepiness along with a NT2 pheno-
type without hypocretin loss after Pandemrix vaccination, the
correlation is currently considered only coincidental.
Biological plausibility between immunological triggers such
as vaccination and NT2 is lacking.
The pathophysiological events leading to the destruction of
hypothalamic hypocretin-producing cells in NT1 are presently
unclear. It has been speculated that especially cell-mediated
autoimmunity, but also autoantibodies against neuronal struc-
tures, or cytotoxicity caused by cytokines and inflammatory
cells in CNS could lead to the destruction of hypocretin-
producing neurons [3,36]. In addition to immune-mediated
destruction of hypocretin neurons, secondary narcolepsy has
been described, for instance, in patients with rare inherited
disorders, brain tumors, traumatic brain injury, and demyelin-
ating disorders which lie outside the scope of this review [37].
Curr Neurol Neurosci Rep (2018) 18:43 Page 3 of 10 43
NT1 is tightly associated with HLA class II allele
DQB1*06:02. More than 98% of NT1 patients are positive
for this allele, making it almost a prerequisite for the disease.
However, this allele is very common in Western countries and
2030% of the general population is positive for this allele.
The relative risk among individuals positive for DQB1*06:02
is ca. 250 times higher than among those individuals who do
not have this allele [38]. The DQB1*06:02 HLA molecule
forms homodimers or heterodimers with another HLA class II
molecule, DQA1*01:02, which also contributes to the relative
risk of NT1 susceptibility [39,40].
HLA class II molecules play a pivotal role in antigen pre-
sentation and cell-mediated immunity. The link between HLA
DQB1*06:02 allele and NT1 is highly indicative of an autoim-
mune background in the disease pathogenesis. Also, HLA class
I alleles and HLA-DP molecules seem to have an independent
role in susceptibility for narcolepsy [41,42]. There are also
multiple protective alleles that reduce the risk of NT1. In the
study by Ollila and coworkers, the most protective heterodimer
was HLA-DPA1*01:03-DPB1*04:02 [41]. HLA-DP loci are
known to have a role in autoimmune diseases such as multiple
sclerosis and type 1 diabetes. Association with HLA class I
alleles also implicates the possibility of the involvement of
cell-mediated cytotoxicity in narcolepsy.
An association with triggering environmental factors such
as H1N1 vaccination, H1N1 virus infection, and streptococcal
infections further strengthens the autoimmune hypothesis in
the pathogenesis of NT1 [3,43]. Nevertheless, no peptides
associated with DQB1*06:02 have been identified; thus, there
is no identification of cross-reactive peptides between micro-
bial pathogens (such as influenza virus or Streptococcus
pyogenes) and human autoantigens, especially those that
could be specific for hypocretin-producing neurons.
In addition, gene polymorphisms in several genes such as
carnitine palmitoyl transferase 1B (CPT1B) and choline kinase
B (CHKB) have been linked to narcolepsy. In some studies, an
increased narcolepsy risk was associated with T cell receptor α
chain gene (TCRA), cathepsin H (CTSH), and purinergic re-
ceptor subtype 2Y11 (P2RY11) (reviewed in [3]). However, the
contribution of gene polymorphisms and other genes apart
from HLA class II genes in narcolepsy susceptibility is rela-
tively low. It is noteworthy that most of the genes associated
with increased narcolepsy risk are involved in the regulation of
the immune system. Thus, these findings further support the
immune-mediated disease mechanisms of NT1.
Moreover, a link between narcolepsy and H1N1 infection
has been seen in immunocompromised mice. Kristensson and
coworkers infected recombinant activating gene 1-deficient
Table 2 Controversies and
unsolved issues in Pandemrix-
related narcolepsy
Factors proving a link between Pandemrix vaccine and narcolepsy
Strong and consistent epidemiological
connection
Increased incidence of narcolepsy shown separately
by different study groups and authorities in all the
countries where the Pandemrix vaccine was used
on a large scale [2••,7••,817].
Immunological connection Higher immune response against H1N1 virus
nucleoprotein in narcoleptic patients than in
healthy controls [30••].
Cross-reactivity between influenza NP and human
hypocretin receptor 2 [31].
Factors affecting robustness of association
Possible biases in observational studies
Confounding by natural H1N1 infection Increased incidence of narcolepsy in China not
related to vaccination [22,23].
Data showing H1N1 virus is capable of entering
into the hypothalamus via the olfactory nerve
and causing narcolepsy-like sleep-wake disruption
in immune-compromised rat [32].
Concomitant circulating H1N1 infection during
the vaccination campaign [27].
Ascertainment, information and recall bias Lack of validation of cases in some studies [2••,7••,821].
Role of increased media attention unclear [2••,7••,821].
Self-reported symptom onset [2••,7••,821].
Missing pathophysiological link
Missing autoantibodies Possible autoantibodies against different neuronal
structures have a low specificity for narcolepsy
and they are also found, though to a lesser extent,
in other sleep disorders or healthy controls.
43 Page 4 of 10 Curr Neurol Neurosci Rep (2018) 18:43
mice, which lacked T and B cells, with the H1N1 influenza A
virus and demonstrated that the infection spread to CNS and
targeted hypocretin-producing neurons in the hypothalamus,
which led to a narcolepsy-like syndrome in these animals [32].
However, most Finnish patients with Pandemrix-associated
narcolepsy were shown not to have antibodies against non-
structural protein 1 (NS1) of the pandemic H1N1 influenza
virus, making it unlikely that the 2009 pandemic H1N1 virus
infection was the causative factor in the development of nar-
colepsy in these patients [28]. NS1 is produced in the host
during the infection and it is not present in influenza vaccines;
thus, anti-NS1 antibodies are found only in humans who suf-
fered a natural influenza A virus infection. This does not rule
out the possibility of a microbial infection being able to trigger
the onset of narcolepsy, but at least in our patients with
Pandemrix-associated narcolepsy, we could not identify any
other environmental factors apart from the Pandemrix vaccine
that was associated with narcolepsy [18••,28].
In addition to a genetic link with factors regulating cell-
mediated immunity and potential narcolepsy-triggering infec-
tions, many groups have identified higher frequencies of au-
toantibodies among narcoleptic patients. These antibodies in-
clude anti-Tribbles homolog 2 and anti-ganglioside antibod-
ies, as well as antibodies against various neuronal structures,
neurotransmitters, and neuron-specific molecules such as
neurexin-1 [44,45]. There is also evidence that hypocretin
receptor 2 may be the target for autoantibodies [31].
Newly diagnosed narcolepsy patients have been shown to
have a higher frequency of anti-Tribbles 2 (TRIB2) homolog
antibodies than control individuals [4649]. The Tribbles fam-
ily of proteins is protein kinase homologs involved in cell
growth regulation, but the precise function of TRIB2 homolog
pseudokinase is not known. Immunoglobulins from narcolep-
sy patients positive for anti-TRIB2 antibodies cause loss of
hypothalamic hypocretin neurons and sleep disturbances
when injected into mice [48]. In addition, TRIB2-
immunized rats displayed autoantibody formation and stain-
ing of hypothalamic structures, including hypocretin neurons.
However, the study failed to demonstrate a direct connection
between anti-TRIB2 antibodies and destruction of hypocretin
neurons, suggesting that the development of anti-TRIB2 anti-
bodies may be a consequence of neuronal damage rather than
the cause [50].
Autoantibodies against neuronal gangliosides have been
linked to some neurological diseases such as the Guillain
Barré syndrome [51]. Recently, Saariaho and coworkers dem-
onstrated that children and adolescent with Pandemrix-
associated narcolepsy have a higher frequency of anti-
human ganglioside GM3 antibodies [45]. An older study
failed to show an association of anti-ganglioside antibodies
and narcolepsy [52], but in that study, the ganglioside pattern
used for autoantibody detection was limited compared to that
in the more recent study. The role of anti-ganglioside
antibodies in the pathogenesis of narcolepsy remains unclear
as it is not known if they contribute to the destruction of
hypocretin neurons or emerge as a consequence of the neuro-
nal damage.
Additional information on the presence of autoantibodies
against neuronal structures in Pandemrix-associated narcolep-
sy was obtained in an immunohistochemical study in rat brain
sections [53]. This study demonstrated that as many as 27%
of patients had autoantibodies against neuronal structures
compared to ca. 10% of healthy controls. A further analysis
revealed that those patients did not have autoantibodies
against hypocretin, but against hypothalamic glutamic acid
isoleucine/α-melanocyte-stimulating hormone (NEI/αMSH),
GABAergic cortical interneurons, and globus pallidus neu-
rons [53]. In the hypothalamus, cells expressing NEI/
αMSH are adjacent to hypocretin neurons. Administration
of NEI/αMSH autoantibody-positive immunoglobulins into
a rat CNS, as in the case of anti-TRIB2 antibodies (see above),
led to neurological symptoms and altered sleep pattern [53].
It was puzzling that the increased frequency of narcolepsy
was associated with the European Pandemrix vaccine and to a
much lesser extent, if at all, with Arepanrix, which was used in
Canada [2••,54]. Since the AS03 adjuvant used in both vac-
cines was produced in the same European factory, the differ-
ences between Pandemrix and Arepanrix are solely in the
H1N1 influenza virus antigen preparation used in the different
vaccines. Vaarala and coworkers showed that the viral nucle-
oprotein (NP) concentration, including polymeric NP, was
higher in Pandemrix than in Arepanrix [30••]. In addition,
immune response against NP was higher in narcoleptic
Pandemrix-vaccinated children than in healthy, matched con-
trol individuals vaccinated with Pandemrix [30••].
Furthermore, antigen absorption experiments pointed out that
the antigen compositions of Pandemrix and Arepanrix are
immunologically different, but the contribution of these dif-
ferences in the development of narcolepsy remains elusive. A
more recent study also confirmed certain differencesin protein
composition and modifications between viral antigens in
Pandemrix and Arepanrix [55].
Molecular mimicry and cross-reactivity between microbial
components and host proteins/molecules may contribute to
the development of autoimmune diseases. A National Center
for Biotechnology Information search for potential cross-
reactive epitopes between Pandemrix viral antigens and host
components led to the identification of a potentially cross-
reactive epitope between influenza NP and human hypocretin
receptor 2 (HCRTR2) [31]. Antibodies against an NP epitope,
which cross-reacted with a similar epitope from human
HCRTR2, were found in sera collected from children and
adolescents who developed narcolepsy after the vaccination
with Pandemrix. The epitope contained seven identical amino
acids within a 20-amino-acid-long structure. Peptide absorp-
tion experiments confirmed the specificity of autoantibodies
Curr Neurol Neurosci Rep (2018) 18:43 Page 5 of 10 43
against the common epitope between NP and HCRTR2 [31].
Nonetheless, sera collected from non-narcoleptic children pri-
or to the 2009 influenza pandemic also showed some autoim-
munity against the cross-reactive epitopes. This raises some
concern regarding the specificity of this cross-reaction as a
causative mechanism in Pandemrix-associated narcolepsy.
Moreover, recent studies demonstrated no anti-HCRTR2-
specific antibodies or a low titer and a very low frequency of
these antibodies in narcoleptic children, suggesting that this
type of autoantibody formation is likely not a general feature
of childhood narcolepsy [56,57]. The situation has recently
become even more complex with the identification of
neurexin-1, methyltransferase-like 22 (METTL22), 5-nucle-
otidase cytosolic IA (NT5C1A) proteins, and the prostaglan-
din D2 receptor being potential autoantigens in narcolepsy.
Patient sera from Pandemrix-associated narcolepsy patients
showed a higher frequency of antibodies against various epi-
topes of those proteins [44,58,59]. However, control indi-
viduals without narcolepsy also showed some autoimmunity
against these proteins.
Numerous studies suggest that the onset of narcolepsy is
genetically linked to several genes, with the HLA class II
system manifesting the highest impact, which implies a role
for cell-mediated immunity in narcolepsy. In addition, various
types of autoantibodies with specificity to hypothalamic cells
and neuronal proteins and peptides propose that humoral im-
munity, too, may contribute to the pathogenesis of narcolepsy.
However, these findings could also be partly explained by a
consequence or a downstream effect of local neuronal cell
death, followed by autoantibody formation. A general inflam-
matory response leading to enhanced cytotoxicity and neuro-
nal damage in CNS is another potential mechanism assisting
the onset of narcolepsy. Figure 1illustrates the potential im-
mune mechanisms contributing to narcolepsy.
There is no evidence of an association between any other
influenza vaccine and narcolepsy. It is possible that there were
other temporary factors, in addition to vaccination, that could
have impaired self-tolerance at the same time. A concurrent
influenza infection was discussed earlier. A Swedish group
found an increased interferon-gamma production against
beta-hemolytic group A streptococcus [60]. We may speculate
that some other factors, such as streptococcal infections, may
have impaired self-tolerance at the time of vaccination.
Furthermore, it has been proposed that at least four other
groups of people are at risk of having autoimmune-like symp-
toms after vaccinations: (a) patients with a history of previous
post-vaccination autoimmune phenomena, (b) patients with a
history of some other autoimmunity, (c) patients with a history
of allergic reactions, and (d) individuals who are prone to
develop autoimmunity (positive family history of autoim-
mune diseases, presence of HLA DQB1*06:02 and/or ab-
sence of HLA DQB1*06:03, etc.) [61]. The latter suggestion
is indirectly supported by findings from our original patient
series, where 17% of patients with Pandemrix-related narco-
lepsy had a history of asthma or atopy, and all patients were
positive for HLA DQB1*06:02 [18••]. A similar connection
was reported in a Spanish study, in which 19% of narcoleptic
patients had one or more associated immune-mediated dis-
eases (odds ratio 3.17 compared to general population) [62].
In contrast, in a French study, only 4.9% of NT1 patients had a
comorbid autoimmune disease, which is an equal prevalence
compared to the general population [63]. In this study, comor-
bid autoimmune diseases were more prevalent in idiopathic
hypersomnia and NT2 than in NT1 [63].
Clinical DescriptionAre There Any
Differences Between Pandemrix-Related
and Idiopathic Narcolepsy?
Some interesting differences have been reported in the
clinical features of Pandemrix-related narcolepsy and
non-vaccine-associated narcolepsy [64,65]. In our expe-
rience, Pandemrix-related NT1 was often characterized by
a rapid onset and short delay between the onset of symp-
toms and diagnosis. A more abrupt onset in Pandemrix-
associated narcolepsy has also been reported in other
studies [18••,65,6668].
Facial hypotonia and cataplectic faciesare increasingly
recognized features in childhood cataplexy. Facial hypotonia
andtongueprotrusionwerereportedtobemorecommonin
children with Pandemrix-related narcolepsy than in those with
idiopathic narcolepsy [68]. The incidence of childhood narco-
lepsy peaks among 1020-year-olds and an early onset in
preschool-aged children is very rare. One striking feature of
vaccine-associated narcolepsy was the emergence of cases
also at preschool age [7••,18••,67,69]. On the other hand,
a few studies have reported a higher mean age at the time of
diagnosis or onset of symptoms in Pandemrix-associated nar-
colepsy [18••,64].
In a comparison between unvaccinated Italian and vacci-
nated Finnish children with narcolepsy, no significant differ-
ences were seen, except for more disturbed sleep character-
istics in Finnish subjects [64]. Even if the onset were more
abrupt, the fully developed clinical picture of Pandemrix-
associated narcolepsy seems to be very similar to idiopathic
narcolepsy [65]. However, the clinical picture of NT1 is
very heterogeneous. Some subjects with Pandemrix-
associated NT1 may manage even without any medication
and be able to fully continue to work or study, while others
are severely handicapped [65]. Objective polysomnographic
and actigraphic characteristics seem to be very similar be-
tween vaccine- and non-vaccine-associated narcolepsy [70].
There were some differences in sleepwake rhythm param-
eters implying an earlier sleep phase in Pandemrix-
associated narcolepsy [70].
43 Page 6 of 10 Curr Neurol Neurosci Rep (2018) 18:43
We have seen some, fortunately only a few, Pandemrix-
associated NT1 subjects developing very severe psychiatric
symptoms. Some of these psychiatric symptoms are so dev-
astating that they resemble psychosis, KlüverBucy syn-
drome, or autoimmune encephalitis. We have screened for a
large repertoire of neuronal autoantibodies in these patients
but failed to find any conclusive evidence for a CNS-
specific autoimmunity.
Treatment
Symptomatic treatment of Pandemrix-associated narcolepsy
mirrors the treatment of idiopathic narcolepsy using, for ex-
ample, modafinil, methylphenidate, and sodium oxybate.
Immunomodulatory treatment has not proven to be effective
in the treatment of Pandemrix-related narcolepsy but the evi-
dence is limited. We have experience of five Finnish post-
Pandemrix patients with intravenous immunoglobulin (IVIg)
treatment administered within a few months of the disease
onset. None of the patients benefited from the treatment.
Danish colleagues have reported similar results [71]. One re-
cent report suggested that an early combined immunotherapy
with methylprednisolone and IVIg may have had a positive
effect on relieving symptoms, but the results have to be taken
cautiously since we know that the natural course of narcolepsy
is very heterogeneous [65,72].
There is a case report that provides a spark of hope for
immune therapy in narcolepsy [73]. The authors demonstrated
the normalization of hypocretin levels and remission of sleep-
iness and cataplexy in a patient treated with IVIg very early
after the symptom onset [73]. Unfortunately, the treatment
effect lasted only for a few months and the subject refused
further treatment. We have reported a patient with a dramatic,
but again unfortunately temporary, improvement of symptoms
after treatment with rituximab [74]. The largest study so far on
the immunomodulatory treatment in pediatric narcolepsy is a
non-randomized controlled open-label trial in France [75]. In
this study, a subset of patients with the highest scores in the
narcolepsy screening questionnaire Ullanlinna Narcolepsy
Scale (UNS) had remission of symptoms sooner than those
who were not treated with IVIg.
Fig. 1 A hypothetical model of autoimmunity in narcolepsy induced by
influenza A virus infection or vaccination or streptococcal infection.
Influenza A virus infection (or S. pyogenes infection) or vaccination
with AS03-adjuvanted pandemic influenza vaccine leads to an infection
or an uptake of viral antigens by dendritic cells (DCs) in peripheral
tissues. This leads to the maturation and migration of infected or
antigen-loaded DCs to local lymph nodes. In the lymph nodes, DCs
present influenza-specific epitopes to CD4- and CD8-positive T cells.
CD4 helper T cells provide help and activation of B cells that have also
taken up soluble viral antigens. Activated T cells and matured IgG-
producing B cells migrate via blood into the central nervous system. In
the brain, in the event that cross-reactive cell-mediated and humoral
immunity has developed, autoimmunity is involved in the destruction
of hypocretin-producing neurons in the hypothalamus. Potentially, three
mechanisms may be involved: (1) activated CD8 T cells destroy neurons
via cytotoxic mechanisms; (2) inflammatory cytokines, produced by
activated T cells, induce cellular cytotoxicity; or (3) autoantibodies,
which recognize cross-reactive epitopes on neurons, induce antibody-
dependent cytotoxicity
Curr Neurol Neurosci Rep (2018) 18:43 Page 7 of 10 43
Taken together, the attempts to utilize immunomodulatory
treatment in narcolepsy have provided controversial results. It
is possible that immune therapies should be administered very
soon after the disease onset, before the permanent damage to
hypocretin system has taken place. The pathogenic cascade
leading to narcolepsy may begin months or even years before
the actual symptoms emerge; thus, in the majority of cases, it
may already be too late when the patient enters the clinic. A
multicenter randomized controlled trial of immunotherapy
near the disease onset might provide an answer to this ques-
tion. In the future, once the autoantigens have been identified
and the disease mechanisms resolved, immune therapy may
become an efficient therapy.
Conclusions
Several studies from different countries using alternative
methods have confirmed the association between narcolepsy
and Pandemrix vaccination. This provides strong evidence of a
true association even if the possible diagnostic biases may some-
what reduce the risk. The classic criteria for an autoimmune
disease are not fully met in the case of narcolepsy, but increasing
evidence suggests an immune-mediated mechanism in the dis-
ease pathogenesis. It is also important to state that seasonal in-
fluenza vaccines, which are given to hundreds of millions of
individuals every year, have not been associated with narcolepsy.
Research on narcolepsy was remarkably enhanced after
Pandemrix-related narcolepsy was identified in 2010 and the
work, for good reason, continues intensively. A conclusive
explanation for the disease mechanism(s) has still remained
elusive. Hopefully, active research efforts on understanding its
pathogenesis may lead to the revelation of the etiology of
narcolepsy, which provides us with better means to prevent
similar events in the future. There is also a great demand for
novel immunotherapeutic and other treatment modalities.
Acknowledgments We thank Damon Tringham for linguistic advice.
Compliance with Ethical Standards
Conflict of Interest Tomi Sarkanen reports a grant from The Finnish
Medical Council, personal fees from Orion, and travel expenses from
UCB, outside the submitted work.
Anniina Alakuijala reports no conflict of interest.
Ilkka Julkunen reports a research grant from the Academy of Finland,
outside the submitted work.
Markku Partinen reports research grants from the Academy of
Finland, personal fees from UCB-Pharma, GSK, Takeda, MSD, Orion,
and participating in clinical trials by Bioprojet, Jazz Pharmaceuticals, and
MSD, all outside the submitted work.
Human and Animal Rights and Informed Consent This article does not
contain any studies with human or animal subjects performed by any of
the authors.
References
Papers of particular interest, published recently, have been
highlighted as:
Of importance
•• Of major importance
1. American Academy of Sleep Medicine. International classification
of sleep disorders. 3rd ed. Darien: American Academy of Sleep
Medicine; 2014.
2.•• Sarkanen T, Alakuijala A, Dauvilliers Y, Partinen M. Incidence of
narcolepsy after H1N1 influenza and vaccinations: systematic re-
view and meta-analysis. Sleep Med Rev. 2017; https://doi.org/10.
1016/j.smrv.2017.06.006.A comprehensive review and meta-
analysis of the observational studies reporting an association
with Pandemrix and narcolepsy. In addition, biases affecting
the risk and lack of an association with other vaccines and
narcolepsy are discussed.
3. Partinen M, Kornum BR, Plazzi G, Jennum P, Julkunen I, Vaarala
O. Narcolepsy as an autoimmune disease: the role of H1N1 infec-
tion and vaccination. Lancet Neurol. 2014;13(6):60013.
4. Dominguez-Cherit G, Lapinsky SE, Macias AE, Pinto R, Espinosa-
Perez L, de la Torre A, et al. Critically ill patients with 2009 influ-
enza A(H1N1) in Mexico. JAMA. 2009;302(17):18807.
5. European Centre for Disease Prevention and Control. Narcolepsy in
association with pandemic influenza vaccination (a multi-country
European epidemiological investigation). Stockholm 2012. https://
ecdc.europa.eu/sites/portal/files/media/en/publications/
Publications/Vaesco%20report%20FINAL%20with%20cover.pdf.
Accessed 13 Nov 2017.
6. GlaxoSmithKline. GSK Public policy positions. Pandemic pre-
paredness. 2014. https://www.gsk.com/media/2951/pandemic-
preparedness-policy.pdf. Accessed 13 Nov 2017.
7.•• Nohynek H, Jokinen J, Partinen M, Vaarala O, Kirjavainen T,
Sundman J, et al. AS03 adjuvanted AH1N1 vaccine associated with
an abrupt increase in the incidence of childhood narcolepsy in
Finland. PLoS One. 2012;7(3):e33536. The first epidemiological
study reporting an increased incidence of narcolepsy after vac-
cination with Pandemrix.
8. Medical Products Agency. A registry based comparative cohort
study in four Swedish counties of the risk for narcolepsy after vac-
cination with Pandemrixa first and preliminary report. 2011.
https://lakemedelsverket.se/upload/nyheter/2011/
PandemrixRegReport110328.pdf. Accessed 13 Nov 2017.
9. Dauvilliers Y, Arnulf I, Lecendreux M, Monaca Charley C, Franco
P, Drouot X, et al. Increased risk of narcolepsy in children and
adults after pandemic H1N1 vaccination in France. Brain.
2013;136(Pt 8):248696.
10. Heier MS, Gautvik KM, Wannag E, Bronder KH, Midtlyng E,
Kamaleri Y, et al. Incidence of narcolepsy in Norwegian children
and adolescents after vaccination against H1N1 influenza A. Sleep
Med. 2013;14(9):86771.
11. Miller E, Andrews N, Stellitano L, Stowe J, Winstone AM,
Shneerson J, et al. Risk of narcolepsy in children and young people
receiving AS03 adjuvanted pandemic A/H1N1 2009 influenza vac-
cine: retrospective analysis. BMJ. 2013;346:f794.
12. OFlanagan D, Barret AS, Foley M, Cotter S, Bonner C, Crowe C,
et al. Investigation of an association between onset of narcolepsy
and vaccination with pandemic influenza vaccine, Ireland April
2009-December 2010. Euro Surveill. 2014;19(17):1525.
13. Oberle D, Pavel J, Mayer G, Geisler P, Keller-Stanislawski B.
Retrospective multicenter matched case-control study on the risk
factors for narcolepsy with special focus on vaccinations (including
43 Page 8 of 10 Curr Neurol Neurosci Rep (2018) 18:43
pandemic influenza vaccination) and infections in Germany. Sleep
Med. 2017;34:7183.
14. Jokinen J, Nohynek H, Honkanen J, Vaarala O, Partinen M, Hublin C
et al. Pandemiarokotteen ja narkolepsian yhteys aikuisilla -
Varmennettuihin rekisteritietoihin perustuva kohorttitutkimus. 2013.
http://urn.fi/URN:ISBN:978-952-245-921-3. Accessed 13 Nov 2017.
15. Persson I, Granath F, Askling J, Ludvigsson JF, Olsson T, Feltelius
N. Risks of neurological and immune-related diseases, including
narcolepsy, after vaccination with Pandemrix: a population- and
registry-based cohort study with over 2 years of follow-up. J
Intern Med. 2014;275(2):17290.
16. Stowe J, Miller E, Andrews N, Kosky C, Leschziner G, Shneerson
JM, et al. Risk of narcolepsy after AS03 adjuvanted pandemic A/
H1N1 2009 influenza vaccine in adults: a case-coverage study in
England. Sleep. 2016;39(5):10517.
17. Jokinen J, Nohynek H, Vaarala O, Kilpi T. Pandemiarokotteen ja
narkolepsian yhteys - vuoden 2012 loppuun mennessä kertyneisiin
rekisteritietoihin perustuva seurantaraportti. 2014. http://urn.fi/
URN:ISBN:978-952-302-255-3. Accessed 13 Nov 2017.
18.•• Partinen M, Saarenpaa-Heikkila O, Ilveskoski I, Hublin C, Linna M,
Olsen P, et al. Increased incidence and clinical picture of childhood
narcolepsy following the 2009 H1N1 pandemic vaccination cam-
paign in Finland. PLoS One. 2012;7(3):e33723. The first article
reporting the clinical picture and increased incidence of narco-
lepsy after vaccination with Pandemrix in the Nordic countries.
19. Verstraeten T, Cohet C, Santos GD, Ferreira GL, Bollaerts K, Bauchau
V, et al. Pandemrix and narcolepsy: a critical appraisal of the observa-
tional studies. Hum Vaccin Immunother. 2016;12(1):18793.
20. Bollaerts K, Shinde V, Dos Santos G, Ferreira G, Bauchau V, Cohet
C, et al. Application of probabilistic multiple-bias analyses to a
cohort- and a case-control study on the association between
Pandemrix and narcolepsy. PLoS One. 2016;11(2):e0149289.
21. Sturkenboom MCJM. The narcolepsy-pandemic influenza story: can
the truth ever be unraveled? Vaccine. 2015;33(Supplement 2):B6B13.
22.Han F, Lin L, Warby SC, Faraco J, Li J, Dong SX, et al. Narcolepsy
onset is seasonal and increased following the 2009 H1N1 pandemic
in China. Ann Neurol. 2011;70(3):4107. The authors report a 3-
fold increase of narcolepsy in the Beijing area in China where
no pandemic vaccines were used. A similar finding was seen in
the Shanghai area, but not in any other country.
23. Han F, Lin L, Li J, Dong XS, Mignot E. Decreased incidence of
childhood narcolepsy 2 years after the 2009 H1N1 winter flu pan-
demic. Ann Neurol. 2013;73(4):560.
24. Wu H, Zhuang J, Stone WS, Zhang L, Zhao Z, Wang Z, et al.
Symptoms and occurrences of narcolepsy: a retrospective study of
162 patients during a 10-year period in eastern China. Sleep Med.
2014;15(6):60713.
25. Choe YJ, Bae GR, Lee DH. No association between influenza
A(H1N1)pdm09 vaccination and narcolepsy in South Korea: an
ecological study. Vaccine. 2012;30(52):743942.
26. Gil Cuesta J, Aavitsland P, Englund H, Gudlaugsson O, Hauge SH,
Lyytikainen O, et al. Pandemic vaccination strategies and influenza
severe outcomes during the influenza A(H1N1)pdm09 pandemic
and the post-pandemic influenza season: the Nordic experience.
Euro Surveill. 2016; https://doi.org/10.2807/1560-7917.ES.2016.
21.16.30208.
27. Van Effelterre T, Dos Santos G, Shinde V. Twin peaks: A/H1N1
pandemic influenza virus infection and vaccination in Norway,
2009-2010. PLoS One. 2016;11(3):e0151575.
28. Melen K, Partinen M, Tynell J, Sillanpaa M, Himanen SL,
Saarenpaa-Heikkila O, et al. No serological evidence of influenza
A H1N1pdm09 virus infection as a contributing factor in childhood
narcolepsy after Pandemrix vaccination campaign in Finland.PLoS
One. 2013;8(8):e68402.
29. Innis B, Boutet P. Comment on: Melen K, Partinen M, Tynell J,
Sillanpaa M, Himanen S-L, et al. No serological evidence of
influenza A H1N1pdm09 virus infection as a contributing factor
in childhood narcolepsy after pandemrix vaccination campaign in
Finland. PLoS One. 2014. http://journals.plos.org/plosone/article/
comment?id=10.1371/annotation/eab58001-4872-4b9d-94d7-
c85a16b4b6d6. Accessed 13 Nov 2017.
30.•• Vaarala O, Vuorela A, Partinen M, Baumann M, Freitag TL, Meri
S, et al. Antigenic differences between AS03 adjuvanted influenza
A (H1N1) pandemic vaccines: implications for Pandemrix-
associated narcolepsy risk. PLoS One. 2014;9(12):e114361. A
study reporting differences in nucleoprotein (NP) of the viral
component in Pandemrix and Arepanrix. Furthermore, in-
creased antibody response against NP was seen in subjects with
the HLA DQB1*06:02 narcolepsy risk allele.
31. Ahmed SS, Volkmuth W, Duca J, Corti L, Pallaoro M, Pezzicoli A,
et al. Antibodies to influenza nucleoprotein cross-react with human
hypocretin receptor 2. Sci Transl Med. 2015;7(294):294ra105.
32. Tesoriero C, Codita A, Zhang MD, Cherninsky A, Karlsson H,
Grassi-Zucconi G, et al. H1N1 influenza virus induces
narcolepsy-like sleep disruption and targets sleep-wake regulatory
neurons in mice. Proc Natl Acad Sci U S A. 2016;113(3):E36877.
33. Sakurai T, AmemiyaA, Ishii M, MatsuzakiI, Chemelli RM, Tanaka
H, et al. Orexins and orexin receptors: a family of hypothalamic
neuropeptides and g protein-coupled receptors that regulate feeding
behavior. Cell. 1998;92:57385.
34. de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, et
al. The hypocretins: hypothalamus-specific peptides with
neuroexcitatory activity. Proc Natl Acad Sci U S A. 1998;95(1):3227.
35. Peyron C, Faraco J, Rogers W, Ripley B, Overeem S, Charnay Y, et
al. A mutation in a case of early onset narcolepsy and a generalized
absence of hypocretin peptides in human narcoleptic brains. Nat
Med. 2000;6(9):9917.
36. Julkunen I, Partinen M. Neuroimmunology: disease mechanisms in
narcolepsy remain elusive. Nat Rev Neurol. 2014;10(11):6167.
37. Nishino S, Kanbayashi T. Symptomatic narcolepsy, cataplexy and
hypersomnia, and their implications in the hypothalamic
hypocretin/orexin system. Sleep Med Rev. 2005;9(4):269310.
38.Tafti M, Hor H, Dauvilliers Y, Lammers GJ, Overeem S, Mayer G,
et al. DQB1 locus alone explains most of the risk and protection in
narcolepsy with cataplexy in Europe. Sleep. 2014;37(1):1925. A
large genome-wide association (GWAS) study with over 1200
narcolepsy patients and 3500 controls. It was shown that the
majority of the genetic risk of narcolepsy is explained by the
HLA DQB1 locus only. HLA DQB1*06:02 positive individuals
carry a 251-fold increased risk of developing narcolepsy.
39. Hor H, Bartesaghi L, Kutalik Z, Vicario JL, de Andres C, Pfister C,
et al. A missense mutation in myelin oligodendrocyte glycoprotein
as a cause of familial narcolepsy with cataplexy. Am J Hum Gen.
2011;89(3):4749.
40. Mignot E, Lin L, Rogers W, Honda Y, Qiu X, Lin X, et al. Complex
HLA-DR and -DQ interactions confer risk of narcolepsy-cataplexy
in three ethnic groups. Am J Hum Gen. 2001;68(3):68699.
41.Ollila HM, Ravel JM, Han F, et al. HLA-DPB1 and HLA class I
confer risk of and protection from narcolepsy. Am J Hum Genet.
2015;96:13646. One of the first reports to find HLA class I
associations in narcolepsy.
42.Tafti M, Lammers GJ, Dauvilliers Y, et al. Narcolepsy-associated
HLA class I alleles implicate cell-mediated cytotoxicity. Sleep.
2016;39:5817. One of the first reports to find HLA class I
associations in narcolepsy.
43. Aran A, Lin L, Nevsimalova S, Plazzi G, HongSC, Weiner K, et al.
Elevated anti-streptococcal antibodies in patients with recent narco-
lepsy onset. Sleep. 2009;32(8):97983.
44. Haggmark-Manberg A, Zandian A, Forsstrom B, Khademi M,
Lima Bomfim I, Hellstrom C, et al. Autoantibody targets in
vaccine-associated narcolepsy. Autoimmunity. 2016;49(6):42133.
Curr Neurol Neurosci Rep (2018) 18:43 Page 9 of 10 43
45. Saariaho AH, Vuorela A, Freitag TL, Pizza F, Plazzi G, Partinen M,
et al. Autoantibodies against ganglioside GM3 are associated with
narcolepsy-cataplexy developing after Pandemrix vaccination
against 2009 pandemic H1N1 type influenza virus. J Autoimmun.
2015;63:6875.
46. Cvetkovic-Lopes V, Bayer L, Dorsaz S, Maret S, Pradervand S,
Dauvilliers Y, et al. Elevated Tribbles homolog 2-specific antibody
levels in narcolepsy patients. J Clin Invest. 2010;120(3):7139.
47. Kawashima M, Lin L, Tanaka S, Jennum P, Knudsen S,
Nevsimalova S, et al. Anti-Tribbles homolog 2 (TRIB2) autoanti-
bodies in narcolepsy are associated with recent onset of cataplexy.
Sleep. 2010;33(7):86974.
48. Katzav A, Arango MT, Kivity S, Tanaka S, Givaty G, Agmon-
Levin N, et al. Passive transfer of narcolepsy: anti-TRIB2 autoan-
tibody positive patient IgG causes hypothalamic orexin neuron loss
and sleep attacks in mice. J Autoimmun. 2013;45:2430.
49. Lind A, Ramelius A, Olsson T, Arnheim-Dahlstrom L, Lamb F,
Khademi M, et al. A/H1N1 antibodies and TRIB2 autoantibodies
in narcolepsy patientsdiagnosed in conjunction with the Pandemrix
vaccination campaign in Sweden 2009-2010. J Autoimmun.
2014;50:99106.
50. Tanaka S, Honda Y, Honda M, Yamada H, Honda K, Kodama T.
Anti-tribbles pseudokinase 2 (TRIB2)-immunization modulates
hypocretin/orexin neuronal functions. Sleep. 2017; https://doi.org/
10.1093/sleep/zsw036.
51. Goodfellow JA, Willison HJ. Antiganglioside, antiganglioside-
complex, and antiglycolipid-complex antibodies in immune-
mediated neuropathies. Curr Opin Neurol. 2016;29(5):57280.
52. Overeem S, Geleijns K, Garssen MP, Jacobs BC, van Doorn PA,
Lammers GJ. Screening for anti-ganglioside antibodies in hypocretin-
deficient human narcolepsy. Neurosci Lett. 2003;341(1):136.
53.BergmanP,AdoriC,VasS,Kai-LarsenY,SarkanenT,Cederlund
A, et al. Narcolepsy patients have antibodies that stain distinct cell
populations in rat brainand influence sleep patterns. Proc Natl Acad
Sci U S A. 2014;111(35):E373544. In this study, 27% of narco-
lepsy patients showed autoantibodies against three different
neuronal structures in immunohistochemical stainings on rat
brain. Although staining against hypocretin neurons was not
seen, autoantibodies against hypothalamic NEI/αMSH neu-
rons were found. Passive transfer of sera from NEI/αMSH-
positive subject into a rat brain produced disturbed REM and
slow wave sleep patterns.
54. Montplaisir J, Petit D, Quinn M-J, Ouakki M, Deceuninck G,
Desautels A, et al. Risk of narcolepsy associated with inactivated
adjuvanted (AS03) A/H1N1 (2009) pandemic influenza vaccine in
Quebec. PLoS One. 2014;9(9):e108489.
55. Jacob L, Leib R, Ollila HM, Bonvalet M, Adams CM, Mignot E.
Comparison of Pandemrix and Arepanrix, two pH1N1 AS03-
adjuvanted vaccines differentially associated with narcolepsy de-
velopment. Brain Behav Immun. 2015;47:4457.
56. Luo G, Lin L, Jacob L, Bonvalet M, Ambati A, Plazzi G, et al.
Absence of anti-hypocretin receptor 2 autoantibodies in post
pandemrix narcolepsy cases. PLoS ONE. 12(12):e0187305.
https://doi.org/10.1371/journal.pone.0187305.
57. Giannoccaro MP, Waters P, Pizza F, Liguori R, Plazzi G, Vincent A.
Antibodies against hypocretin receptor 2 are rare in narcolepsy.
Sleep. 2017;40 https://doi.org/10.1093/sleep/zsw056.
58.Zandian A, Forsstrom B, Haggmark-Manberg A, Schwenk JM,
Uhlen M, Nilsson P, et al. Whole-proteome peptide microarrays
for profiling autoantibody repertoires within multiple sclerosis
and narcolepsy. J Proteome Res. 2017;16(3):130014. The au-
thors presented a novel method for screening autoantibodies,
namely high-density peptide arrays. They recognized a poten-
tial new autoantigen neurexin 1 in narcolepsy.
59.Sadam H, Pihlak A, Kivil A, Pihelgas S, Jaago M, Adler P, et al.
Prostaglandin D2 receptor DP1 antibodies predict vaccine-induced
and spontaneous narcolepsy type 1: large-scale study of antibody
profiling. EBioMedicine. 2018;29:4759. The authors describe a
potentially new autoantigen prostaglandin D2 receptor against
which narcolepsy patient have developed autoantibodies.
60. Ambati A, Poiret T, Svahn BM, Valentini D, Khademi M, Kockum
I, et al. Increased beta-haemolytic group A streptococcal M6 sero-
type and streptodornase B-specific cellular immune responses in
Swedish narcolepsy cases. J Intern Med. 2015;278(3):26476.
61. Soriano A, Nesher G, Shoenfeld Y. Predicting post-vaccination au-
toimmunity: who might be at risk? Pharmacol Res. 2015;92:1822.
62. Martinez-Orozco FJ, Vicario JL, Villalibre-Valderrey I, De Andres
C, Fernandez-Arquero M, Peraita-Adrados R. Narcolepsy with cat-
aplexy and comorbid immunopathological diseases. J Sleep Res.
2014;23(4):4149.
63. Barateau L, Lopez R, Arnulf I, Lecendreux M, Franco P, Drouot X,
et al. Comorbidity between central disorders of hypersomnolence
and immune-based disorders. Neurology. 2017;88(1):93100.
64. Pizza F, Peltola H, Sarkanen T, Moghadam KK, Plazzi G, Partinen
M. Childhood narcolepsy with cataplexy: comparison between
post-H1N1 vaccination and sporadic cases. Sleep Med.
2013;15(2):2625.
65.Sarkanen T, Alakuijala A, Partinen M. Clinical course of H1N1-
vaccine-related narcolepsy. Sleep Med. 2016;19:1722. The first
longitudinal study of the clinical phenotype of Pandemrix-
related narcolepsy. The symptoms of narcolepsy and severity
of the disease seem very heterogeneous.
66. Szakacs A, Darin N, Hallbook T. Increased childhood incidence of
narcolepsy in western Sweden after H1N1 influenza vaccination.
Neurology. 2013;80(14):131521.
67. Medical Products Agency. Occurrence of narcolepsy with cata-
plexy among children and adolescents in relation to the H1N1 pan-
demic and Pandemrix vaccinationsresults of a case inventory
studybytheMPAinSwedenduring20092010. https://
lakemedelsverket.se/upload/nyheter/2011/Fallinventeringsrapport_
pandermrix_110630.pdf. Accessed 13 Nov 2017.
68. Winstone AM, Stellitano L, Verity C, Andrews N, Miller E, Stowe
J, et al. Clinical features of narcolepsy in children vaccinated with
AS03 adjuvanted pandemic A/H1N1 2009 influenza vaccine in
England. Dev Med Child Neurol. 2014;56(11):111723.
69. ECDC comment. Update on narcolepsy cases associated with
Pandemrix vaccination in 2009 in the Netherlands. https://ecdc.
europa.eu/en/news-events/update-narcolepsy-cases-associated-
pandemrix-vaccination-2009-netherlands. Accessed 13 Nov 2017.
70. Alakuijala A, Sarkanen T, Partinen M. Polysomnographic and
actigraphic characteristics of patients with H1N1-vaccine-related
and sporadic narcolepsy. Sleep Med. 2015;16(1):3944.
71. Knudsen S, Biering-Sorensen B, Kornum BR, Petersen ER,
Petersen ER, Ibsen JD, et al. Early IVIg treatment has no effect
on post-H1N1 narcolepsy phenotype or hypocretin deficiency.
Neurology. 2012;79(1):1023.
72. Viste R, Soosai J, Vikin T, Thorsby PM, Nilsen KB, Knudsen S.
Long-term improvement after combined immunomodulation in ear-
ly post-H1N1 vaccination narcolepsy. Neurol Neuroimmunol
Neuroinflamm. 2017;4(5):e389.
73. Dauvilliers Y, Abril B, Mas E, Michel F, Tafti M. Normalization of
hypocretin-1 in narcolepsy after intravenous immunoglobulin treat-
ment. Neurology. 2009;73(16):13334.
74. Sarkanen T, Alen R, Partinen M. Transient impact of rituximab in
H1N1 vaccination-associated narcolepsy with severe psychiatric
symptoms. Neurologist. 2016;21(5):856.
75. Lecendreux M, Berthier J, Corny J, Bourdon O, Dossier C,
Delclaux C. Intravenous immunoglobulin therapy in pediatric nar-
colepsy: a nonrandomized, open-label, controlled, longitudinal ob-
servational study. J Clin Sleep Med. 2017;13(3):44153.
43 Page 10 of 10 Curr Neurol Neurosci Rep (2018) 18:43
... Children affected by narcolepsy after the vaccination were reported to have a more sudden onset of the disease compared to previously known cases. Sweden had a high vaccination rate of around 60% of the population, and the subsequent increase in narcolepsy was eventually interpreted as an effect of the extensive Pandemrix vaccinations [29][30][31][32]. Notably, in China, where no vaccinations were performed, studies also indicated an increase in seasonal narcolepsy incidence, most likely due to the H1N1 infection [33]. ...
Article
Full-text available
(1) Background: In the context of the H1N1 pandemic and the Pandemrix vaccination campaign, an increased number of narcolepsy cases were noted in several countries. In Sweden, this phenomenon was attributed to the effect of the Pandemrix vaccination in the first place. Studies from China indicated that narcolepsy could occur as a consequence of the H1N1 infection itself. We performed an analysis of the increase, with a specific interest in age and sex distribution. We also aimed to validate the origin of the excess cases, post hoc. (2) Methods: Data for narcolepsy patients (ICD code G 47.4, both type 1 and type 2) distributed by sex and age at 5-year intervals, annually between 2005 and 2017, were retrieved from the National Patient Register. Information on the total population was collected from the Swedish Population Register. (3) Results: The number of narcolepsy cases increased markedly from 2009 to 2014 compared to the period before 2009. A particular increase in 2011 among children and teenagers was observed. The sex ratio did not change significantly during the study period. (4) Conclusions: Our results support an association between the increased prevalence of narcolepsy cases and Pandemrix vaccination, but the effect of the virus itself cannot be ruled out as a contributing factor.
... In the context of the H1N1 pandemic and the Pandemrix vaccination in 2009-2010, an increased number of narcolepsy cases were noted in several countries (28). Sweden had a high vaccination rate of around 60% of the population and the subsequent increase in narcolepsy was eventually interpreted as an effect of the extensive Pandemrix vaccinations (29)(30)(31)(32). Notably, in China, where no vaccinations were performed, studies indicated an increase in seasonal narcolepsy incidence as well, most probably due to the H1N1 infection (33). ...
Preprint
Full-text available
In the context of the H1N1 pandemic and the Pandemrix vaccination campaign, an increased number of narcolepsy cases were noted in several countries. In Sweden, this phenomenon was attributed to the effect of the Pandemrix vaccination in the first place. Interestingly, Denmark, a neighboring country, also had an increased incidence during this period, despite a low vaccination rate. Additionally, studies from China indicated that narcolepsy could occur as a consequence of the H1N1 infection itself. Against these background factors, and due to the clinical impression that narcolepsy patients who were vaccinated tend to dominate in the Swedish narcolepsy polyclinic, we performed an analysis of the increase with a specific interest in age and sex distribution. We also aimed to validate the origin of the excess cases, post hoc. Materials & Methods: The data for narcolepsy patients (ICD code G 47.4) distributed by sex and age at 5-year intervals, annually between 2005 and 2017, was retrieved from the National Patient Register. Information on the total population was collected from the Swedish Population Register and was used for calculations of the narcolepsy prevalence. Results: The number of narcolepsy cases increased markedly from 2009 to 2014 compared to the period before 2009. A particular increase in 2011 among children and teenagers was observed, which resulted in increased cases among the age group 20-39 years in the following years. The sex ratio did not change significantly during the study period. The excess cases found in our study exceeded the number of patients who so far (December 2023) received economic compensation for vaccine side effects (n=450). Conclusions: Our results confirm an association between the increased prevalence of narcolepsy cases and the Pandemrix vaccination. Consequently, the effect of the virus itself cannot be ruled out as a contributing factor. The delayed timing in diagnostics is crucial when determining the number of cases with onset during the pandemic.
... Acute rheumatic fever occurs in 2% to 3% of people with type A streptococcal pharyngitis. During the 2009 global influenza A (H1N1) pandemic, narcolepsy was reported in more than 1300 people who received Pandemrix vaccine (17,18). Shahed (19) reported that Guillain-Barre syndrome (GBS) was associated with influenza infection, but Kim (20) held the opposite view that the incidence of GBS increased slightly but not significantly during the influenza pandemic in Korea. ...
Article
Full-text available
Introduction Microbial infections are associated with the occurrence of autoimmune diseases, but the mechanisms of microbial infection inducing autoimmune diseases are not fully understood. The existence of heterophilic antigens between microorganisms and human tissues may explain part of the pathogenesis of autoimmune diseases. Here, we investigate the distribution of heterophilic antigens and its relationship with autoimmune diseases. Methods Monoclonal antibodies against a variety of microorganisms were prepared. The titer, subclass and reactivity of antibodies with microorganisms were identified, and heterophilic antibodies that cross-reacted with human tissues were screened by human tissue microarray. The reactivity of these heterophilic antibodies with different individuals and different species was further examined by immunohistochemistry. Results In this study, 21 strains of heterophilic antibodies were screened. The results showed that these heterophilic antibodies were produced due to the existence of heterophilic antigens between microorganism and human body and the distribution of heterophilic antigens had individual, tissue and species differences. Conclusion Our study showed that heterophilic antigens exist widely between microorganisms and human body, and the heterophilic antigens carried by microorganisms may break the immune tolerance of the body through carrier effect and initiate immune response, which may be one of the important mechanisms of infection inducing autoimmune diseases.
... Major (and rare) side effects of vaccines are typically materializing within weeks or months post-vaccination and are either caused by the vaccine directly (e.g., an attenuated virus reverting or being virulent in an immunocompromised individuals-oral polio vaccine, yellow fever vaccine, etc.) or by an overreacting immune system (e.g., Guillain-Barre syndrome with influenza vaccines, narcolepsy with pandemic H1N1 vaccine). In both cases, symptoms arise mostly within a short period of time after vaccination [125][126][127]. Issues that arise many years post-vaccination have not been reported and it would be hard to think of mechanisms for those. ...
Article
Full-text available
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged late in 2019 and caused the coronavirus disease 2019 (COVID-19) pandemic that has so far claimed approximately 20 million lives. Vaccines were developed quickly, became available in the end of 2020, and had a tremendous impact on protection from SARS-CoV-2 mortality but with emerging variants the impact on morbidity was diminished. Here I review what we learned from COVID-19 from a vaccinologist’s perspective.
Article
Narcolepsy type 1 (NT1), characterized by the loss of hypocretin/orexin (HCRT) production in the lateral hypothalamus, has been linked to Pandemrix vaccination during the 2009 H1N1 pandemic, especially in children and adolescents. It is still unknown why this vaccination increased the risk of developing NT1. This study investigated the effects of Pandemrix vaccination during adolescence on Hcrt mRNA expression in mice. Mice received a primary vaccination (50 µL i.m.) during prepubescence and a booster vaccination during peri-adolescence. Hcrt expression was measured at three-time points after the vaccinations. Control groups included both a saline group and an undisturbed group of mice. Hcrt expression was decreased after both Pandemrix and saline injections, but 21 days after the second injection, the saline group no longer showed decreased Hcrt expression, while the Pandemrix group still exhibited a significant reduction of about 60% compared to the undisturbed control group. This finding suggests that Pandemrix vaccination during adolescence influences Hcrt expression in mice into early adulthood. The Hcrt mRNA level did not reach the low levels known to induce NT1 symptoms, instead, our finding supports the multiple-hit hypothesis of NT1 that states that several insults to the HCRT system may be needed to induce NT1 and that Pandemrix could be one such insult.
Chapter
Sleep is a physiological behaviour, cyclical and reversible, that occupies one third of our lives, even if some consider this time as regrettably wasted. But its ultimate explanation is still unknown, although multiple theories have tried to clarify it throughout the centuries. The issue of sleep and its abnormalities has attracted man’s interest from the beginning of time and many philosophers, poets and artists have addressed the topic. But the actual science of sleep, as we know it today, is relatively young. If we give Hans Berger, the father of electroencephalography, the merit of introducing a new technique to study the brain of the sleeper in 1929, then the history of somnology is less than 100 years old. Another significant discovery was the dichotomy between REM sleep, NREM sleep and their relation to wakefulness, encompassing the three stages of human consciousness. Sleep disorders comprise manifold phenomena affecting not only sleep itself but daytime functioning and some of them can implicate injuries or serious health outcomes. Some of these issues are reviewed in this chapter.
Article
Objective/background: Psychiatric symptoms and cognitive deficits add significantly to impairment in academic achievement and quality of life in patients with narcolepsy. The primary aim of this study was to evaluate the prevalence of psychiatric disorders and executive dysfunctions, secondly to explore the association between psychiatric comorbidity, executive dysfunctions, subjective and objective sleep measures, and severity of cerebrospinal fluid (CSF) hypocretin-1 deficiency in pediatric narcolepsy type 1 (PNT1). Patients/methods: Cross-sectional study of 59 consecutively included PNT1 patients (age: 6-20 years; 34:25 girls: boys; 54/59 H1N1 (Pandemrix®)-vaccinated). Core narcolepsy symptoms including subjective sleepiness, polysomnography and multiple sleep latency test results, CSF hypocretin-1 levels, psychiatric disorders (by semistructured diagnostic interview Kaufmann Schedule for Affective Disorders and Schizophrenia Present and Lifetime version (KSADS)), and executive dysfunction (by Behavior Rating of Executive Function (BRIEF)) were assessed. Results: 52.5% of the patients had one or more psychiatric comorbid disorder, and 64.7% had executive dysfunction in a clinically relevant range, with no sex difference in prevalence, while older age was associated with poorer executive function (p=0.013). Having any psychiatric comorbid disorder was associated with poorer executive functions (p=0.001). CSF hypocretin-1 deficiency severity was significantly associated with presence of psychiatric comorbidity (p=0.022) and poorer executive functions (p=0.030), and poorer executive functions was associated with subjective sleepiness (p=0.009). Conclusions: The high occurrence of, and association between, psychiatric comorbidity and executive dysfunction underlines the importance of close attention to both these comorbidities in clinical care of NT1.
Article
Full-text available
Background A recent publication suggested molecular mimicry of a nucleoprotein (NP) sequence from A/Puerto Rico/8/1934 (PR8) strain, the backbone used in the construction of the reassortant strain X-179A that was used in Pandemrix® vaccine, and reported on anti-hypocretin (HCRT) receptor 2 (anti-HCRTR2) autoantibodies in narcolepsy, mostly in post Pandemrix® narcolepsy cases (17 of 20 sera). In this study, we re-examined this hypothesis through mass spectrometry (MS) characterization of Pandemrix®, and two other pandemic H1N1 (pH1N1)-2009 vaccines, Arepanrix® and Focetria®, and analyzed anti-HCRTR2 autoantibodies in narcolepsy patients and controls using three independent strategies. Methods MS characterization of Pandemrix® (2 batches), Arepanrix® (4 batches) and Focetria® (1 batch) was conducted with mapping of NP 116I or 116M spectrogram. Two sets of narcolepsy cases and controls were used: 40 post Pandemrix® narcolepsy (PP-N) cases and 18 age-matched post Pandemrix® controls (PP-C), and 48 recent (≤6 months) early onset narcolepsy (EO-N) cases and 70 age-matched other controls (O-C). Anti-HCRTR2 autoantibodies were detected using three strategies: (1) Human embryonic kidney (HEK) 293T cells with transient expression of HCRTR2 were stained with human sera and then analyzed by flow cytometer; (2) In vitro translation of [³⁵S]-radiolabelled HCRTR2 was incubated with human sera and immune complexes of autoantibody and [³⁵S]-radiolabelled HCRTR2 were quantified using a radioligand-binding assay; (3) Optical density (OD) at 450 nm (OD450) of human serum immunoglobulin G (IgG) binding to HCRTR2 stably expressed in Chinese hamster ovary (CHO)-K1 cell line was measured using an in-cell enzyme-linked immunosorbent assay (ELISA). Results NP 116M mutations were predominantly present in all batches of Pandemrix®, Arepanrix® and Focetria®. The wild-type NP109-123 (ILYDKEEIRRIWRQA), a mimic to HCRTR234-45 (YDDEEFLRYLWR), was not found to bind to DQ0602. Three or four subjects were found positive for anti-HCRTR2 autoantibodies using two strategies or the third one, respectively. None of the post Pandemrix® narcolepsy cases (0 of 40 sera) was found positive with all three strategies. Conclusion Anti-HCRTR2 autoantibody is not a significant biological feature of narcolepsy or of post Pandemrix® autoimmune responses.
Article
Full-text available
Study objectives: Previous case reports of intravenous immunoglobulins (IVIg) in pediatric narcolepsy have shown contradictory results. Methods: This was a nonrandomized, open-label, controlled, longitudinal observational study of IVIg use in pediatric narcolepsy with retrospective data collection from medical files obtained from a single pediatric national reference center for the treatment of narcolepsy in France. Of 56 consecutively referred patients with narcolepsy, 24 received IVIg (3 infusions administered at 1-mo intervals) in addition to standard care (psychostimulants and/or anticataplectic agents), and 32 continued on standard care alone (controls). Results: For two patients in each group, medical files were unavailable. Of the 22 IVIg patients, all had cerebrospinal fluid (CSF) hypocretin ≤ 110 pg/mL and were HLA-DQB1*06:02 positive. Of the 30 control patients, 29 were HLA-DQB1*06:02 positive and of those with available CSF measurements, all 12 had hypocretin ≤ 110 pg/mL. Compared with control patients, IVIg patients had shorter disease duration, shorter latency to sleep onset, and more had received H1N1 vaccination. Mean (standard deviation) follow-up length was 2.4 (1.1) y in the IVIg group and 3.9 (1.7) y in controls. In multivariate-adjusted linear mixed-effects analyses of change from baseline in Ullanlinna Narcolepsy Scale (UNS) scores, high baseline UNS, but not IVIg treatment, was associated with a reduction in narcolepsy symptoms. On time-to-event analysis, among patients with high baseline UNS scores, control patients achieved a UNS score < 14 (indicating remission) less rapidly than IVIg patients (adjusted hazard ratio 0.18; 95% confidence interval: 95% confidence interval: 0.03, 0.95; p = 0.043). Shorter or longer disease duration did not influence treatment response in any analysis. Conclusions: Overall, narcolepsy symptoms were not significantly reduced by IVIg. However, in patients with high baseline symptoms, a subset of IVIg-treated patients achieved remission more rapidly than control patients.
Article
Full-text available
Study objectives: Recently, antibodies to the hypocretin receptor 2 (HCRTR2-Abs) were reported in a high proportion of narcolepsy patients who developed the disease following Pandemrix® vaccination. We tested a group of narcolepsy patients for the HCRTR2-Abs using a newly established cell-based assay. Methods: Sera from 50 narcolepsy type 1 (NT1) and 11 with type 2 (NT2), 22 patients with other sleep disorders, 15 healthy controls and 93 disease controls were studied. CSFs from 3 narcoleptic patients were also subsequently included. Human embryonic kidney cells were transiently transfected with human HCRTR2, incubated with patients' sera for 1 hour at 1:20 dilution and then fixed. Binding of antibodies was detected by fluorescently-labelled secondary antibodies to human IgG and the different IgG subclasses. A non-linear visual scoring system was used from 0 to 4; samples scoring ≥ 1 were considered positive. Results: Only 3/61 patients (5%) showed a score ≥ 1, one with IgG1- and two with IgG3-antibodies, but titers were low (1:40 - 1:100). CSFs from these patients were negative. The three positive patients included one NT1 case with associated psychotic features, one NT2 patient and a NT1 patient with normal hypocretin CSF levels. Conclusions: Low levels of IgG1 or IgG3 antibodies against HCRTR2 were found in 3/61 patients with narcolepsy, although only one presented with full-blown NT1. HCRTR2-Abs are not common in narcolepsy unrelated to vaccination.
Article
Background: Neuropathological findings support an autoimmune etiology as an underlying factor for loss of orexin-producing neurons in spontaneous narcolepsy type 1 (narcolepsy with cataplexy; sNT1) as well as in Pandemrix influenza vaccine-induced narcolepsy type 1 (Pdmx-NT1). The precise molecular target or antigens for the immune response have, however, remained elusive. Methods: Here we have performed a comprehensive antigenic repertoire analysis of sera using the next-generation phage display method - mimotope variation analysis (MVA). Samples from 64 children and adolescents were analyzed: 10 with Pdmx-NT1, 6 with sNT1, 16 Pandemrix-vaccinated, 16 H1N1 infected, and 16 unvaccinated healthy individuals. The diagnosis of NT1 was defined by the American Academy of Sleep Medicine international criteria of sleep disorders v3. Findings: Our data showed that although the immunoprofiles toward vaccination were generally similar in study groups, there were also striking differences in immunoprofiles between sNT1 and Pdmx-NT1 groups as compared with controls. Prominent immune response was observed to a peptide epitope derived from prostaglandin D2 receptor (DP1), as well as peptides homologous to B cell lymphoma 6 protein. Further validation confirmed that these can act as true antigenic targets in discriminating NT1 diseased along with a novel epitope of hemagglutinin of H1N1 to delineate exposure to H1N1. Interpretation: We propose that DP1 is a novel molecular target of autoimmune response and presents a potential diagnostic biomarker for NT1. DP1 is involved in the regulation of non-rapid eye movement (NREM) sleep and thus alterations in its functions could contribute to the disturbed sleep regulation in NT1 that warrants further studies. Together our results also show that MVA is a helpful method for finding novel peptide antigens to classify human autoimmune diseases, possibly facilitating the design of better therapies.
Article
An increased incidence of narcolepsy was seen in many countries after the pandemic H1N1 influenza vaccination campaign in 2009-2010. The H1N1 vaccine - narcolepsy connection is based on observational studies that are prone to various biases, e.g. confounding by H1N1 infection, and ascertainment, recall and selection biases. A direct pathogenic link has, however, remained elusive. We conducted a systematic review and meta-analysis to analyze the magnitude of H1N1 vaccination related risk and to examine if there was any association with H1N1 infection itself. We searched all articles from PubMed, Web of Science and Scopus, and other relevant sources reporting the incidence and risk of post-vaccine narcolepsy. In our paper, we show that the risk appears to be limited to only one vaccine (Pandemrix®). During the first year after vaccination, the relative risk of narcolepsy was increased 5-14-fold in children and adolescents and 2-7-fold in adults. The vaccine attributable risk in children and adolescents was around 1 per 18,400 vaccine doses. Studies from Finland and Sweden also appear to demonstrate an extended risk of narcolepsy into the second year following vaccination, but such conclusions should be interpreted with a word of caution due to possible biases. Benefits of immunization outweigh the risk of vaccination-associated narcolepsy, which remains a rare disease.
Article
Objective Studies associate pandemic influenza vaccination with narcolepsy. In Germany, a retrospective, multicenter, matched case–control study was performed to identify risk factors for narcolepsy, particularly regarding vaccinations (seasonal and pandemic influenza vaccination) and infections (seasonal and pandemic influenza) and to quantify the detected risks. Methods Patients with excessive daytime sleepiness who had been referred to a sleep center between April 2009 and December 2012 for multiple sleep latency test (MSLT) were eligible. Case report forms were validated according to the criteria for narcolepsy defined by the Brighton Collaboration (BC). Confirmed cases of narcolepsy (BC level of diagnostic certainty 1−4a) were matched with population-based controls by year of birth, gender, and place of residence. A second control group was established including patients in whom narcolepsy was definitely excluded (test-negative controls). Results A total of 103 validated cases of narcolepsy were matched with 264 population-based controls. The second control group included 29 test-negative controls. A significantly increased odd ratio (OR) to develop narcolepsy (crude OR [cOR] = 3.9, 95% confidence interval [CI] = 1.8–8.5; adjusted OR [aOR] = 4.5, 95% CI = 2.0–9.9) was detected in individuals immunized with pandemic influenza A/H1N1/v vaccine prior to symptoms onset as compared to nonvaccinated individuals. Using test-negative controls, in individuals immunized with pandemic influenza A/H1N1/v vaccine prior to symptoms onset, a nonsignificantly increased OR of narcolepsy was detected when compared to nonvaccinated individuals (whole study population, BC levels 1−4a: cOR = 1.9, 95% CI = 0.5–6.9; aOR = 1.8, 95% CI = 0.3–10.1). Conclusions The findings of this study support an increased risk for narcolepsy after immunization with pandemic influenza A/H1N1/v vaccine.
Article
The underlying molecular mechanisms of autoimmune diseases are poorly understood. In order to unravel the autoimmune processes across diseases, comprehensive and unbiased analyses of proteins targets recognized by the adaptive immune system are needed. Here, we present an approach starting from high-density peptide arrays to characterize autoantibody repertoires and to identify new autoantigens. A set of ten plasma and serum samples from subjects with multiple sclerosis, narcolepsy and without any disease diagnosis were profiled on a peptide array representing the whole proteome, hosting 2.2 million 12-mer peptides with a six amino acid lateral shift. Based on the IgG reactivities found on these whole-proteome peptide microarrays, a set of 23 samples was then studied on a targeted array with 174,000 12-mer peptides of single amino acid lateral shift. Finally, verification of IgG reactivities were conducted with a larger sample set (n=448) using the bead-based peptide microarrays. The presented workflow employed three different peptide microarray formats to discover and resolve the epitopes of human autoantibodies, and revealed two potentially new autoantigens: MAP3K7 in MS and NRXN1 in narcolepsy. The presented strategy provides insights into antibody repertoire reactivity at a peptide level and may accelerate the discovery and validation of autoantigens in human diseases.
Article
In [1], the following error was published on page 177. The correction is regarding the definition of type 1 diabetes mellitus (T1D) in Table 2. The outcome T1D was defined by the presence of a diagnosis code of insulin-dependent diabetes mellitus (ICD-10 = E10) in the Patient Register in combination with a filled prescriptions of insulin (ATC code = A10Axxx) in the Prescribed Drug Register (PDR). Any filled prescription of drugs used in diabetes (ATC code = A10xxxx) in the PDR were also used for exclusion of prevalent disease. Thus, two criteria were used for definition of T1D cases in our study, as originally planned and for the purpose of helping to define the outcome/diagnosis as specifically as possible. In summary, information on our use of prescription data from the PDR, as the second criterion to define the T1D diagnosis in our study, was erroneously left out in the Table 2 and should be added. (Table presented.). © 2016 The Association for the Publication of the Journal of Internal Medicine
Article
Objective: To assess and compare the frequencies of personal and family history of autoimmune diseases (AID), autoinflammatory disorders (ID), and allergies in a population of patients, adults and children, with narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), and idiopathic hypersomnia (IH), 3 central hypersomnia disorders, and healthy controls. Methods: Personal and family history of AID, ID, and allergies were assessed by questionnaire and medical interview in a large cohort of 450 consecutive adult patients (206 NT1, 106 NT2, 138 IH) and 95 pediatric patients (80 NT1) diagnosed according to the third International Classification of Sleep Disorders criteria in national reference centers for narcolepsy in France and 751 controls (700 adults, 51 children) from the general population. Results: Ten adults with NT1 (4.9%) had a comorbid AID vs 3.4% of adult controls, without between-group differences in adjusted models. AID frequency did not differ between children with NT1 and controls. Conversely, compared with controls, AID frequency was higher in adults with NT2 (p = 0.002), whereas ID (p = 0.0002) and allergy (p = 0.003) frequencies were higher in adults with IH. A positive family history of AID was found in the NT1 group and of ID in the IH group. Conclusions: NT1 is not associated with increased risk of comorbid immune disorders, in favor of a potentially unique pathophysiology. Conversely, compared with controls, the frequency of autoimmune diseases was higher in adults with NT2, whereas allergies and autoinflammatory disorders were more common in adults with IH, suggesting an immune dysregulation mechanism in these conditions.