You are on page 1of 8

J Endocrinol Invest

DOI 10.1007/s40618-017-0740-9

SHORT REVIEW

Polyglandular autoimmune syndromes


G. J. Kahaly1 · L. Frommer1 

Received: 25 July 2017 / Accepted: 3 August 2017


© Italian Society of Endocrinology (SIE) 2017

Abstract  physicians to take appropriate action in order to prevent full-


Background  In recent years, scientific knowledge pertain- blown PAS disease.
ing to the rare ORPHAN polyglandular autoimmune syn-
drome (registered code ORPHA 282196) has accumulated. Keywords  Polyglandular autoimmune disease · Clinical
Objective  To offer current demographic, clinical, serologi- picture · Diagnosis · Epidemiology · Treatment
cal and immunogenic data on PAS.
Methods  Review of the pertinent and current literature.
Results  Polyglandular autoimmune syndromes (PAS) are Definition and classification
multifactorial diseases with at least two coexisting autoim-
mune-mediated endocrinopathies. PAS show a great hetero- Polyglandular autoimmune syndromes (PAS) are defined by
geneity of syndromes and manifest sequentially with a large the coexistence of at least two autoimmune-mediated endo-
time interval between the occurrence of the first and second crinopathies [1, 2]. Specific clustering of monoglandular
glandular autoimmune disease. PAS cluster with several autoimmune diseases depends on genetic and non-genetic
non-endocrine autoimmune diseases. In most endocrinopa- environmental factors and differs considerably at the time
thies of PAS, the autoimmune process causes an irrevers- of presentation allowing distinguishing between two major
ible loss of function, while chronic autoimmune aggressions subtypes of PAS. The juvenile PAS type I shows a mono-
can simultaneously modify physiological processes in the genetic inheritance, whereas multiple genes contribute to
affected tissue and lead to altered organ function. The rare the etiopathogenesis of the adult PAS [3, 4]. Based on the
juvenile PAS type I is inherited in a monogenetic manner, various combinations of autoimmune endocrinopathies, the
whereas several susceptibility gene polymorphisms have adult PAS is subdivided into the types II–IV [5, 6]. Patients
been reported for the more prevalent adult types. Relevant with PAS are at high risk for developing non-glandular auto-
for a timely diagnosis at an early stage is the screening for immune diseases [7] (Table 1).
polyglandular autoimmunity in patients with monoglandular A disproportionately high co-occurrence of glandular dis-
autoimmune disease and/or first degree relatives of patients eases was described 1908 for the first time and was defined
with PAS. The most prevalent adult PAS type is the combi- as a separate entity [8]. In 1978, genetic analysis and defini-
nation of type 1 diabetes with autoimmune thyroid disease. tion of susceptibility genes led to the classification of PAS
Conclusions  Early detection of specific autoantibodies with different subtypes, that was modified several times in
and latent organ-specific dysfunction is advocated to alert consideration of continuously more profound comprehen-
sion of the pathogenesis [9–11]. The current classification
distinguishes between four PAS types.
PAS type I, also known as autoimmune polyendocrinop-
* G. J. Kahaly athy, candidiasis and ectodermal dystrophy (APECED) or
Kahaly@ukmainz.de
multiple endocrine deficiency autoimmune candidiasis syn-
1
Department of Medicine I, Johannes Gutenberg University drome is a rare disease characterized by the manifestation of
Medical Center, 55101 Mainz, Germany autoimmune endocrinopathies in a peculiar sequence during

13
Vol.:(0123456789)
J Endocrinol Invest

Table 1  Characteristics of juvenile and adult PAS


Juvenile PAS Adult PAS

Age of manifestation Childhood Adulthood


Incidence [1/a] <1:100,000 1–2:100,000
Gender ratio [M:F] 3:4 1:3
Inheritance Monogenic Polygenic
Serology Interferon α/ω-Ab Organ-specific Ab
Autoimmune endocrinopathies Hypoparathyroidism (80–85%) Autoimmune thyropathy (70–75%)
Addison’s disease (60–70%) Type 1 diabetes (40–60%)
Type 1 diabetes (2–33%) Addison’s disease (40–50%)
Hypogonadism (12%) Hypoparathyroidism (≤5%)
Autoimmune thyropathy (10%) Hypogonadism (≤3%)
Hypopituitarism (≤2%)
Additional non-glandular auto- Mucocutaneous candidiasis (70–80%); autoimmune Atrophic gastritis; pernicious anemia; atopic eczema;
immune diseases hepatitis; atrophic gastritis; alopecia areata; vitiligo, alopecia areata; myasthenia gravis; systemic lupus
keratoconjunctivitis erythematosus; rheumatoid arthritis; autoimmune
hepatitis

infancy. In most affected children, chronic mucocutaneous glandular autoimmune diseases are common in PAS III. In
candidiasis caused by Candida albicans occurs before the comparison, PAS type IV is very heterogeneous involving a
age of 5 years affecting up to 5% of the body surface. During large variety of glandular autoimmune diseases that are not
the following years, a primary hypoparathyroidism manifests considered within PAS II–III. Being less well defined it is
as the first autoimmune-induced endocrine disease compo- often incorrectly described as a combination of monoglandu-
nent. The clinical phenotype is a hypocalcemia tetany in lar autoimmune disease with one non-glandular autoimmune
approximately 75%. The complete clinical picture of PAS disease. In fact, PAS type IV including various combinations
type I usually occurs prior to the age of 15 years with a of autoimmune hypopituitarism, hypergonadotropic hypo-
primary autoimmune-induced adrenal insufficiency (Addi- gonadism or hypoparathyroidism with T1D or an immune
son’s disease, AD) as second autoimmune endocrinopathy. thyropathy is rarely described in the literature [18, 19].
Incomplete subclinical forms make it difficult to timely diag- Further autoimmune polyendocrine syndromes exist, such
nose and provide early treatment. There is a high number of as the immunodysregulation polyendocrinopathy enteropa-
undetected cases, thus up to 15% of children with autoim- thy X-linked syndrome (IPEX) or the polyneuropathy orga-
mune adrenalitis may be PAS I positive [12]. Additionally nomegaly endocrinopathy monoclonal protein skin changes
non-endocrine autoimmune diseases such as malabsorption syndrome (POEMS), which are extremely rare and shall only
syndromes, necrotizing hepatitis or mucocutaneous candid- be mentioned for completeness.
iasis-related carcinomatosis have been observed in PAS I
patients [5, 13–15].
The adult forms of PAS show a great heterogeneity, differ Epidemiology
in prevalence and genetic background as well as in disease
associations and therefore are subdivided into PAS types The prevalence of PAS I is estimated at 1:100,000 ranging
II–IV. The major endocrine component of PAS II is AD. In from 1:43,000 (Slovenia) and 1:80,000 (Norway) up to
approximately 40–50% of cases with AD, additional autoim- 1:130,000 (Poland/Ireland). In populations with rather low
mune endocrine diseases occur. Up to 1 year after onset of genetic diversity, higher prevalence has been described,
AD, less than 50% of those patients only are detected due to e.g., Iranian Jews (1:9000), Sardinians (1:14,000) or Finns
the broad range of unspecific symptoms and the long time (1:25,000). The prevalence of PAS I is lower in Japanese
interval between the occurrences of the different endocrine populations (1:10,000,000) [20–23]. The female/male ratio
components [16]. With a relative prevalence of approxi- of PAS I is 0.8–2.4 showing a slight female predominance
mately 40%, PAS type III is the most frequent subtype and [7, 24]. The manifestation peak is in infancy. In contrast,
encompasses type 1 diabetes (T1D) and an autoimmune with a prevalence of 1:20,000, the adult form of PAS is far
thyroid disease, AITD [1]. T1D may occur with an autoim- more prevalent with an annual incidence of 1–2:100,000.
mune-induced hypothyroidism (generally caused by chronic Further, the prevalence of subclinical incomplete forms
lymphocytic Hashimoto’s thyroiditis, HT) or hyperthyroid- is estimated to be 150:100,000. The gender ratio of PAS
ism (caused by Graves’ disease, GD) differing in genetic, II–IV shows a female predominance with a female per-
serologic, and clinical presentation [17]. Additional (non-) centage of 75% [11, 25]. Due to its sequential course, the

13
J Endocrinol Invest

adult form of PAS is usually diagnosed beyond an age of Pathomechanism


20 years with a manifestation peak in the 4th and 5th dec-
ade depending on the combination of the various autoim- In most endocrine components of PAS, the autoimmune pro-
mune endocrinopathies [1, 26]. cess causes an irreversible and progressive loss of function as
in T1D, HT or AD. However, chronic autoimmune aggres-
sions can also simultaneously modify physiological processes
in the affected tissue and therefore lead to an altered specific
Etiology organ function as seen in patients with GD. Non-endocrine
autoimmune diseases, frequently accompanying PAS, can
Juvenile PAS show a systemic distribution pattern with unspecific, varying
tissue damage. Each of those forms of autoimmune processes
PAS I is one of the very rare monogenetic autoimmune mainly depends on the underlying pathomechanism that led
diseases. The underlying genetic defect is in the auto- to the loss of self-tolerance. The PAS shows exemplary fea-
immune regulator (AIRE) gene located on chromosome tures of both cellular and serologic autoimmunity. While
21q22.3. Over 60 different mutations are known varying, the cellular immunologic response is primarily analyzed for
in their frequency of occurrence, on region, ethnicity, and academic purposes, the serologic markers provide a tool for
genetic pool [27]. Most frequent mutations of AIRE are screening and monitoring, respectively. But organ-specific
R257X in exon 6 and 67–979del13 bp in exon 8 [20, 28]. autoantibodies can also be detected in healthy subjects. The
The coded protein is a transcriptional factor primarily difference between pathologic and physiologic autoantibod-
found in medullary thymic epithelial cells where central ies is found in the quantity as well as quality of the segments.
self-tolerance is provided via negative or positive selec- In general, natural antibodies show low serum titers with
tion, respectively [29]. By upregulating the number of the predominance of µ-isotypes and low antigen specificity,
expressed and presented self-antigens in healthy individu- while the V antibody region is characterized by non-mutated
als, the transcription factor protects against autoimmune germ line V gene segments (34). On the other side, patho-
diseases by eliminating autoreactive T cells. Thus, muta- genic autoantibodies are predominantly γ-isotype with high
tions in the AIRE gene leading to dysfunctional proteins antigen specificity and an alteration in V region due to a T ×
are strongly susceptible for autoimmunity. Patients with stimulation of B cells in the context of an adaptive immune
the same mutations can differ in their phenotype empha- response [34]. The serum titers are generally high though the
sizing the impact of environment or other susceptibility duration and progress must be considered in this regard, since
genes such as human leucocyte antigen (HLA) class I the serum titer of an organ-specific autoantibody declines in
and II modifying the clinical course of PAS I, i.e., HLA- the course of time and often correlates with the severity/level
DQB1*0301 correlating positively with PAS I severity of histopathological destruction [35].
[30, 31]. Origin and signaling pathway leading to occurrence
and manifestation of endocrine autoimmunity are not fully
understood. Several concepts may be hypothesized as expla-
Adult PAS nations for the breakdown of self-tolerance on level of T-
and B cells.
The adult PAS shows a multifactorial etiology with genes
determining a general susceptibility but not eventual dis- (a) Failure of deletion/suppression of autoreactive cells: a
ease manifestation with a concordance in monozygotic low number of autoreactive T- and B clones is consid-
twins being less than 100%. With respect to the polygen- ered to be physiologic with regulation due to T cells
etic inheritance, genes on chromosome 6 play a predomi- with immunomodulatory activity [34].
nant role [1]. By encoding the major histocompatibility (b) Molecular mimicry: microbial antigens being similar to
complex for the presentation of self- and foreign antigens, self-antigens cause immune reactions directed against
those genes have a great impact on the occurrence of auto- human body cells.
immunity in this context, too. Furthermore, several poly- (c) Abnormal presentation of (non-)self-antigens: com-
morphisms carry susceptibility for PAS. Immunogenet- plexes of self-antigens with foreign antigens can lead
ics and epidemiologic patterns differentiate between both to an aberrant presentation with inadequate T-cell acti-
mono- and polyglandular autoimmunity as well as between vation.
the subgroups of adult PAS types [32, 33]. Further envi- (d) Epitope spreading: epitope-specific immune responses
ronmental risk factors i.e., nicotine consumption, infec- lead to the release of antigens with occasional process-
tions or hormonal influence have been postulated. ing, presentation, and immunization against further

13
J Endocrinol Invest

epitopes as reported in stroke-induced adaptive auto- mature B and T lymphocytes is associated with several auto-
immune responses to novel neuronal antigens [36]. immune disorders. FOXP3 on the other hand regulates the
(e) Polyclonal lymphocyte activation: polyclonal B-cell differentiation of regulatory T cells and therefore might pro-
activation resulting in serum hypergammaglobulinemia. mote the development of autoimmune endocrinopathies and
PAS III, in particular (41).
While alternatives a-d can be considered as general patho-
mechanisms of autoimmune (endocrine) diseases, polyclonal
lymphocyte activation plays a minor part in organ-specific Clinical phenotype
autoimmunity.
Juvenile PAS
Immunogenetics
Juvenile PAS type I is characterized by the co-occurrence
The genetic risk factors of the adult PAS and their relevance of at least two of the three main component AD, primary
for disease manifestation and course of each autoimmune hypoparathyroidism, and candidiasis. Most patients suf-
component are still matter of current research. Several poly- fer from additional (non-) glandular autoimmunopathies.
morphisms have been reported as susceptibility genes and Approximately 70% of all patients with APECED have
others are assumed to be involved. Genes that have already three to five diseases associated with PAS I [42]. While
been identified to confer susceptibility encompass several primary hypoparathyroidism is found in about 75–90% and
HLA class I and II alleles which show concordant influence AD in about 65–80%, the prevalence of (transient) chronic
on both mono- and polyglandular autoimmunopathies but mucocutaneous candidiasis varies from 95–100% in Finish
simultaneously differentiate between PAS types II and III or Sardinian patients to 20% in Iranian Jews [43]. Further
[32, 33, 37]. In addition, the HLA genes on chromosome 6 ectodermal diseases such as alopecia, keratopathies, gingival
have the highest implications. PAS II-defining disorders are hyperplasia, dental enamel defects, and nail dystrophies are
associated with an increased frequency of the HLA haplotype observed [44]. Up to 24% patients with PAS I were reported
A1, B8, DR3, DQA1*0501, DQB1*0201 [38]. AD, in par- to suffer from intestinal dysfunction other than atrophic gas-
ticular, shows a strong association with DR3 and DR4 with a tritis or pernicious anemia (e.g., steatorrhea or constipation)
relative risk of 6.0, 4.6, and 26.5 for DR3, DR4, and DR3/4, with unknown etiology. A few patients have also endocrine
respectively. Both as mono and as component of PAS II, AD and exocrine pancreas insufficiency [13]. Autoimmune hepa-
is also correlated with DQ2/DQ8 with DRB1*0404 [39, titis is often referred to as rare component of PAS I and
40]. In contrast, in patients with PAS III, HLA-DRB1*03, is detected in 10–20% of the Finish and in 20–30% of the
*04, -DQA1*03 and -DQB1*02 are increased compared to Sardinian populations, respectively. Varying from asymp-
patients with isolated AITD, also allowing to distinguish tomatic to fulminant clinical course, autoimmune hepatitis
between HT and GD as PAS III components [33]. Numer- caused death in 25% of the Finish patients with PAS I. Fur-
ous population studies indicate that both HLA haplotypes ther glandular autoimmune diseases such as T1D or AITD
DR3-DQB1*0201 and DR4-DQB1*0302 contribute to PAS occur in about 10–20%, while hypogonadism is found in
III with DR3 conferring the susceptibility for T1D to a large 15–30% of male and 60–65% of female patients. Hypopi-
extent [38]. However, the polymorphisms −2221 Msp (C/T) tuitarism is a rare component being found in less than 5%.
and −23 Hphl (A/T) in the insulin gene are frequently found
in T1D as monoglandular disease but not within the scope of Adult PAS
PAS. Additionally, polymorphisms in the cytotoxic T-lym-
phocyte-associated antigen 4 (CTLA-4), protein tyrosine The adult PAS is not associated with chronic mucocutaneous
phosphatase non-receptor type 22 (PTPN22), and the fork- candidiasis, keratinopathies or other dystrophies and differs in
head box P3 (FOXP3) have been identified as risk factors the prevalence of glandular components by definition. Over-
for PAS emphasizing the relevance of the immunological all, AITD (45–46) represent the most frequent autoimmune
synapse [41]. It represents the concept of molecular interac- endocrinopathies within the scope of PAS and are present
tions underlying regulation of the immune response between in 70–75% of the patients [15]. AITD within PAS can occur
antigen presenting cells and T-cell receptors, while different with or without Graves’ orbitopathy [45–47].The second most
HLA class II alleles vary in subsequent receptor affinities for prevalent endocrine autoimmune disease in the adult PAS
peptides such as self-antigens, an alteration in the length of type is T1D with 50–60% [48]. Combination of both diseases
CTLA-4 genes leads to an impaired inhibitory function of defines PAS type III, whereas AD, essential for PAS II, occurs
T-cell activation. In line with this, the limitation of immuno- in 40–50% of all cases with PAS. In contrast to the juvenile
logic response to antigens via lymphoid tyrosine phosphatase PAS, primary hypogonadism is diagnosed in 4–11% and both
encoded by PTPN22 and expressed in immature as well as primary hypoparathyroidism and hypopituitarism are found

13
J Endocrinol Invest

in 1–5% only of the patients with adult PAS [49]. The adult autoimmunity. Concerning PAS I, interferon-ω and -α anti-
PAS manifests sequentially with a large time interval between bodies show the highest prevalence (95–100%) with early
occurrence of the first and second glandular autoimmune dis- emergence and have been proposed for confirmation of
ease. Additional non-glandular autoimmune diseases manifest diagnosis [13, 20]. Most organ-specific autoantibodies can
in the course of time and are observed in 40–50% [50]. be found in both PAS I and the respective isolated autoim-
mune disease. Additionally, NACHT leucine-rich repeat
protein 5 (NALP 5) correlating with primary hypoparathy-
Associated non‑glandular autoimmune diseases
roidism and ovarian insufficiency, tryptophan hydroxylase
antibodies (TPH) correlating with intestinal malabsorption
The prevalence of additional non-glandular autoimmune
and autoimmune hepatitis as well as aromatic l-amino acid
diseases associated with PAS I differs from the prevalence
decarboxylase antibodies (AADC) correlating with autoim-
in patients suffering from adult PAS. Especially autoim-
mune hepatitis and vitiligo can be found in patients with juve-
mune diseases with cutaneous manifestation appear to
nile PAS. Each of these above stated antibodies is present in
cluster in patients with a mutation in the AIRE gene, i.e.,
approximately 50% of the patients with NALP5 showing high
vitiligo (8–25%) and alopecia (16–40%), and are diagnosed
specificity but low sensitivity and TPH/AADC showing high
more often than in the adult type with 5–10 and 3–10%,
sensitivity but low to medium specificity. In contrast to the
respectively [51, 52]. Type A autoimmune, atrophic gastritis
adult PAS with polygenic dominant inheritance, patients at
(potentially associated with pernicious anemia) is found in
high risk for PAS I can theoretically be screened by genetic
4–10% (1–2%) with PAS II–IV and in 10–20% (1–6%) twice
testing of the AIRE gene. Further specific autoantibodies e.g.,
as often in the juvenile PAS. This holds true for autoim-
against IL-22, IL-17F, and IL-17A have been identified and
mune hepatitis diagnosed in 5–20% of PAS I patients but in
allow to distinguish isolated autoimmune diseases from those
less than 10% of the adult PAS patients. Sjögren syndrome
occurring in association with PAS I [53, 54].
occurs equally often in both juvenile and adult types (1–3%).
Patients with monoglandular autoimmune diseases should
be clinically and serologically screened further for both glan-
dular autoimmune diseases as well as non-endocrine auto-
Diagnosis and screening immune disorders (Fig. 1). As an example, TSH receptor
antibody testing is useful to diagnose AITD and untreated
Early diagnosis of the rare PAS I is difficult because of its autoimmune hyperthyroidism in particular [55]. Because of
clinical variability and inter-individual differences in pres- familial autoimmunity with autoimmune diseases clustering
entation with sequential occurrence of associated diseases. in relatives due to common pathomechanism, first-degree
Except for the detection of at least two of the three major relatives and members of multiplex families (at least two
components mentioned above, the manifestation of one major family members suffering from monoglandular autoimmune
component in a sibling of a patient diagnosed with PAS I disease) should also be screened [56]. Since overt (non-)
allows diagnosis, too. PAS I should be considered in patients glandular autoimmune diseases are defined by insufficiency
with chronic mucocutaneous candidiasis, adrenal insuffi- or complete loss of function of the affected organ, following
ciency or hypoparathyroidism in young age combined with functional testing is needed to confirm diagnosis.
at least one minor component such as chronic diarrhea/severe
constipation, keratitis/enamel hypoplasia, periodic rash with
fever, autoimmune hepatitis, vitiligo or alopecia [13]. Therapy
Crucial for the diagnosis of PAS II–IV at an early stage
is considering the presence of polyglandular autoimmun- Treatment of glandular autoimmune diseases consists
ity in patients with still uncomplete syndrome. Screening primarily of substitution of hormones, modification of
and regular follow-up examinations should be performed endocrine function, symptomatic therapy, and preven-
to detect endocrine insufficiencies before the occurrence of tion of potential complications. Since autoimmune patho-
potentially severe acute (e.g., AD crisis) or chronic com- mechanisms cause chronic mucocutaneous candidiasis,
plications (e.g., long-term damages in T1D). In fact, the immunomodulators have become of interest in the treat-
consideration of an increased prevalence of non-glandular ment of PAS I. Topical and systemic application of der-
autoimmune diseases in patients with PAS can help to avail matologic medications with symptomatic treatment is
missing the therapeutic window of opportunity for early combined with immunosuppression adjusted to the overt
treatment (e.g., rheumatoid arthritis in patients with PAS autoimmune disease and individual response. In PAS I,
suffering from joint problems). the most important life-threatening complications occur
Besides clinical symptoms, serological testing is indis- during childhood and require immediate aggressive immu-
pensable for the screening of patients with polyglandular nosuppression. Successful treatment of gastrointestinal

13
J Endocrinol Invest

Transglutamin
Serological screening
TPO- / TG- / 17-OH Ab CaSR - Ab PC-Ab / IF-Ab
Ab / C-peptide 21-OH Ab ase - Ab
TSH-R-Ab

positive

positive

positive

positive
positive

positive

positive
Functional screening

Endoscopy /
LH / FSH PTH / Ca. / Endoscopy /
OGTT TSH / fT4 Cortisol / ACTH biopsy
E/T Phosphate RBC

positive
positive

positive
positive
positive

positive

positive
Therapy

Iron / vitamin
Insulin LT4 / MMI Hydrocortisone E/T Vitamin D Gluten-free diet
B12

Fig. 1  The serologic and functional screening for associated auto- will be managed first with the administration of anti-thyroid drugs
immune diseases in patients with a monoglandular autoimmune dis- (e.g., methimazole, MMI). Ab antibody, OGTT oral glucose tolerance
ease performed at the onset of endocrinopathy and during follow-up test, ACTH adrenocorticotropic hormone, Ca-Sensor calcium-sensing
appointments every 2  years. After diagnosis of type 1 diabetes, thy- receptor, E estradiol, FSH follicle-stimulating hormone, fT4 free thy-
roid dysfunction, adrenal failure, primary hypogonadism, hypopar- roxine, Hypo hypothyroidism, Hyperthyr hyperthyroidism, IF intrin-
athyroidism, type A autoimmune gastritis with or without pernicious sic factor, LH luteinizing hormone, PC parietal cell, PTH parathyroid
anemia, and celiac disease, substitution proceeds with levothyroxine, hormone, RBC red blood cell count, T total testosterone, transglutam-
hydrocortisone, estradiol (E) or testosterone (T), vitamin D, iron tab- inase antibodies, TG thyroglobulin, TPO thyroid peroxidase, TSH-R
lets, and vitamin B12 intramuscularly, with a strict gluten-free diet. TSH receptor, TSH thyrotropin, Vit vitamin, 17-OH 17-hydroxylase,
In contrast, hyperthyroidism due to the autoimmune Graves’ disease 21-OH 21-hydroxylase

dysfunction, alopecia, and keratoconjunctivitis with cyclo- candidiasis can be observed requiring adequate antimicro-
sporine has been reported, while autoimmune hepatitis bial or virustatic drugs (e.g., septrin, itraconazole, and acy-
responded to steroids and azathioprine in several patients clovir). Especially systemic application of azole antifungals
[31, 57]. Additionally, gastrointestinal dysfunction and may lead to severe liver damage and should be avoided in
exocrine pancreatic insufficiency showed remission and patients with autoimmune hepatitis. With regard to immu-
partial amelioration, respectively, in immunosuppressive nosuppressive drugs, severe side-effects such as sirolimus-
regimes including tacrolimus and mycophenolate mofetil associated acute interstitial nephritis must be considered and
or oral methotrexate, respectively [58, 59]. Experimental regular controls must be performed at close intervals.
therapy with either anti-CD52 (Alemtuzumab) or anti- In patients with PAS, physiologic interactions of hor-
CD20 monoclonal antibodies (rituximab) was effective mones need to be considered. levothyroxine, for instance,
on treatment-resistant components of PAS I. increases hepatic metabolism of cortisol and might lead to
In overt endocrine insufficiency, substitution therapy acute AD failure in patients with PAS type II. Furthermore,
depends on the residual glandular function and is admin- AD causes both a reduction of insulin requirement as well as
istered in accordance to monoglandular autoimmune dis- TSH increase due to absence of glucocorticoid-transmitted
eases. In patients with the adult PAS, immunomodulatory inhibition of TSH secretion [1, 60]. Because of the complex-
therapy does not take a central role and is restricted to ity of hormonal therapy, patients need to be instructed and
special indications such as Graves’ orbitopathy (47) or informed about these interactions to avoid complications
additional non-glandular autoimmune diseases. when taking the drug (e.g., simultaneous intake of levothy-
Especially during immunosuppressive therapy in patients roxine and vitamin D/calcium) and/or side effects (gastric
with PAS I, an aggravation of chronic mucocutaneous ulceration during steroid therapy).

13
J Endocrinol Invest

Compliance with ethical standards  17. Horie I, Kawasaki E, Ando T, Kuwahara H, Abiru N, Usa T,
Yamasaki H, Ejima E, Kawakami A (2012) Clinical and genetic
characteristics of autoimmune polyglandular syndrome type
Conflict of interest  The authors have no conflicts of interest to de-
3 variant in the Japanese population. J Clin Endocrinol Metab
clare.
97(6):E1043–E1050. doi:10.1210/jc.2011-3109
18. Yoshioka K, Ohsawa A, Yoshida T, Yokoh S (1993) Insulin-
Ethical approval  This review does not contain any studies with
dependent diabetes mellitus associated with Graves’ disease
human participants or animals performed by any of the authors.
and idiopathic hypoparathyroidism. J Endocrinol Investig
16(8):643–646
Informed consent  No informed consent needed. 19. Lupi I, Raffaelli V, Di Cianni G, Caturegli P, Manetti L, Ciccarone
AM, Bogazzi F, Mariotti S, Del Prato S, Martino E (2013) Pitui-
tary autoimmunity in patients with diabetes mellitus and other
endocrine disorders. J Endocrinol Investig 36(2):127–131
20. Kisand K, Peterson P (2015) Autoimmune polyendocrinopathy
candidiasis ectodermal dystrophy. J Clin Immunol 35(5):463–478.
References doi:10.1007/s10875-015-0176-y
21. Myhre AG, Halonen M, Eskelin P, Ekwall O, Hedstrand H,
1. Kahaly GJ (2009) Polyglandular autoimmune syndromes. Eur Rorsman F, Kampe O, Husebye ES (2001) Autoimmune poly-
J Endocrinol Eur Fed Endocr Soc 161(1):11–20. doi:10.1530/ endocrine syndrome type 1 (APS I) in Norway. Clin Endocrinol
eje-09-0044 54(2):211–217
2. Eisenbarth GS, Gottlieb PA (2004) Autoimmune polyendocrine 22. Sato K, Nakajima K, Imamura H, Deguchi T, Horinouchi S,
syndromes. N Engl J Med 350(20):2068–2079. doi:10.1056/ Yamazaki K, Yamada E, Kanaji Y, Takano K (2002) A Novel
NEJMra030158 Missense Mutation of <I>AIRE</I> gene in a patient with
3. Cutolo M (2014) Autoimmune polyendocrine syndromes. Autoim- autoimmune polyendocrinopathy, candidiasis and ectodermal
mun Rev 13(2):85–89. doi:10.1016/j.autrev.2013.07.006 dystrophy (APECED), accompanied with progressive muscular
4. Martinez Lopez MM, Gonzalez Casado I, Alvarez Doforno R, Del- atrophy: case report and review of the literature in Japan. Endocr
gado Cervino E, Gracia Bouthelier R (2006) AIRE gene mutation in J 49(6):625–633. doi:10.1507/endocrj.49.625
polyglandular syndrome type 1. Anales de pediatria 64(6):583–587 23. Dominguez M, Crushell E, Ilmarinen T, McGovern E, Collins
5. Hansen MP, Kahaly GJ (2013) Autoimmune polyglandular syn- S, Chang B, Fleming P, Irvine AD, Brosnahan D, Ulmanen I,
dromes. Deutsche medizinische Wochenschrift 138(7):319–326. Murphy N, Costigan C (2006) Autoimmune polyendocrinopathy-
doi:10.1055/s-0032-1327355 (quiz 327-318) candidiasis-ectodermal dystrophy (APECED) in the Irish popula-
6. Forster G, Krummenauer F, Kuhn I, Beyer J, Kahaly G (1999) tion. J Pediatr Endocrinol Metab 19(11):1343–1352
Polyglandular autoimmune syndrome type II: epidemiology and 24. Betterle C, Dal Pra C, Mantero F, Zanchetta R (2002) Autoim-
forms of manifestation. Dtsch Med Wochenschr 124(49):1476– mune adrenal insufficiency and autoimmune polyendocrine syn-
1481. doi:10.1055/s-2008-1035684 dromes: autoantibodies, autoantigens, and their applicability in
7. Hansen MP, Matheis N, Kahaly GJ (2015) Type 1 diabetes and diagnosis and disease prediction. Endocr Rev 23(3):327–364.
polyglandular autoimmune syndrome: a review. World J Diabetes doi:10.1210/edrv.23.3.0466
6(1):67–79. doi:10.4239/wjd.v6.i1.67 25. Betterle C, Lazzarotto F, Presotto F (2004) Autoimmune polyglan-
8. Claude HGH (1908) Insuffisance pluriglandulaire endocrinienne. dular syndrome type 2: the tip of an iceberg? Clin Exp Immunol
J Physiol Pathol Gen 10:469–480 137(2):225–233. doi:10.1111/j.1365-2249.2004.02561.x
9. Eisenbarth GS, Wilson PW, Ward F, Buckley C, Lebovita H 26. Fatma M, Mouna E, Raouf H, Hajer F, Hatem M, Mohammed
(1979) The polyglandular failure syndrome: disease inheritance, A (2014) Autoimmune polyglandular syndrome type II: epide-
HLA type, and immune function. Ann Intern Med 91(4):528–533 miological, clinical and immunological data. J Endocrinol Metab
10. Muir A, She JX (1999) Advances in the genetics and immunology 4(4):101–109
of autoimmune polyglandular syndrome II/III and their clinical 27. Waterfield M, Anderson MS (2010) Clues to immune tolerance:
applications. Ann Med Interne 150(4):301–312 the monogenic autoimmune syndromes. Ann N Y Acad Sci
11. Betterle C, Zanchetta R (2003) Update on autoimmune polyendo- 1214:138–155. doi:10.1111/j.1749-6632.2010.05818.x
crine syndromes (APS). Acta bio-medica: Atenei Parm 74(1):9–33 28. Wolff AS, Erichsen MM, Meager A, Magitta NF, Myhre AG,
12. Arlt W, Allolio B (2003) Adrenal insufficiency. Lancet Bollerslev J, Fougner KJ, Lima K, Knappskog PM, Husebye ES
361(9372):1881–1893. doi:10.1016/S0140-6736(03)13492-7 (2007) Autoimmune polyendocrine syndrome type 1 in Norway:
13. Husebye ES, Perheentupa J, Rautemaa R, Kampe O (2009) Clini- phenotypic variation, autoantibodies, and novel mutations in the
cal manifestations and management of patients with autoimmune autoimmune regulator gene. J Clin Endocrinol Metab 92(2):595–
polyendocrine syndrome type I. J Intern Med 265(5):514–529. 603. doi:10.1210/jc.2006-1873
doi:10.1111/j.1365-2796.2009.02090.x 29. Chan AY, Anderson MS (2015) Central tolerance to self revealed
14. Bensing S, Brandt L, Tabaroj F, Sjoberg O, Nilsson B, Ekbom A, by the autoimmune regulator. Ann N Y Acad Sci 1356(1):80–89.
Blomqvist P, Kampe O (2008) Increased death risk and altered doi:10.1111/nyas.12960
cancer incidence pattern in patients with isolated or combined 30. Halonen M, Eskelin P, Myhre AG, Perheentupa J, Husebye ES,
autoimmune primary adrenocortical insufficiency. Clin Endo- Kampe O, Rorsman F, Peltonen L, Ulmanen I, Partanen J (2002)
crinol 69(5):697–704. doi:10.1111/j.1365-2265.2008.03340.x AIRE mutations and human leukocyte antigen genotypes as
15. Dittmar M, Kahaly GJ (2003) Polyglandular autoimmune syn- determinants of the autoimmune polyendocrinopathy-candidi-
dromes: immunogenetics and long-term follow-up. J Clin Endo- asis-ectodermal dystrophy phenotype. J Clin Endocrinol Metab
crinol Metab 88(7):2983–2992. doi:10.1210/jc.2002-021845 87(6):2568–2574. doi:10.1210/jcem.87.6.8564
16. Quinkler M (2012) Addison’s disease. Medizinische Klinik, Inten- 31. Meloni A, Willcox N, Meager A, Atzeni M, Wolff AS, Husebye
sivmedizin und Notfallmedizin 107(6):454–459. doi:10.1007/ ES, Furcas M, Rosatelli MC, Cao A, Congia M (2012) Autoim-
s00063-012-0112-3 mune polyendocrine syndrome type 1: an extensive longitudinal

13
J Endocrinol Invest

study in Sardinian patients. J Clin Endocrinol Metab 97(4):1114– L, Nardi M, Curro N, Boschi A, Bernard M, von Arx G, Per-
1124. doi:10.1210/jc.2011-2461 ros P, Kahaly GJ, European Group on Graves O (2017) Does
32. Barkia Beradhi S, Flesch BK, Hansen MP, Matheis N, Kahaly GJ early response to intravenous glucocorticoids predict the final
(2015) HLA class II differentiates between thyroid and polyglan- outcome in patients with moderate-to-severe and active Graves’
dular autoimmunity. Horm Metab Res 48(4):232–237. doi:10.10 orbitopathy? J Endocrinol Investig 40(5):547–553. doi:10.1007/
55/s-0035-1559622 s40618-017-0608-z
33. Flesch BK, Matheis N, Alt T, Weinstock C, Bux J, Kahaly GJ 48. Van den Driessche A, Eenkhoorn V, Van Gaal L, De Block C
(2014) HLA class II haplotypes differentiate between the adult (2009) Type 1 diabetes and autoimmune polyglandular syndrome:
autoimmune polyglandular syndrome types II and III. J Clin Endo- a clinical review. Neth J Medicine 67(11):376–387
crinol Metab 99(1):E177–E182. doi:10.1210/jc.2013-2852 49. Kobayashi I, Inukai T, Takahashi M, Ishii A, Ohshima K, Mori
34. Bellone M (2001) Autoimmune disease: pathogenesis. In: eLS. M, Shimomura Y, Kobayashi S, Hashimoto A, Sugiura M (1988)
Wiley. doi:10.1038/npg.els.0004000 Anterior pituitary cell antibodies detected in Hashimoto’s thyroid-
35. Kang SY, Oh JH, Song SK, Lee JS, Choi JC, Kang JH (2015) Both itis and Graves’ disease. Endocrinologia japonica 35(5):705–708
binding and blocking antibodies correlate with disease severity in 50. Wallaschofski H, Meyer A, Tuschy U, Lohmann T (2003) HLA-
myasthenia gravis. Neurol Sci: Off J Ital Neurol Soc Ital Soc Clin DQA1*0301-associated susceptibility for autoimmune pol-
Neurophysiol 36(7):1167–1171. doi:10.1007/s10072-015-2236-8 yglandular syndrome type II and III. Hormone and metabolic
36. Ortega SB, Noorbhai I, Poinsatte K, Kong X, Anderson A, research=Hormon- und Stoffwechselforschung=Hormones et
Monson NL, Stowe AM (2015) Stroke induces a rapid adaptive metabolisme 35(2):120–124. doi:10.1055/s-2003-39059
autoimmune response to novel neuronal antigens. Discov Med 51. Betterle C, Greggio NA, Volpato M (1998) Clinical review 93:
19(106):381–392 autoimmune polyglandular syndrome type 1. J Clin Endocrinol
37. Ramos-Lopez E, Lange B, Kahles H, Willenberg HS, Meyer G, Metab 83(4):1049–1055. doi:10.1210/jcem.83.4.4682
Penna-Martinez M, Reisch N, Hahner S, Seissler J, Badenhoop 52. Betterle C, Volpato M, Greggio AN, Presotto F (1996) Type 2 pol-
K (2008) Insulin gene polymorphisms in type 1 diabetes, Addi- yglandular autoimmune disease (Schmidt’s syndrome). J Pediatr
son’s disease and the polyglandular autoimmune syndrome type Endocrinol Metab 9(Suppl 1):113–123
II. BMC Med Genet 9:65. doi:10.1186/1471-2350-9-65 53. Puel A, Doffinger R, Natividad A, Chrabieh M, Barcenas-Morales
38. Huang W, Connor E, Rosa TD, Muir A, Schatz D, Silverstein G, Picard C, Cobat A, Ouachee-Chardin M, Toulon A, Bustamante
J, Crockett S, She JX, Maclaren NK (1996) Although DR3- J, Al-Muhsen S, Al-Owain M, Arkwright PD, Costigan C, McCo-
DQB1*0201 may be associated with multiple component dis- nnell V, Cant AJ, Abinun M, Polak M, Bougneres PF, Kumararatne
eases of the autoimmune polyglandular syndromes, the human D, Marodi L, Nahum A, Roifman C, Blanche S, Fischer A, Bode-
leukocyte antigen DR4-DQB1*0302 haplotype is implicated only mer C, Abel L, Lilic D, Casanova JL (2010) Autoantibodies against
in beta-cell autoimmunity. J Clin Endocrinol Metab 81(7):2559– IL-17A, IL-17F, and IL-22 in patients with chronic mucocutaneous
2563. doi:10.1210/jcem.81.7.8675578 candidiasis and autoimmune polyendocrine syndrome type I. J Exp
39. Maclaren NK, Riley WJ (1986) Inherited susceptibility to autoim- Med 207(2):291–297. doi:10.1084/jem.20091983
mune Addison’s disease is linked to human leukocyte antigens- 54. Kisand K, Boe Wolff AS, Podkrajsek KT, Tserel L, Link M,
DR3 and/or DR4, except when associated with type I autoimmune Kisand KV, Ersvaer E, Perheentupa J, Erichsen MM, Bratanic N,
polyglandular syndrome. J Clin Endocrinol Metab 62(3):455–459. Meloni A, Cetani F, Perniola R, Ergun-Longmire B, Maclaren N,
doi:10.1210/jcem-62-3-455 Krohn KJ, Pura M, Schalke B, Strobel P, Leite MI, Battelino T,
40. Robles DT, Fain PR, Gottlieb PA, Eisenbarth GS (2002) The Husebye ES, Peterson P, Willcox N, Meager A (2010) Chronic
genetics of autoimmune polyendocrine syndrome type II. Endo- mucocutaneous candidiasis in APECED or thymoma patients cor-
crinol Metab Clin North Am 31(2):353–368 relates with autoimmunity to Th17-associated cytokines. J Exp
41. Dittmar M, Kahaly GJ (2010) Genetics of the autoimmune pol- Med 207(2):299–308. doi:10.1084/jem.20091669
yglandular syndrome type 3 variant. Thyroid: Off J Am Thyroid 55. Diana T, Wuster C, Kanitz M, Kahaly GJ (2016) Highly variable
Assoc 20(7):737–743. doi:10.1089/thy.2010.1639 sensitivity of five binding and two bio-assays for TSH-receptor
42. Brabant G, Manns MP, Vogel A, Strassburg CP (2002) Autoim- antibodies. J Endocrinol Invest 39(10):1159–1165. doi:10.1007/
mun polyglanduläre Syndrome: Aspekte zu Pathogenese, Prog- s40618-016-0478-9
nose und Therapie. Dtsch Arztebl Int 99(21):1428 56. Cardenas-Roldan J, Rojas-Villarraga A, Anaya JM (2013) How do
43. Zlotogora J, Shapiro MS (1992) Polyglandular autoimmune syn- autoimmune diseases cluster in families? A systematic review and
drome type I among Iranian Jews. J Med Genet 29(11):824–826 meta-analysis. BMC Med 11:73. doi:10.1186/1741-7015-11-73
44. Ahonen P, Myllarniemi S, Sipila I, Perheentupa J (1990) Clinical 57. Ward L, Paquette J, Seidman E, Huot C, Alvarez F, Crock P, Delvin
variation of autoimmune polyendocrinopathy-candidiasis-ectoder- E, Kampe O, Deal C (1999) Severe autoimmune polyendocrinop-
mal dystrophy (APECED) in a series of 68 patients. N Engl J Med athy-candidiasis-ectodermal dystrophy in an adolescent girl with a
322(26):1829–1836. doi:10.1056/NEJM199006283222601 novel AIRE mutation: response to immunosuppressive therapy. J
45. Bartalena L, Chiovato L, Vitti P (2016) Management of hyper- Clin Endocrinol Metab 84(3):844–852. doi:10.1210/jcem.84.3.5580
thyroidism due to Graves’ disease: frequently asked questions 58. O’Gorman CS, Shulman R, Lara-Corrales I, Pope E, Marcon M,
and answers (if any). J Endocrinol Invest 39(10):1105–1114. Grasemann H, Schneider R, Upton J, Sochett EB, Koltin D, Cohen
doi:10.1007/s40618-016-0505-x E (2013) A child with autoimmune polyendocrinopathy candidi-
46. Bartalena L, Masiello E, Magri F, Veronesi G, Bianconi E, Zerbini asis and ectodermal dysplasia treated with immunosuppression:
F, Gaiti M, Spreafico E, Gallo D, Premoli P, Piantanida E, Tanda a case report. J Med Case Rep 7:44. doi:10.1186/1752-1947-7-44
ML, Ferrario M, Vitti P, Chiovato L (2016) The phenotype of 59. Abinun M, Hodges S, Cheetham T, Ognjanovic M, Hopper N,
newly diagnosed Graves’ disease in Italy in recent years is milder Burt A, Wood K, Lilic D (2014) ESID-0264 immunomodulatory
than in the past: results of a large observational longitudinal therapy for severe autoimmune polyendocrinopathy type-1 (APS-
study. J Endocrinol Investig 39(12):1445–1451. doi:10.1007/ 1). J Clin Immunol 34(Suppl 2):139–515
s40618-016-0516-7 60. Michels AW, Eisenbarth GS (2009) Autoimmune polyendocrine
47. Bartalena L, Veronesi G, Krassas GE, Wiersinga WM, Marcocci syndrome type 1 (APS-1) as a model for understanding autoim-
C, Marino M, Salvi M, Daumerie C, Bournaud C, Stahl M, Sassi mune polyendocrine syndrome type 2 (APS-2). J Intern Med
L, Azzolini C, Boboridis KG, Mourits MP, Soeters MR, Baldeschi 265(5):530–540. doi:10.1111/j.1365-2796.2009.02091.x

13

You might also like