Skip to main content

The combined prevalence of classified rare rheumatic diseases is almost double that of ankylosing spondylitis

Abstract

Background

Rare diseases (RDs) affect less than 5/10,000 people in Europe and fewer than 200,000 individuals in the United States. In rheumatology, RDs are heterogeneous and lack systemic classification. Clinical courses involve a variety of diverse symptoms, and patients may be misdiagnosed and not receive appropriate treatment. The objective of this study was to identify and classify some of the most important RDs in rheumatology. We also attempted to determine their combined prevalence to more precisely define this area of rheumatology and increase awareness of RDs in healthcare systems. We conducted a comprehensive literature search and analyzed each disease for the specified criteria, such as clinical symptoms, treatment regimens, prognoses, and point prevalences. If no epidemiological data were available, we estimated the prevalence as 1/1,000,000. The total point prevalence for all RDs in rheumatology was estimated as the sum of the individually determined prevalences.

Results

A total of 76 syndromes and diseases were identified, including vasculitis/vasculopathy (n = 15), arthritis/arthropathy (n = 11), autoinflammatory syndromes (n = 11), myositis (n = 9), bone disorders (n = 11), connective tissue diseases (n = 8), overgrowth syndromes (n = 3), and others (n = 8). Out of the 76 diseases, 61 (80%) are classified as chronic, with a remitting-relapsing course in 27 cases (35%) upon adequate treatment. Another 34 (45%) diseases were predominantly progressive and difficult to control. Corticosteroids are a therapeutic option in 49 (64%) syndromes. Mortality is variable and could not be determined precisely. Epidemiological studies and prevalence data were available for 33 syndromes and diseases. For an additional eight diseases, only incidence data were accessible. The summed prevalence of all RDs was 28.8/10,000.

Conclusions

RDs in rheumatology are frequently chronic, progressive, and present variable symptoms. Treatment options are often restricted to corticosteroids, presumably because of the scarcity of randomized controlled trials. The estimated combined prevalence is significant and almost double that of ankylosing spondylitis (18/10,000). Thus, healthcare systems should assign RDs similar importance as any other common disease in rheumatology.

Background

Rare diseases (RDs) are a complex problem in medicine, and the definition of a RD varies around the world. The European Union (EU) defines a disease as rare when it affects less than 5 people in 10,000 living in the EU, which translates to approximately 370,500 individuals being affected. In the United States, the Rare Diseases Act of 2002 defined a disease as rare when it affects less than 200,000 people. Patients frequently remain undiagnosed for many years, and treatment is often inefficacious. Data on prevalence, disease burden, treatment regimens, access to healthcare systems, and mortality are, to a great extent, unknown. Thus, RDs remain an unresolved challenge in modern medicine.

The classification, overall prevalence, and treatment options of rare rheumatic diseases are poorly defined. Disorders may affect the musculoskeletal apparatus with arthritis and myalgia but also involve other tissues in the form of myositis, vasculitis, autoimmune organ involvement, or bone diseases. Patients are often diagnosed as having a psychosomatic disorder due to missing or unrecognized somatic and/or objective findings. Patients not only have to cope with their disease burden, but are also at risk of developing additional psychiatric comorbidities. For example, patients with undiagnosed diseases in Germany have a prevalence of depressive symptoms three times higher than the average population (22% vs. 8.1%) [1, 2]. On the other hand, a verified diagnosis may aid the patient in accepting their diagnosis and coping with the ensuing symptoms and challenges. However, due to the scarcity of randomized controlled trials, treatment options for RDs are often limited and remain empirical. In addition, the definitions of key symptoms often vary in clinical studies, hampering uniform analyses.

RDs in rheumatology may be analyzed systematically by prevalence, genetic background and pathogenesis, clinical involvement, treatment options, and prognosis. Prevalence data vary according to age, as well as global and ethnic background. For example, seronegative symmetrical synovitis with pitting edema (RS3PE) has a high prevalence among the elderly, with 0.09% of all individuals over the age of 50 years being affected in Japan [3], but seems to be a quite rare disorder among younger individuals. Similarly, the prevalence of adult onset Still’s disease (AOSD) varies globally: 3.4–6.9/100,000 in Norway [4], 6.77/100,000 in Turkey [5], and 3.9/100,000 in Japan [6]. With respect to ethnic background, the estimated worldwide prevalence of Gaucher’s disease is 1–2/100,000, but in Ashkenazi-Jews the prevalence may be as high as 1/850 [7].

The pathogenesis and genetic backgrounds of some RDs in rheumatology have been studied increasingly in recent years, and in some cases well elucidated. Blau syndrome was described in 2001 and is characterized by mutations in the CARD15/NOD2 gene [8] and overexpression of autoinflammatory cytokines [9]. Interestingly, mutations in CARD15/NOD2 are also associated with other diseases with inflammatory involvement, such as Crohn´s disease and arthritis [10]. Familial cold urticaria (FCU), Muckle-Wells syndrome (MWS), and neonatal-onset multisystem inflammatory disease (NOMID or chronic infantile neurologic cutaneous articular syndrome [CINCA]) were originally thought to be three similar but distinct diseases. Further evidence has shown that all three syndromes are the result of mutations in the same gene, CIAS1. They are now referred to as different phenotypes of the same disorder, namely cryoporin-associated periodic syndrome (CAPS) [11, 12]. The MEFV gene, best known for causing familial Mediterranean fever (FMF) equally demonstrates the importance of genetics in RDs [13, 14]. Heterozygous mutations in MEFV are also found in many children with periodic fever, stomatitis, pharyngitis, adenitis (PFAPA) [15, 16]. Findings suggest that exon variants in MEFV may also be associated with AOSD [17].

With respect to pathogenesis, infectious agents may also play a role in RDs. As some diseases follow a distinct seasonal pattern, infectious pathogenesis has been suggested for Kawasaki disease [18, 19], and IgA-vasculitis (formerly Henoch-Schönlein purpura) [20, 21].

Furthermore, some diseases are probably modulated by hormonal alterations, such as pachydermoperiostosis [22]. Although a disease-causing genetic mutation has been detected [23], males are seemingly more commonly and severely affected [24]. RDs in rheumatology involve a variety of tissues and organ systems. For example, joints are affected by pigmented villonodular synovitis [25], the skeletal system by Camurati-Engelmann disease [26], and internal organs by AOSD [27]. The skin is involved in pyogenic arthritis, pyoderma gangrenosum, acne (PAPA) syndrome [28], blood vessels in granulomatosis with polyangiitis (GPA) [29], connective tissue in systemic sclerosis [30], and muscles in inclusion body myositis (IBM) [31].

Treatment options often, but not exclusively, include corticosteroids, such as in RS3PE [32] and eosinophilic-myalgia syndrome [33]. Although research on RDs is often limited to retrospective, single center trials or case reports only, randomized controlled trials (RCTs) have been increasingly available in recent years for some conditions, such as granulomatosis with polyangiitis [34], PFAPA syndrome [35], and FMF [36].

The disease course may be classified as self-limited (e.g., Kawasaki disease), chronic with a variable remitting-relapsing course during treatment (e.g., Takayasu arteritis), and chronic with a predominantly progressive and difficult to control course (e.g., systemic sclerosis).

The prognosis for RD varies and may be affected by the primary disease, complications, and treatment, especially long-term immunosuppression.

The objective of this study was to analyze the complexity of RDs in rheumatology. Based on data from the literature, we identified some of the most important sets of rheumatic diseases and calculated their combined prevalence. Our data may give better insight into this area of rheumatology, aid specialized centers for RDs, and raise overall awareness in the healthcare field.

Results

The diseases extracted from the databases are summarized in Table 2 of Appendix. The 76 syndromes and diseases were classified as follows: vasculitis/vasculopathy (n = 15), arthritis/arthropathy (n = 11), autoinflammatory syndromes (n = 11), myositis (n = 9), bone disorders (n = 11), connective tissue diseases (n = 8), which include inflammatory as well as non-inflammatory conditions, overgrowth syndromes (n = 3), and others (n = 8). A definitive genetic cause was identified in 26 diseases (34%). Out of the 76 conditions, 34 diseases (44%) were classified as chronic, primarily progressive, and difficult to control. Twenty-seven diseases (35%) were classified as chronic with a variable remitting-relapsing course. Six diseases (7%) were classified as self-limited. Acute phase reactants may be elevated in 49 diseases (64%). Corticosteroids are used as a therapeutic option in 49 diseases (64%). The mortality was variable and could not be determined precisely, but nine diseases (11%) were considered severe and potentially lethal if left untreated.

Prevalence data were available for 28 syndromes and diseases. For an additional five diseases, estimated prevalence data were already available. For another eight diseases, only incidence data were available. The prevalence of 38 diseases was estimated at 1/1,000,000, for 4 diseases at 1/100,000, and for 1 disease at 1/10,000. The combined prevalence per 10,000 is given in Table 1 (see also Figs. 1 and 2). The summed prevalence of all available and estimated RDs was 28.8/10,000.

Fig. 1
figure 1

Overview of the total number of diseases in each classification

Fig. 2
figure 2

Flow chart of syndrome inclusion in the analysis. After conducting the literature search and analysis with regard to prevalence, four diseases were excluded due to high prevalence, and seven diseases were merged into three, resulting in a total of 76 syndromes included in further analysis

Discussion

RDs are challenging for patients, healthcare professionals, and societies. Signs and symptoms often remain unrecognized and patients are excluded from specific treatment. Patients are frequently diagnosed with a psychosomatic disorder. Conversely, some patients with true extrasomatic diagnoses insist on the presence of a RD and cause significant expenses to healthcare systems. To overcome this bias, the importance of RDs should be recognized in public healthcare.

The knowledge of RDs is mostly available from case reports or case studies. On one hand, these studies are important in order to document essential information, such as commonly reported symptoms, different treatment regimens, and outcomes. However, such studies may involve reporting bias and, thus, are difficult to compare. For example, multicentric histiocytosis is a disease primarily reported in Caucasian women. However, this may be due to increased awareness in Western countries. Furthermore, women may consult a doctor more often and simulate a female Caucasian predominance [37].

Most studies scrutinizing RDs are designed as single center, retrospective studies due to a lack of patient numbers or networks. Larger registries would provide an opportunity to conduct retrospective or prospective and multicenter studies with a greater number of participating patients. Therefore, further development of international expert centers and registries is in great demand. In recent years, advances have been made due to the establishment of international expert/reference centers. For example, the Eurofever project for autoinflammatory diseases provided classification criteria and evaluated treatment options for multiple disorders [38, 39].

In addition, genetic testing has become increasingly available and, thus, more important in recent years. For example, whole exome sequencing in patients with similar symptoms without previous knowledge of candidate genes led to the identification of WISP 3 and SLCO2A1 as the pathogenic mutations in progressive pseudorheumatoid dysplasia [40] and primary hypertrophic osteoarthropathy [23].

In this study, we aimed to identify and classify RDs in rheumatology. We were able to show that the most common symptoms in rare rheumatic diseases are arthritis (31.0%, 89.3/100,000), followed by vasculitis (26.6%, 76.8/100,000), and connective tissue involvement (16.0%, 46.1/100,000), which in this study includes inflammatory as well as non-inflammatory conditions. Importantly, the total prevalence of a symptom was commonly dominated by only a few comparatively prevalent diseases. For example, systemic sclerosis (prevalence: 22.5/100,000) makes up 48.8% of all rare rheumatic diseases with connective tissue involvement.

Our study has several limitations. First, the nomenclature for diseases and syndromes is often used interchangeably, and the same disease or a variation in the same group of diseases is sometimes named differently. For example, CAPS is formerly known as Muckle-Wells, CINCA/NOMID, or familial cold autoinflammatory syndrome [12]. This may lead to incomplete search results and impede the comparability of available studies. Second, most prevalence data are almost exclusively based on retrospective analyses of hospital information or questionnaires. Prevalence data also vary according to ethnic background, geography, and age, which makes the use of overall prevalence data quite uncertain. For example, FMF or Behçet´s are more common in Mediterranean and Middle Eastern populations [41, 42] and rare in other regions, demonstrating the difficulty in using a local geographic prevalence. Similarly, although currently considered a RD by the European definition, an Italian study found an unexpectedly high overall prevalence of 8.5/10,000 for cryoglobulinemic vasculitis, which would no longer be defined as a RD. Although this study had some limitations, including the implementation of a questionnaire leading to a higher participation rate in the elderly population than the younger age groups, we decided to exclude cryoglobulinemic vasculitis from our list of rare rheumatological diseases. Our reasoning for this was the methodically more accurate estimation of prevalence by this study [43]. A well-known, verified RD, systemic sclerosis is quite common in Choctaw Native Americans (prevalence 66/100,000 in Oklahoma Choctaws and 469/100,000 in full-blooded Choctaws [44]), but rare among all other studied populations [45]. This may be due to a unique HLA haplotype, but other environmental factors may also play a role. Furthermore, our estimates of prevalence data may be somewhat inaccurate. We probably underestimated the prevalence by choosing 1 in 1,000,000 rather than 1 in 100,000, and the total prevalence of rare rheumatic diseases is likely to be even higher than our estimate of 29.6/10,000.

Prevalence data may also differ depending on age. For example, giant cell arteritis is probably extremely rare in younger patients but quite frequent in patients older than 55 years of age (UK 250/100,000 [46]), with age-independent prevalence data being difficult to obtain. Because the overall estimated prevalence may be even higher than 1/10,000, we also excluded giant cell arteritis from our list of RDs in rheumatology.

Similarly, we also excluded systemic lupus erythematodes, one of the better known “rare” diseases in rheumatology from our study, as recent epidemiological studies suggest that it is probably not a rare disease by the above mentioned European definition [47]. Furthermore, we also excluded rhupus syndrome, because it is suspected to be an overlap of systemic lupus erythematodes and rheumatoid arthritis and thus its prevalence may be difficult to obtain and distinguish [48].

The classification system we used also has its limitations. Conditions can be classified by their etiology or by their clinical appearance. As the etiology of many rare diseases remains unknown, we decided to classify diseases according to their main organ system involved in the clinical appearance of the disease. Exceptions include the category of autoinflammatory and overgrowth syndromes, where multiple organ systems may be involved, and the conditions in one group have a basic (suspected) etiology in common. However, overlaps between included conditions may have occurred because of the complex nature of some RDs.

Another pitfall is that diagnostic and/or classification criteria may differ in varying definitions of the diseases (e.g., familial Mediterranean fever), are not well established, and have been suggested based on radiographic or histological findings. In most cases, no validation studies are available to confirm specificity and sensitivity.

We observed that RCTs are available only for some RDs, such as Behcet´s disease [49] or ANCA-associated vasculitis [34]. Furthermore, most follow-ups of patients with RDs are rarely published. Larger patient cohorts would be necessary to obtain reliable data on outcome, disease progression, morbidity, and mortality. Treatment complications in most, if not all, RDs may be due to the disease itself or adverse events to immunosuppressive treatment. These data merit in-depth analyses, as they may shed more light on the disease and its pathophysiology or potential treatment options.

Incidence and prevalence data have become available for increasingly more RDs and, in some cases, the prevalence has even increased in recent years (e.g., Kawasaki disease) [50]. This trend may be due to a true increase in incidence, increased diagnosis of previously undiagnosed patients due to an upsurge in physician or patient awareness (internet, patient support groups, RD associations, etc.), or simply due to better national reporting systems and databases. In addition, an increased overall life expectancy in the general population has led to an increased incidence and prevalence of conditions that predominantly affect the elderly (e.g., GCA). Continuously better treatment options may also lead to prolonged overall survival with an increase in prevalence rates.

In this study, we found that the cumulative prevalence of RDs in rheumatology is at least 28.8/10,000, which is almost double that of ankylosing spondylitis (18/10,000) [51], a rather common disease seen in practice. This observation suggests that symptoms should be carefully acknowledged in all patients, especially when an overt psychopathology is present, as many patients with a yet undiagnosed disorder show signs of depression or other psychosomatic disorders [1], which can obscure the differentiation between primary disease and secondary complication even more for the treating physician. Our study may aid physicians as a simple tool for diagnosing patients with an undiagnosed rheumatic disease by comparing the symptoms, prevalence, and likeliness of one disorder to another.

Conclusion

Our study shows that RDs in rheumatology are not as rare as previously thought, affecting at least 28.8/10,000 people. Therefore, for every patient diagnosed with ankylosing spondylitis, 1.6 patients may suffer from a rare rheumatic disease. Although research and case reports of RDs are important, international expert centers are necessary to initiate and perpetuate patient cohorts and registries, establish classification/diagnostic criteria, and conduct clinical trials. Standard questionnaires and laboratory analyses may aid in obtaining better insight into the pathophysiology of RDs.

Methods and materials

The abstract archives of the European League Against Rheumatism, the American College of Rheumatology, Orpha.net, and the PubMed database were searched for the following terms: “rare” in combination with arthritis, arteritis, connective tissue disease, rheumatic, and vasculitis. Furthermore, terms were used in various combinations including arthralgia, autoimmune, fever, inflammation, joint pain, muscle pain, myalgia, and swollen joint. Identified syndromes were then classified according to their main appearance under the following terms: arthritis/arthropathy, bone disorders, autoinflammatory syndromes, connective tissue diseases, myositis/myopathy, overgrowth syndromes, vasculitis/vasculopathy, and others. Furthermore, we conducted a search of the literature and analyzed each disease according to the following criteria: prevalence, genetics/pathogenesis, diagnostic criteria, symptoms, laboratory findings, therapy, and prognosis. Diseases and syndromes were assessed for their overall prevalence and excluded if they did not meet the European definition of a RD (< 5/10,000) (see Fig. 3).

Fig. 3
figure 3

Overview of the combined prevalence per 100,000 per classification

If more than one prevalence was available, the prevalence data were averaged accordingly.

For statistical reasons, we estimated the prevalence for diseases for which no epidemiological data were available as one of three possible values: < 1/1,000,000, 1/100,000 or 1/10,000. The overall total prevalence for all RDs in rheumatology was estimated by summing up the available or estimated individual prevalence of each disease.

Availability of data and materials

All data generated or analysed during this study are included in this published article. Data citations are included in the reference list.

Abbreviations

AKT:

Serine/threonine kinase 1

ANA:

Antinuclear antibody

ANCA:

Anti-neutrophil cytoplasmic antibody

ANKH:

ANKH inorganic pyrophosphate transport regulator

ANTRX2:

Anthrax toxin receptor 2

AP:

Alkaline phosphatase

BMP:

Bone morphogenetic protein

BUN:

Blood urea nitrogen

CAPS:

Cryoporin-associated periodic syndrome

CCL18:

CC-chemokine ligand 18

CECR1:

Cat eye syndrome chromosome region

CIAS1:

Cold-induced autoinflammatory syndrome 1

CINCA:

Chronic infantile neurological, cutaneous, and articular syndrome

CMG2:

Capillary morphogenesis gene 2

COL1A1:

Collagen type 1 alpha 1

CPTII:

Carnitine palmitoyltransferase 2

CPPD:

Calcium pyrophosphate dehydrate

DM:

Dermatomyositis

DMARD:

Disease-modifying anti-rheumatic drug

DNASE1L3:

Deoxyribonuclease 1 like 3

DVT:

Deep vein thrombosis

FBN1:

Fibrillin 1

FCGR2A/3A:

Fc fragment of IgG receptor 2A/3A

GAG:

Glycosaminoglycan

GALNS:

Galactosamine (N-acetyl)-6-sulfatase

GBA1:

Glucocerebrosidase beta

GCSF:

Granulocyte colony-stimulating factor

GI:

Gastrointestinal

GNE:

Glucosamine (UDP-N-acetyl)-2-epimerase/N-acetylmannosamine kinase

HDAC9:

Histone deacetylase 9

HLA:

Human leukocyte antigene

HMGCoA:

3-Hydroxy-3-methylglutaryl-coenzyme A

HPGD:

Hydroxyprostaglandin dehydrogenase

IDS:

Iduronate 2-sulfatase

IL:

Interleukin

ILAR:

International League of Associations for Rheumatology

ILD:

Interstitial lung disease

ING5:

Inhibitor of growth family member 5

IOMID:

Infantile-onset multisystem inflammatory disease

JIA:

Juvenile idiopathic arthritis

KIT:

KIT proto-oncogene receptor tyrosine kinase

MIF:

Macrophage migration inhibitory factor

HLA:

Human leukocyte antigene

MAS:

Macrophage activation syndrome

MDS:

Myelodysplastic syndrome

MEFV:

Mediterranean fever

MIF:

Macrophage migration inhibitory factor

MMP3:

Matrix metalloproteinase-3

MPO:

Myeloperoxidase

MVK:

Mevalonate kinase

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B-cells

NOD2:

Nucleotide binding oligomerization domain containing 2

NOMID:

Neonatal onset multisystem inflammatory disease

NSAID:

Nonsteroidal anti-inflammatory drug

PAH:

Pulmonary arterial hypertension

PFKM:

Muscle phosphofructokinase

PGYM:

Muscle glycogen phosphorylase

PIK3CA:

Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit Α

PM:

Polymyositis

PPD/PPRD:

Progressive pseudorheumatoid dysplasia

PSMB8:

Proteasome subunit beta 8

PSTPIP1:

Proline-serine-threonine phosphatase interacting protein 1

PTX3:

Pentraxine 3

RF:

Rheumatoid factor

RA:

Rheumatoid arthritis

SAA:

Serum amyloid A

SAPHO:

Synovitis-acne-pustulosis-hyperostosis-osteitis-syndrome

SEDT-PA:

Spondyloepiphyseal dysplasia tarda with progressive arthropathy

SLE:

Systemic lupus erythematosus

TGFB1:

Transforming growth factor

TIMP1:

Tissue inhibitor of metalloproteinase

TNFi:

Tumor necrosis factor inhibitor

TNFRSF:

Tumor necrosis factor receptor superfamily

VEGF:

Vascular endothelial growth factor

WBC:

White blood cell

WISP3:

WNT1 inducible signaling pathway protein

References

  1. Windheuser IC, Mücke M, Klawonn F, Stieber C. Patients without diagnosis: A profile. (Poster Presentation 2nd World Congress on Rare Diseases and Orphan Drugs. London 2017)

  2. Busch MA, Maske UE, Ryl L, Schlack R, Hapke U. Prävalenz von depressiver Symptomatik und diagnostizierter Depression bei Erwachsenen in Deutschland: Ergebnisse der Studie zur Gesundheit Erwachsener in Deutschland (DEGS1). Bundesgesundheitsblatt - Gesundheitsforschung - Gesundheitsschutz. 2013;56:733–9.

    Article  CAS  PubMed  Google Scholar 

  3. Okumura T, Tanno S, Ohhira M, Nozu T. The rate of polymyalgia rheumatica (PMR) and remitting seronegative symmetrical synovitis with pitting edema (RS3PE) syndrome in a clinic where primary care physicians are working in Japan. Rheumatol Int. 2012;32:1695–9.

    Article  PubMed  Google Scholar 

  4. Evensen KJ, Nossent HC. Epidemiology and outcome of adult-onset Still’s disease in Northern Norway. Scand J Rheumatol. 2006;35:48–51.

    Article  CAS  PubMed  Google Scholar 

  5. Balci MA, Pamuk ON, Pamuk GE, Uzundere FK, Donmez S. AB1142 epidemiology and outcome of adult-onset still9s disease in northwestern thrace region in Turkey. Ann Rheum Dis. 2015;74:1284–1284.

    Article  Google Scholar 

  6. Asanuma YF, Mimura T, Tsuboi H, Noma H, Miyoshi F, Yamamoto K, et al. Nationwide epidemiological survey of 169 patients with adult Still’s disease in Japan. Mod Rheumatol. 2015;25:393–400.

    Article  PubMed  Google Scholar 

  7. Mistry PK, Cappellini MD, Lukina E, Özsan H, Pascual SM, Rosenbaum H, et al. Consensus conference: a reappraisal of Gaucher disease—diagnosis and disease management algorithms. Am J Hematol. 2011;86:110–5.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Miceli-Richard C, Lesage S, Rybojad M, Prieur A-M, Manouvrier-Hanu S, Häfner R, et al. CARD15 mutations in Blau syndrome. Nat Genet. 2001;29:19–20.

    Article  CAS  PubMed  Google Scholar 

  9. Wouters CH, Maes A, Foley KP, Bertin J, Rose CD. Blau Syndrome, the prototypic auto-inflammatory granulomatous disease. Pediatr Rheumatol Online J. 2014;12:33.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Zhong Y, Kinio A, Saleh M. Functions of NOD-like receptors in human diseases. Front Immunol [Internet]. 2013 [cited 2018 Aug 29];4. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC3797414/

  11. Levy R, Gérard L, Kuemmerle-Deschner J, Lachmann HJ, Koné-Paut I, Cantarini L, et al. Phenotypic and genotypic characteristics of cryopyrin-associated periodic syndrome: a series of 136 patients from the Eurofever Registry. Ann Rheum Dis. 2015;74:2043–9.

    Article  CAS  PubMed  Google Scholar 

  12. Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle–Wells syndrome. Nat Genet. 2001;29:301–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Consortium TFF, Bernot A, Clepet C, Dasilva C, Devaud C, Petit J-L, et al. A candidate gene for familial Mediterranean fever. Nat Genet. 1997;17:25–31.

    Article  Google Scholar 

  14. The International FMF Consortium. Ancient missense mutations in a new member of the RoRet gene family are likely to cause familial mediterranean fever. Cell. 1997;90:797–807.

    Article  Google Scholar 

  15. Celiksoy MH, Ogur G, Yaman E, Abur U, Fazla S, Sancak R, et al. Could familial Mediterranean fever gene mutations be related to PFAPA syndrome? Pediatr Allergy Immunol. 2016;27:78–82.

    Article  PubMed  Google Scholar 

  16. Taniuchi S, Nishikomori R, Iharada A, Tuji S, Heike T, Kaneko K. MEFV variants in patients with PFAPA syndrome in Japan. Open Rheumatol J. 2013;7:22–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nonaka F, Migita K, Jiuchi Y, Shimizu T, Umeda M, Iwamoto N, et al. Increased prevalence of MEFV exon 10 variants in Japanese patients with adult-onset Still’s disease. Clin Exp Immunol. 2015;179:392–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yim D, Curtis N, Cheung M, Burgner D. Update on Kawasaki disease: epidemiology, aetiology and pathogenesis. J Paediatr Child Health. 2013;49:704–8.

    Article  PubMed  Google Scholar 

  19. Rodó X, Ballester J, Cayan D, Melish ME, Nakamura Y, Uehara R, et al. Association of Kawasaki disease with tropospheric wind patterns. Sci Rep [Internet]. 2011 [cited 2018 Aug 30];1. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC3240972/

  20. Yan M, Wang Z, Niu N, Zhao J, Peng J. Relationship between chronic tonsillitis and Henoch–Schonlein purpura. Int J Clin Exp Med. 2015;8:14060–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Piram M, Mahr A. Epidemiology of immunoglobulin A vasculitis (Henoch–Schönlein): current state of knowledge. Curr Opin Rheumatol. 2013;25:171–8.

    Article  CAS  PubMed  Google Scholar 

  22. Bianchi L, Lubrano C, Carrozzo AM, Iraci S, Tomassoli M, Spera G, et al. Pachydermoperiostosis: study of epidermal growth factor and steroid receptors. Br J Dermatol. 1995;132:128–33.

    Article  CAS  PubMed  Google Scholar 

  23. Zhang Z, Xia W, He J, Zhang Z, Ke Y, Yue H, et al. Exome sequencing identifies SLCO2A1 mutations as a cause of primary hypertrophic osteoarthropathy. Am J Hum Genet. 2012;90:125–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Castori M, Sinibaldi L, Mingarelli R, Lachman R, Rimoin D, Dallapiccola B. Pachydermoperiostosis: an update. Clin Genet. 2005;68:477–86.

    Article  CAS  PubMed  Google Scholar 

  25. Xie G, Jiang N, Liang C, Zeng J, Chen Z, Xu Q, et al. Pigmented Villonodular Synovitis: A Retrospective Multicenter Study of 237 Cases. PLoS One [Internet]. 2015 [cited 2017 Sep 8];10. Available from: http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4370558/

  26. Janssens K, Vanhoenacker F, Bonduelle M, Verbruggen L, Van Maldergem L, Ralston S, et al. Camurati-Engelmann disease: review of the clinical, radiological, and molecular data of 24 families and implications for diagnosis and treatment. J Med Genet. 2006;43:1–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kadavath S, Efthimiou P. Adult-onset Still’s disease—pathogenesis, clinical manifestations, and new treatment options. Ann Med. 2015;47:6–14.

    Article  CAS  PubMed  Google Scholar 

  28. Smith EJ, Allantaz F, Bennett L, Zhang D, Gao X, Wood G, et al. Clinical, molecular, and genetic characteristics of PAPA syndrome: a review. Curr Genom. 2010;11:519–27.

    Article  CAS  Google Scholar 

  29. Comarmond C, Cacoub P. Granulomatosis with polyangiitis (Wegener): clinical aspects and treatment. Autoimmun Rev. 2014;13:1121–5.

    Article  PubMed  Google Scholar 

  30. Kowal-Bielecka O, Fransen J, Avouac J, Becker M, Kulak A, Allanore Y, et al. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76:1327–39.

    Article  PubMed  Google Scholar 

  31. Needham M, Mastaglia FL. Inclusion body myositis: current pathogenetic concepts and diagnostic and therapeutic approaches. Lancet Neurol. 2007;6:620–31.

    Article  PubMed  Google Scholar 

  32. Karmacharya P, Donato AA, Aryal MR, Ghimire S, Pathak R, Shah K, et al. RS3PE revisited: a systematic review and meta-analysis of 331 cases. Clin Exp Rheumatol. 2016;34:404–15.

    PubMed  Google Scholar 

  33. Hertzman PA. The eosinophilia-myalgia syndrome: status of 205 patients and results of treatment 2 years after onset. Ann Intern Med. 1995;122:851.

    Article  CAS  PubMed  Google Scholar 

  34. Stone JH, Merkel PA, Spiera R, Seo P, Langford CA, Hoffman GS, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N E J Med. 2010;363:221–32.

    Article  CAS  Google Scholar 

  35. Renko M, Salo E, Putto-Laurila A, Saxen H, Mattila PS, Luotonen J, et al. A randomized, controlled trial of tonsillectomy in periodic fever, aphthous stomatitis, pharyngitis, and adenitis syndrome. J Pediatr. 2007;151:289–92.

    Article  CAS  PubMed  Google Scholar 

  36. Ben-Zvi I, Kukuy O, Giat E, Pras E, Feld O, Kivity S, et al. Anakinra for colchicine-resistant familial mediterranean fever: a randomized, double-blind, placebo-controlled trial. Arthritis Rheumatol. 2017;69:854–62.

    Article  CAS  PubMed  Google Scholar 

  37. Tariq S, Hugenberg ST, Hirano-Ali SA, Tariq H. Multicentric reticulohistiocytosis (MRH): case report with review of literature between 1991 and 2014 with in depth analysis of various treatment regimens and outcomes. Springerplus [Internet]. 2016 [cited 2017 Sep 13];5. Available from: http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4766148/

  38. Federici S, Sormani MP, Ozen S, Lachmann HJ, Amaryan G, Woo P, et al. Evidence-based provisional clinical classification criteria for autoinflammatory periodic fevers. Ann Rheum Dis. 2015;74:799–805.

    Article  PubMed  Google Scholar 

  39. ter Haar N, Lachmann H, Özen S, Woo P, Uziel Y, Modesto C, et al. Treatment of autoinflammatory diseases: results from the Eurofever Registry and a literature review. Ann Rheum Dis. 2013;72:678–85.

    Article  PubMed  Google Scholar 

  40. Rai E, Mahajan A, Kumar P, Angural A, Dhar MK, Razdan S, et al. Whole Exome Screening Identifies Novel and Recurrent WISP3 Mutations Causing Progressive Pseudorheumatoid Dysplasia in Jammu and Kashmir-India. Sci Rep [Internet]. 2016 [cited 2017 Jun 23];6. Available from: http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4904372/

  41. Adeeb F, Stack AG, Fraser AD. Knitting the Threads of Silk through Time: Behçet’s Disease—Past, Present, and Future. Int J Rheumatol [Internet]. 2017 [cited 2017 Nov 30];2017. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5610876/

  42. Ben-Chetrit E, Touitou I. Familial mediterranean fever in the world. Arthritis Rheum. 2009;61:1447–53.

    Article  CAS  PubMed  Google Scholar 

  43. Monti G, Saccardo F, Castelnovo L, Novati P, Sollima S, Riva A, et al. Prevalence of mixed cryoglobulinaemia syndrome and circulating cryoglobulins in a population-based survey: the Origgio study. Autoimmun Rev. 2014;13:609–14.

    Article  CAS  PubMed  Google Scholar 

  44. Arnett FC, Howard RF, Tan F, Moulds JM, Bias WB, Durban E, et al. Increased prevalence of systemic sclerosis in a native american tribe in Oklahoma. Association with an Amerindian HLA haplotype. Arthritis Rheum. 1996;39:1362–70.

    Article  CAS  PubMed  Google Scholar 

  45. Barnes J, Mayes MD. Epidemiology of systemic sclerosis: incidence, prevalence, survival, risk factors, malignancy, and environmental triggers. Curr Opin Rheumatol. 2012;24:165–70.

    Article  PubMed  Google Scholar 

  46. Yates M, Graham K, Watts RA, MacGregor AJ. The prevalence of giant cell arteritis and polymyalgia rheumatica in a UK primary care population. BMC Musculoskelet Disord [Internet]. 2016 [cited 2017 Dec 4];17. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4946178/

  47. Stojan G, Petri M. Epidemiology of systemic lupus erythematosus: an update. Curr Opin Rheumatol. 2018;30:144–50.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Tani C, D’Aniello D, Sedie AD, Carli L, Cagnoni M, Possemato N, et al. Rhupus syndrome: Assessment of its prevalence and its clinical and instrumental characteristics in a prospective cohort of 103 SLE patients. Autoimmun Rev. 2013;12:537–41.

    Article  CAS  PubMed  Google Scholar 

  49. Vallet H, Riviere S, Sanna A, Deroux A, Moulis G, Addimanda O, et al. Efficacy of anti-TNF alpha in severe and/or refractory Behçet’s disease: multicenter study of 124 patients. J Autoimmun. 2015;62:67–74.

    Article  CAS  PubMed  Google Scholar 

  50. Descriptive Epidemiology of Kawasaki Disease in Japan. 2011–2012: from the results of the 22nd Nationwide Survey. J Epidemiol. 2015;25:239–45.

    Google Scholar 

  51. Exarchou S, Lindström U, Askling J, Eriksson JK, Forsblad-d’Elia H, Neovius M, et al. The prevalence of clinically diagnosed ankylosing spondylitis and its clinical manifestations: a nationwide register study. Arthritis Res Ther [Internet]. 2015 [cited 2018 Sep 25];17. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4424886/

  52. Arkkila PE. Thromboangiitis obliterans (Buerger’s disease). Orphanet J Rare Dis. 2006;1:14.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Zheng J-F, Chen Y-M, Chen D-Y, Lin C-H, Chen H-H. The incidence and prevalence of thromboangiitis obliterans in Taiwan: a nationwide, population-based analysis of data collected from 2002 to 2011. Clinics (Sao Paulo). 2016;71:399–403.

    Article  Google Scholar 

  54. Puéchal X, Fiessinger J-N. Thromboangiitis obliterans or Buerger’s disease: challenges for the rheumatologist. Rheumatology (Oxford). 2007;46:192–9.

    Article  Google Scholar 

  55. Shionoya S. Diagnostic criteria of Buerger’s disease. Int J Cardiol. 1998;66:S243–5.

    Article  PubMed  Google Scholar 

  56. Olin JW. Thromboangiitis Obliterans (Buerger’s Disease). N E J Med. 2000;343:864–9.

    Article  CAS  Google Scholar 

  57. Sada K-E, Amano K, Uehara R, Yamamura M, Arimura Y, Nakamura Y, et al. A nationwide survey on the epidemiology and clinical features of eosinophilic granulomatosis with polyangiitis (Churg–Strauss) in Japan. Mod Rheumatol. 2014;24:640–4.

    Article  PubMed  Google Scholar 

  58. Mahr A, Guillevin L, Poissonnet M, Aymé S. Prevalences of polyarteritis nodosa, microscopic polyangiitis, Wegener’s granulomatosis, and Churg–Strauss syndrome in a French urban multiethnic population in 2000: A capture–recapture estimate. Arthritis Rheum. 2004;51:92–9.

    Article  PubMed  Google Scholar 

  59. Mohammad AJ, Jacobsson LTH, Mahr AD, Sturfelt G, Segelmark M. Prevalence of Wegener’s granulomatosis, microscopic polyangiitis, polyarteritis nodosa and Churg–Strauss syndrome within a defined population in southern Sweden. Rheumatology (Oxford). 2007;46:1329–37.

    Article  CAS  Google Scholar 

  60. Comarmond C, Pagnoux C, Khellaf M, Cordier J-F, Hamidou M, Viallard J-F, et al. Eosinophilic granulomatosis with polyangiitis (Churg–Strauss): Clinical characteristics and long-term followup of the 383 patients enrolled in the French Vasculitis Study Group cohort. Arthritis Rheum. 2013;65:270–81.

    Article  PubMed  Google Scholar 

  61. Mahr A, Moosig F, Neumann T, Szczeklik W, Taillé C, Vaglio A, et al. Eosinophilic granulomatosis with polyangiitis (churg–strauss): evolutions in classification, etiopathogenesis, assessment and management. Curr Opin Rheumatol. 2014;26:16–23.

    Article  CAS  PubMed  Google Scholar 

  62. Greco A, Rizzo MI, De Virgilio A, Gallo A, Fusconi M, Ruoppolo G, et al. Churg–Strauss syndrome. Autoimmun Rev. 2015;14:341–8.

    Article  CAS  PubMed  Google Scholar 

  63. Masi AT, Hunder GG, Lie JT, Michel BA, Bloch DA, Arend WP, et al. The American College of Rheumatology 1990 criteria for the classification of churg-strauss syndrome (allergic granulomatosis and angiitis). Arthritis Rheum. 1990;33:1094–100.

    Article  CAS  PubMed  Google Scholar 

  64. Theodoridis A, Konstantinidou A, Makrantonaki E, Zouboulis C. Malignant and benign forms of atrophic papulosis (Köhlmeier–Degos disease): systemic involvement determines the prognosis. Br J Dermatol. 2014;170:110–5.

    Article  CAS  PubMed  Google Scholar 

  65. Theodoridis A, Makrantonaki E, Zouboulis CC. Malignant atrophic papulosis (Köhlmeier–Degos disease)—a review. Orphanet J Rare Dis. 2013;8:10.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Magro CM, Wang X, Garrett-Bakelman F, Laurence J, Shapiro LS, DeSancho MT. The effects of Eculizumab on the pathology of malignant atrophic papulosis. Orphanet J Rare Dis. 2013;8:185.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Shapiro LS, Toledo-Garcia AE, Farrell JF. Effective treatment of malignant atrophic papulosis (Köhlmeier–Degos disease) with treprostinil—early experience. Orphanet J Rare Dis. 2013;8:52.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Khalid U, Saleem T. Hughes-Stovin Syndrome. Orphanet J Rare Dis. 2011;6:15.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Erkan D, Yazici Y, Sanders A, Trost D, Yazici H. Is Hughes–Stovin syndrome Behçet’s disease? Clin Exp Rheumatol. 2004;22:S64-68.

    CAS  PubMed  Google Scholar 

  70. Jachiet M, Flageul B, Deroux A, Le Quellec A, Maurier F, Cordoliani F, et al. The clinical spectrum and therapeutic management of hypocomplementemic urticarial vasculitis: data from a French Nationwide study of fifty-seven patients. Arthritis Rheumatol. 2015;67:527–34.

    Article  CAS  PubMed  Google Scholar 

  71. Özçakar ZB, Foster J, Diaz-Horta O, Kasapcopur O, Fan Y-S, Yalçınkaya F, et al. DNASE1L3 mutations in hypocomplementemic urticarial vasculitis syndrome. Arthritis Rheum. 2013;65:2183–9.

    Article  PubMed  CAS  Google Scholar 

  72. Wakamatsu R, Watanabe H, Suzuki K, Suga N, Kitagawa W, Miura N, et al. Hypocomplementemic urticarial vasculitis syndrome is associated with high levels of serum IgG4: a clinical manifestation that mimics IgG4-related disease. Intern Med. 2011;50:1109–12.

    Article  CAS  PubMed  Google Scholar 

  73. Schwartz HR, McDuffie FC, Black LF, Schroeter AL, Conn DL. Hypocomplementemic urticarial vasculitis: association with chronic obstructive pulmonary disease. Mayo Clin Proc. 1982;57:231–8.

    CAS  PubMed  Google Scholar 

  74. Buck A, Christensen J, McCarty M. Hypocomplementemic urticarial vasculitis syndrome. J Clin Aesthet Dermatol. 2012;5:36–46.

    PubMed  PubMed Central  Google Scholar 

  75. Uehara R, Belay ED. Epidemiology of Kawasaki Disease in Asia, Europe, and the United States. J Epidemiol. 2012;22:79–85.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Singh S, Vignesh P, Burgner D. The epidemiology of Kawasaki disease: a global update. Arch Dis Child. 2015;100:1084–8.

    Article  PubMed  Google Scholar 

  77. Takahashi K, Oharaseki T, Yokouchi Y. Update on etio and immunopathogenesis of Kawasaki disease. Curr Opin Rheumatol. 2014;26:31–6.

    Article  CAS  PubMed  Google Scholar 

  78. Arora A, Wetter DA, Gonzalez-Santiago TM, Davis MDP, Lohse CM. Incidence of leukocytoclastic vasculitis, 1996–2010: a population-based study in Olmsted County, Minnesota. Mayo Clin Proc. 2014;89:1515–24.

    Article  PubMed  Google Scholar 

  79. Bouiller K, Audia S, Devilliers H, Collet E, Aubriot MH, Leguy-Seguin V, et al. Etiologies and prognostic factors of leukocytoclastic vasculitis with skin involvement. Medicine (Baltimore) [Internet]. 2016 [cited 2017 Nov 30];95. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4956825/

  80. Johnson E, Wetter D, Lehman J, Hand J, Davis D, Tollefson M. Leukocytoclastic vasculitis in children: clinical characteristics, subtypes, causes and direct immunofluorescence findings of 56 biopsy-confirmed cases. J Eur Acad Dermatol Venereol. 2017;31:544–9.

    Article  CAS  PubMed  Google Scholar 

  81. Kallenberg CGM. The diagnosis and classification of microscopic polyangiitis. J Autoimmun. 2014;48–49:90–3.

    Article  PubMed  CAS  Google Scholar 

  82. Greco A, De Virgilio A, Rizzo MI, Gallo A, Magliulo G, Fusconi M, et al. Microscopic polyangiitis: advances in diagnostic and therapeutic approaches. Autoimmun Rev. 2015;14:837–44.

    Article  CAS  PubMed  Google Scholar 

  83. Mahr A, Belarbi L, Wechsler B, Jeanneret D, Dhote R, Fain O, et al. Population-based prevalence study of Behçet’s disease: differences by ethnic origin and low variation by age at immigration. Arthritis Rheum. 2008;58:3951–9.

    Article  PubMed  Google Scholar 

  84. Saadoun D, Wechsler B. Behçet’s disease. Orphanet J Rare Dis. 2012;7:20.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Hatemi G, Silman A, Bang D, Bodaghi B, Chamberlain AM, Gul A, et al. EULAR recommendations for the management of Behçet disease. Ann Rheum Dis. 2008;67:1656–62.

    Article  CAS  PubMed  Google Scholar 

  86. Gilchrist H, Patterson JW. Erythema nodosum and erythema induratum (nodular vasculitis): diagnosis and management. Dermatol Ther. 2010;23:320–7.

    Article  PubMed  Google Scholar 

  87. Mascaró JM, Baselga E. Erythema induratum of Bazin. Dermatol Clin. 2008;26:439–45.

    Article  PubMed  Google Scholar 

  88. Caorsi R, Penco F, Grossi A, Insalaco A, Omenetti A, Alessio M, et al. ADA2 deficiency (DADA2) as an unrecognised cause of early onset polyarteritis nodosa and stroke: a multicentre national study. Ann Rheum Dis. 2017;76:1648–56.

    Article  CAS  PubMed  Google Scholar 

  89. De Virgilio A, Greco A, Magliulo G, Gallo A, Ruoppolo G, Conte M, et al. Polyarteritis nodosa: a contemporary overview. Autoimmun Rev. 2016;15:564–70.

    Article  PubMed  Google Scholar 

  90. Hajj-Ali RA, Calabrese LH. Diagnosis and classification of central nervous system vasculitis. J Autoimmun. 2014;48–49:149–52.

    Article  PubMed  Google Scholar 

  91. Salvarani C, Brown RD, Christianson T, Miller DV, Giannini C, Huston J, et al. An Update of the Mayo Clinic Cohort of Patients With Adult Primary Central Nervous System Vasculitis. Medicine (Baltimore) [Internet]. 2015 [cited 2017 Dec 4];94. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4616419/

  92. Salvarani C, Brown RD, Christianson TJH, Huston J, Giannini C, Miller DV, et al. Adult primary central nervous system vasculitis treatment and course: analysis of one hundred sixty-three patients. Arthritis Rheumatol. 2015;67:1637–45.

    Article  CAS  PubMed  Google Scholar 

  93. Calabrese LH, Mallek JA. Primary angiitis of the central nervous system. Report of 8 new cases, review of the literature, and proposal for diagnostic criteria. Medicine (Baltimore). 1988;67:20–39.

    Article  CAS  PubMed  Google Scholar 

  94. Trapani S, Micheli A, Grisolia F, Resti M, Chiappini E, Falcini F, et al. Henoch Schonlein Purpura in childhood: epidemiological and clinical analysis of 150 cases over a 5-year period and review of literature. Semin Arthritis Rheum. 2005;35:143–53.

    Article  PubMed  Google Scholar 

  95. Ozen S, Pistorio A, Iusan SM, Bakkaloglu A, Herlin T, Brik R, et al. EULAR, PRINTO, PRES criteria for Henoch–Schönlein purpura, childhood polyarteritis nodosa, childhood Wegener granulomatosis and childhood Takayasu arteritis: Ankara 2008. Part II: Final classification criteria. Ann Rheum Dis. 2008;2010(69):798–806.

    Google Scholar 

  96. Park SJ, Kim HJ, Park H, Hann HJ, Kim KH, Han S, et al. Incidence, prevalence, mortality and causes of death in Takayasu Arteritis in Korea—a nationwide, population-based study. Int J Cardiol. 2017;235:100–4.

    Article  PubMed  Google Scholar 

  97. Watts R, Al-Taiar A, Mooney J, Scott D, MacGregor A. The epidemiology of Takayasu arteritis in the UK. Rheumatology (Oxford). 2009;48:1008–11.

    Article  Google Scholar 

  98. Gudbrandsson B, Molberg Ø, Garen T, Palm Ø. Prevalence, incidence, and disease characteristics of takayasu arteritis by ethnic background: data from a large, population-based cohort resident in southern Norway. Arthritis Care Res. 2017;69:278–85.

    Article  Google Scholar 

  99. Birlik M, Kücükyavas Y, Aksu K, Solmaz D, Can G, Taylan A, et al. Epidemiology of Takayasu’s arteritis in Turkey. Clin Exp Rheumatol. 2016;34:S33-39.

    PubMed  Google Scholar 

  100. Saruhan-Direskeneli G, Hughes T, Aksu K, Keser G, Coit P, Aydin SZ, et al. Identification of multiple genetic susceptibility loci in Takayasu Arteritis. Am J Hum Genet. 2013;93:298–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Lutalo PMK, D’Cruz DP. Diagnosis and classification of granulomatosis with polyangiitis (aka Wegener’s granulomatosis). J Autoimmun. 2014;48–49:94–8.

    Article  PubMed  CAS  Google Scholar 

  102. Yamaguchi M, Ohta A, Tsunematsu T, Kasukawa R, Mizushima Y, Kashiwagi H, et al. Preliminary criteria for classification of adult Still’s disease. J Rheumatol. 1992;19:424–30.

    CAS  PubMed  Google Scholar 

  103. Fautrel B, Zing E, Golmard J, Le Moel G, Bissery A, Rioux C, et al. Proposal for a new set of classification criteria for adult-onset still disease. Medicine. 2002;81:194–200.

    Article  PubMed  Google Scholar 

  104. Wynne-Davies R, Hall C, Ansell BM. Spondylo-epiphysial dysplasia tarda with progressive arthropathy. A “new” disorder of autosomal recessive inheritance. Bone Joint J. 1982;64-B:442–5.

    Google Scholar 

  105. Wynne-Davies R, Gormley J. The prevalence of skeletal dysplasias. An estimate of their minimum frequency and the number of patients requiring orthopaedic care. Bone Joint J. 1985;67:133–7.

    CAS  Google Scholar 

  106. Delague V, Chouery E, Corbani S, Ghanem I, Aamar S, Fischer J, et al. Molecular study of WISP3 in nine families originating from the Middle-East and presenting with progressive pseudorheumatoid dysplasia: identification of two novel mutations, and description of a founder effect. Am J Med Genet A. 2005;138A:118–26.

    Article  PubMed  Google Scholar 

  107. Cassa CA, Smith SE, Docken W, Hoffman E, McLaughlin H, Chun S, et al. An argument for early genomic sequencing in atypical cases: a WISP3 variant leads to diagnosis of progressive pseudorheumatoid arthropathy of childhood. Rheumatology (Oxford). 2016;55:586–9.

    Google Scholar 

  108. Nakamura Y, Weidinger G, Liang JO, Aquilina-Beck A, Tamai K, Moon RT, et al. The CCN family member Wisp3, mutant in progressive pseudorheumatoid dysplasia, modulates BMP and Wnt signaling. J Clin Invest. 2007;117:3075–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Garcia Segarra N, Mittaz L, Campos-Xavier AB, Bartels CF, Tuysuz B, Alanay Y, et al. The diagnostic challenge of progressive pseudorheumatoid dysplasia (PPRD): a review of clinical features, radiographic features, and WISP3 mutations in 63 affected individuals. Am J Med Genet. 2012;160C:217–29.

    Article  PubMed  CAS  Google Scholar 

  110. Williams CJ, Zhang Y, Timms A, Bonavita G, Caeiro F, Broxholme J, et al. Autosomal dominant familial calcium pyrophosphate dihydrate deposition disease is caused by mutation in the transmembrane protein ANKH. Am J Hum Genet. 2002;71:985–91.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Andrew LJ, Brancolini V, de la Pena LS, Devoto M, Caeiro F, Marchegiani R, et al. Refinement of the chromosome 5p locus for familial calcium pyrophosphate dihydrate deposition disease. Am J Hum Genet. 1999;64:136–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Netter P, Bardin T, Bianchi A, Richette P, Loeuille D. The ANKH gene and familial calcium pyrophosphate dihydrate deposition disease. Joint Bone Spine. 2004;71:365–8.

    Article  PubMed  Google Scholar 

  113. Pendleton A, Johnson MD, Hughes A, Gurley KA, Ho AM, Doherty M, et al. Mutations in ANKH cause chondrocalcinosis. Am J Hum Genet. 2002;71:933–40.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Rosales-Alexander JL, Balsalobre Aznar J, Magro-Checa C. Calcium pyrophosphate crystal deposition disease: diagnosis and treatment. Open Access Rheumatol. 2014;6:39–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Verspoor FGM, Zee AAG, Hannink G, van der Geest IC, Veth RPH, et al. Long-term follow-up results of primary and recurrent pigmented villonodular synovitis. Rheumatology (Oxford). 2014;53:2063–70.

    Article  CAS  PubMed  Google Scholar 

  116. Myers BW, Masi AT. Pigmented villonodular synovitis and tenosynovitis: a clinical epidemiologic study of 166 cases and literature review. Medicine (Baltimore). 1980;59:223–38.

    Article  CAS  Google Scholar 

  117. Fiocco U, Sfriso P, Lunardi F, Pagnin E, Oliviero F, Scagliori E, et al. Molecular pathways involved in synovial cell inflammation and tumoral proliferation in diffuse pigmented villonodular synovitis. Autoimmun Rev. 2010;9:780–4.

    Article  CAS  PubMed  Google Scholar 

  118. Cassier PA, Italiano A, Gomez-Roca CA, Le Tourneau C, Toulmonde M, Cannarile MA, et al. CSF1R inhibition with emactuzumab in locally advanced diffuse-type tenosynovial giant cell tumours of the soft tissue: a dose-escalation and dose-expansion phase 1 study. Lancet Oncol. 2015;16:949–56.

    Article  CAS  PubMed  Google Scholar 

  119. Dwivedi N, Upadhyay J, Neeli I, Khan S, Pattanaik D, Myers L, et al. Felty’s syndrome autoantibodies bind to deiminated histones and neutrophil extracellular chromatin traps. Arthritis Rheum. 2012;64:982–92.

    Article  CAS  PubMed  Google Scholar 

  120. Hellmich B, Csernok E, Schatz H, Gross WL, Schnabel A. Autoantibodies against granulocyte colony-stimulating factor in Felty’s syndrome and neutropenic systemic lupus erythematosus. Arthritis Rheum. 2002;46:2384–91.

    Article  CAS  PubMed  Google Scholar 

  121. Balint GP, Balint PV. Felty’s syndrome. Best Pract Res Clin Rheumatol. 2004;18:631–45.

    Article  PubMed  Google Scholar 

  122. Narváez J, Domingo-Domenech E, Gómez-Vaquero C, López-Vives L, Estrada P, Aparicio M, et al. Biological agents in the management of felty’s syndrome: a systematic review. Semin Arthritis Rheum. 2012;41:658–68.

    Article  PubMed  CAS  Google Scholar 

  123. Li H, Altman RD, Yao Q. RS3PE: clinical and research development. Curr Rheumatol Rep. 2015;17:49.

    Article  CAS  PubMed  Google Scholar 

  124. Origuchi T, Arima K, Umeda M, Kawashiri S, Tamai M, Nakamura H, et al. Clinical outcomes in the first year of remitting seronegative symmetrical synovitis with pitting edema (RS3PE) syndrome. Mod Rheumatol. 2017;27:150–4.

    Article  CAS  PubMed  Google Scholar 

  125. Selmi C, Greenspan A, Huntley A, Gershwin ME. Multicentric Reticulohistiocytosis: a Critical Review. Current Rheumatology Reports [Internet]. 2015 [cited 2017 Sep 13];17. https://0-doi-org.brum.beds.ac.uk/10.1007/s11926-015-0511-6

  126. Islam AD, Naguwa SM, Cheema GS, Hunter JC, Gershwin ME. Multicentric reticulohistiocytosis: a rare yet challenging disease. Clin Rev Allergy Immunol. 2013;45:281–9.

    Article  PubMed  Google Scholar 

  127. Brackenridge A, Bashir T, Wheatley T. Multicentric reticulohistiocytosis and pregnancy. BJOG Int J Obst Gynaecol. 2005;112:672–3.

    Article  Google Scholar 

  128. Hayem G, Bouchaud-Chabot A, Benali K, Roux S, Palazzo E, Silbermann-Hoffman O, et al. SAPHO syndrome: a long-term follow-up study of 120 cases. Semin Arthritis Rheum. 1999;29:159–71.

    Article  CAS  PubMed  Google Scholar 

  129. Witt M, Meier J, Hammitzsch A, Proft F, Schulze-Koops H, Grunke M. Disease burden, disease manifestations and current treatment regimen of the SAPHO syndrome in Germany: results from a nationwide patient survey. Semin Arthritis Rheum. 2014;43:745–50.

    Article  PubMed  Google Scholar 

  130. Nguyen MT, Borchers A, Selmi C, Naguwa SM, Cheema G, Gershwin ME. The SAPHO syndrome. Semin Arthritis Rheum. 2012;42:254–65.

    Article  PubMed  Google Scholar 

  131. Zimmermann P, Curtis N. Synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome—a challenging diagnosis not to be missed. J Infect. 2016;72:S106–14.

    Article  PubMed  Google Scholar 

  132. Rukavina I. SAPHO syndrome: a review. J Child Orthop. 2015;9:19–27.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Prakken B, Albani S, Martini A. Juvenile idiopathic arthritis. The Lancet. 2011;377:2138–49.

    Article  Google Scholar 

  134. Vastert SJ, Kuis W, Grom AA. Systemic JIA: new developments in the understanding of the pathophysiology and therapy. Best Pract Res Clin Rheumatol. 2009;23:655–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Woo P. Systemic juvenile idiopathic arthritis: diagnosis, management, and outcome. Nat Clin Pract Rheumatol. 2006;2:28–34.

    Article  PubMed  Google Scholar 

  136. Petty RE, Southwood TR, Manners P, Baum J, Glass DN, Goldenberg J, et al. International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol. 2004;31:390–2.

    PubMed  Google Scholar 

  137. Ombrello MJ, Arthur VL, Remmers EF, Hinks A, Tachmazidou I, Grom AA, et al. Genetic architecture distinguishes systemic juvenile idiopathic arthritis from other forms of juvenile idiopathic arthritis: clinical and therapeutic implications. Ann Rheum Dis. 2017;76:906–13.

    Article  CAS  PubMed  Google Scholar 

  138. Ayaz NA, Özen S, Bilginer Y, Ergüven M, Taşkıran E, Yılmaz E, et al. MEFV mutations in systemic onset juvenile idiopathic arthritis. Rheumatology (Oxford). 2009;48:23–5.

    Article  CAS  Google Scholar 

  139. De Benedetti F, Meazza C, Vivarelli M, Rossi F, Pistorio A, Lamb R, et al. Functional and prognostic relevance of the −173 polymorphism of the macrophage migration inhibitory factor gene in systemic-onset juvenile idiopathic arthritis. Arthritis Rheum. 2003;48:1398–407.

    Article  PubMed  CAS  Google Scholar 

  140. Quartier P, Allantaz F, Cimaz R, Pillet P, Messiaen C, Bardin C, et al. A multicentre, randomised, double-blind, placebo-controlled trial with the interleukin-1 receptor antagonist anakinra in patients with systemic-onset juvenile idiopathic arthritis (ANAJIS trial). Ann Rheum Dis. 2010;70:747–54.

    Article  PubMed  CAS  Google Scholar 

  141. van Dijkhuizen EHP, Wulffraat NM. Early predictors of prognosis in juvenile idiopathic arthritis: a systematic literature review. Ann Rheum Dis. 2015;74:1996–2005.

    Article  PubMed  CAS  Google Scholar 

  142. Edouard S, Fenollar F, Raoult D. The rise of Tropheryma whipplei: a 12-year retrospective study of PCR diagnoses in our reference center. J Clin Microbiol. 2012;50:3917–20.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Ramharter M, Harrison N, Bühler T, Herold B, Lagler H, Lötsch F, et al. Prevalence and risk factor assessment of Tropheryma whipplei in a rural community in Gabon: a community-based cross-sectional study. Clin Microbiol Infect. 2014;20:1189–94.

    Article  CAS  PubMed  Google Scholar 

  144. Fenollar F, Trani M, Davoust B, Salle B, Birg M, Rolain J, et al. Prevalence of asymptomatic Tropheryma whipplei carriage among humans and nonhuman primates. J Infect Dis. 2008;197:880–7.

    Article  CAS  PubMed  Google Scholar 

  145. Feurle GE, Junga NS, Marth T. Efficacy of ceftriaxone or meropenem as initial Therapies in Whipple’s disease. Gastroenterology. 2010;138:478–86.

    Article  CAS  PubMed  Google Scholar 

  146. Feurle GE, Moos V, Bläker H, Loddenkemper C, Moter A, Stroux A, et al. Intravenous ceftriaxone, followed by 12 or three months of oral treatment with trimethoprim-sulfamethoxazole in Whipple’s disease. J Infect. 2013;66:263–70.

    Article  PubMed  Google Scholar 

  147. Feurle GE, Moos V, Schinnerling K, Geelhaar A, Allers K, Biagi F, et al. The immune reconstitution inflammatory syndrome in whipple disease: a cohort study. Ann Intern Med. 2010;153:710–7.

    Article  PubMed  Google Scholar 

  148. Lagier J-C, Fenollar F, Lepidi H, Giorgi R, Million M, Raoult D. Treatment of classic Whipple’s disease: from in vitro results to clinical outcome. J Antimicrob Chemother. 2014;69:219–27.

    Article  CAS  PubMed  Google Scholar 

  149. Moos V, Schneider T. Tropheryma whipplei: Erreger verschiedener Infektionen und Pathogen des Morbus Whipple. Dtsch med Wochenschr. 2015;140:428–32.

    Article  PubMed  Google Scholar 

  150. Pareek A, Sanders TL, Wu IT, Larson DR, Saris DBF, Krych AJ. Incidence of symptomatic osteochondritis dissecans lesions of the knee: a population-based study in Olmsted County. Osteoarthr Cartil. 2017;25:1663–71.

    Article  CAS  Google Scholar 

  151. Chambers HG, Shea KG, Carey JL. AAOS Clinical Practice Guideline: diagnosis and treatment of osteochondritis dissecans. J Am Acad Orthop Surg. 2011;19:307–9.

    Article  PubMed  Google Scholar 

  152. Kocher MS, Tucker R, Ganley TJ, Flynn JM. Management of Osteochondritis Dissecans of the Knee: Current Concepts Review. Am J Sports Med. 2006;34:1181–91.

    Article  PubMed  Google Scholar 

  153. Aróstegui JI, Arnal C, Merino R, Modesto C, Antonia Carballo M, Moreno P, et al. NOD2 gene-associated pediatric granulomatous arthritis: clinical diversity, novel and recurrent mutations, and evidence of clinical improvement with interleukin-1 blockade in a Spanish cohort. Arthritis Rheum. 2007;56:3805–13.

    Article  PubMed  CAS  Google Scholar 

  154. Rosé CD, Aróstegui JI, Martin TM, Espada G, Scalzi L, Yagüe J, et al. NOD2-associated pediatric granulomatous arthritis, an expanding phenotype: study of an international registry and a national cohort in Spain. Arthritis Rheum. 2009;60:1797–803.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Caso F, Galozzi P, Costa L, Sfriso P, Cantarini L, Punzi L. Autoinflammatory granulomatous diseases: from Blau syndrome and early-onset sarcoidosis to NOD2-mediated disease and Crohn’s disease. RMD Open [Internet]. 2015 [cited 2017 Jun 27];1. Available from: http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4612691/

  156. Rosé CD, Pans S, Casteels I, Anton J, Bader-Meunier B, Brissaud P, et al. Blau syndrome: cross-sectional data from a multicentre study of clinical, radiological and functional outcomes. Rheumatology (Oxford). 2015;54:1008–16.

    Article  CAS  Google Scholar 

  157. Hoffmann AL, Milman N, Byg KE. Childhood sarcoidosis in Denmark 1979–1994: incidence, clinical features and laboratory results at presentation in 48 children. Acta Paediatr. 2004;93:30–6.

    Article  CAS  PubMed  Google Scholar 

  158. Mensa-Vilaro A, Cham WT, Tang SP, Lim SC, González-Roca E, Ruiz-Ortiz E, et al. Brief Report: First Identification of Intrafamilial Recurrence of Blau Syndrome due to Gonosomal NOD2 Mosaicism. Arthritis Rheumatol (Hoboken, NJ). 2016;68:1039–44.

    Article  CAS  Google Scholar 

  159. Finetti M, Omenetti A, Federici S, Caorsi R, Gattorno M. Chronic Infantile Neurological Cutaneous and Articular (CINCA) syndrome: a review. Orphanet J Rare Dis [Internet]. 2016 [cited 2017 Sep 21];11. Available from: http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5142346/

  160. Cuisset L, Jeru I, Dumont B, Fabre A, Cochet E, Le Bozec J, et al. Mutations in the autoinflammatory cryopyrin-associated periodic syndrome gene: epidemiological study and lessons from eight years of genetic analysis in France. Ann Rheum Dis. 2011;70:495–9.

    Article  CAS  PubMed  Google Scholar 

  161. Kuemmerle-Deschner JB. CAPS—pathogenesis, presentation and treatment of an autoinflammatory disease. Semin Immunopathol. 2015;37:377–85.

    Article  CAS  PubMed  Google Scholar 

  162. Koné-Paut I, Hentgen V, Touitou I. Current data on familial Mediterranean fever. Joint Bone Spine. 2011;78:111–4.

    Article  PubMed  Google Scholar 

  163. Ozen S, Demirkaya E, Amaryan G, Koné-Paut I, Polat A, Woo P, et al. Results from a multicentre international registry of familial Mediterranean fever: impact of environment on the expression of a monogenic disease in children. Ann Rheum Dis. 2014;73:662–7.

    Article  PubMed  Google Scholar 

  164. Sari I, Birlik M, Kasifoglu T. Familial Mediterranean fever: an updated review. Eur J Rheumatol. 2014;1:21–33.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Yalçınkaya F, Özen S, Özçakar ZB, Aktay N, Çakar N, Düzova A, et al. A new set of criteria for the diagnosis of familial Mediterranean fever in childhood. Rheumatology (Oxford). 2009;48:395–8.

    Article  Google Scholar 

  166. Livneh A, Langevitz P, Zemer D, Zaks N, Kees S, Lidar T, et al. Criteria for the diagnosis of familial mediterranean fever. Arthritis Rheum. 1997;40:1879–85.

    Article  CAS  PubMed  Google Scholar 

  167. Haas D, Hoffmann GF. Mevalonate kinase deficiencies: from mevalonic aciduria to hyperimmunoglobulinemia D syndrome. Orphanet J Rare Dis. 2006;1:13.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Lainka E, Neudorf U, Lohse P, Timmann C, Bielak M, Stojanov S, et al. Incidence and clinical features of hyperimmunoglobulinemia D and periodic fever syndrome (HIDS) and spectrum of mevalonate kinase (MVK) mutations in German children. Rheumatol Int. 2012;32:3253–60.

    Article  CAS  PubMed  Google Scholar 

  169. Ozen S, Kuemmerle-Deschner JB, Cimaz R, Livneh A, Quartier P, Kone-Paut I, et al. International retrospective chart review of treatment patterns in severe familial mediterranean fever, tumor necrosis factor receptor-associated periodic syndrome, and mevalonate kinase deficiency/hyperimmunoglobulinemia d syndrome: real-world experience in three periodic fever syndromes. Arthritis Care Res. 2017;69:578–86.

    Article  CAS  Google Scholar 

  170. van der Hilst JCH, Bodar EJ, Barron KS, Frenkel J, Drenth JPH, van der Meer JWM, et al. Long-term follow-up, clinical features, and quality of life in a series of 103 patients with hyperimmunoglobulinemia D syndrome. Medicine. 2008;87:301–10.

    Article  PubMed  CAS  Google Scholar 

  171. Arima K, Kinoshita A, Mishima H, Kanazawa N, Kaneko T, Mizushima T, et al. Proteasome assembly defect due to a proteasome subunit beta type 8 (PSMB8) mutation causes the autoinflammatory disorder, Nakajo–Nishimura syndrome. Proc Natl Acad Sci USA. 2011;108:14914–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. McDermott A, Jacks J, Kessler M, Emanuel PD, Gao L. Proteasome-associated autoinflammatory syndromes: advances in pathogeneses, clinical presentations, diagnosis, and management. Int J Dermatol. 2015;54:121–9.

    Article  PubMed  Google Scholar 

  173. Kanazawa N. Nakajo-Nishimura syndrome: an autoinflammatory disorder showing pernio-like rashes and progressive partial lipodystrophy. Allergol Int. 2012;61:197–206.

    Article  CAS  PubMed  Google Scholar 

  174. Schellevis MA, Stoffels M, Hoppenreijs EPAH, Bodar E, Simon A, van der Meer JWM. Variable expression and treatment of PAPA syndrome. Ann Rheum Dis. 2011;70:1168–70.

    Article  PubMed  Google Scholar 

  175. Wise CA, Gillum JD, Seidman CE, Lindor NM, Veile R, Bashiardes S, et al. Mutations in CD2BP1 disrupt binding to PTP PEST and are responsible for PAPA syndrome, an autoinflammatory disorder. Hum Mol Genet. 2002;11:961–9.

    Article  CAS  PubMed  Google Scholar 

  176. Lindor NM, Arsenault TM, Solomon H, Seidman CE, McEvoy MT. A new autosomal dominant disorder of pyogenic sterile arthritis, pyoderma gangrenosum, and acne: PAPA syndrome. Mayo Clin Proc. 1997;72:611–5.

    Article  CAS  PubMed  Google Scholar 

  177. Yeon HB, Lindor NM, Seidman JG, Seidman CE. Pyogenic arthritis, pyoderma gangrenosum, and acne syndrome maps to chromosome 15q. Am J Hum Genet. 2000;66:1443–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Førsvoll J, Kristoffersen EK, Øymar K. Incidence, clinical characteristics and outcome in Norwegian children with periodic fever, aphthous stomatitis, pharyngitis and cervical adenitis syndrome; a population-based study. Acta Paediatr. 2013;102:187–92.

    Article  PubMed  Google Scholar 

  179. Thomas KT, Feder HM, Lawton AR, Edwards KM. Periodic fever syndrome in children. J Pediatr. 1999;135:15–21.

    Article  CAS  PubMed  Google Scholar 

  180. Vanoni F, Theodoropoulou K, Hofer M. PFAPA syndrome: a review on treatment and outcome. Pediatr Rheumatol Online J [Internet]. 2016 [cited 2017 Oct 12];14. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4924332/

  181. Marshall GS, Edwards KM, Butler J, Lawton AR. Syndrome of periodic fever, pharyngitis, and aphthous stomatitis. J Pediatr. 1987;110:43–6.

    Article  CAS  PubMed  Google Scholar 

  182. Cantarini L, Vitale A, Sicignano LL, Emmi G, Verrecchia E, Patisso I, et al. Diagnostic Criteria for Adult-Onset Periodic Fever, Aphthous Stomatitis, Pharyngitis, and Cervical Adenitis (PFAPA) Syndrome. Front Immunol [Internet]. 2017 [cited 2018 Sep 5];8. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5609591/

  183. Simon A, Asli B, Braun-Falco M, De Koning H, Fermand J-P, Grattan C, et al. Schnitzler’s syndrome: diagnosis, treatment, and follow-up. Allergy. 2013;68:562–8.

    Article  CAS  PubMed  Google Scholar 

  184. Krause K, Tsianakas A, Wagner N, Fischer J, Weller K, Metz M, et al. Efficacy and safety of canakinumab in Schnitzler syndrome: a multicenter randomized placebo-controlled study. J Allergy Clin Immunol. 2017;139:1311–20.

    Article  CAS  PubMed  Google Scholar 

  185. Tinazzi E, Puccetti A, Patuzzo G, Sorleto M, Barbieri A, Lunardi C. Schnitzler syndrome, an autoimmune–autoinflammatory syndrome: Report of two new cases and review of the literature. Autoimmun Rev. 2011;10:404–9.

    Article  PubMed  Google Scholar 

  186. Gusdorf L, Asli B, Barbarot S, Néel A, Masseau A, Puéchal X, et al. Schnitzler syndrome: validation and applicability of diagnostic criteria in real-life patients. Allergy. 2017;72:177–82.

    Article  CAS  PubMed  Google Scholar 

  187. Vastert SJ, van Wijk R, D’Urbano LE, de Vooght KMK, de Jager W, Ravelli A, et al. Mutations in the perforin gene can be linked to macrophage activation syndrome in patients with systemic onset juvenile idiopathic arthritis. Rheumatology (Oxford). 2010;49:441–9.

    Article  CAS  Google Scholar 

  188. Davì S, Minoia F, Pistorio A, Horne A, Consolaro A, Rosina S, et al. Performance of current guidelines for diagnosis of macrophage activation syndrome complicating systemic juvenile idiopathic arthritis. Arthritis Rheumatol. 2014;66:2871–80.

    Article  PubMed  CAS  Google Scholar 

  189. Ravelli A, Minoia F, Davì S, Horne A, Bovis F, Pistorio A, et al. Classification criteria for macrophage activation syndrome complicating systemic juvenile idiopathic arthritis. Ann Rheum Dis Ann Rheum Dis. 2016;75:481–9.

    Article  CAS  PubMed  Google Scholar 

  190. Henter J-I, Horne A, Aricó M, Egeler RM, Filipovich AH, Imashuku S, et al. HLH-2004: Diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2007;48:124–31.

    Article  PubMed  Google Scholar 

  191. Minoia F, Davì S, Horne A, Demirkaya E, Bovis F, Li C, et al. Clinical features, treatment, and outcome of macrophage activation syndrome complicating systemic juvenile idiopathic arthritis: a multinational, multicenter study of 362 patients. Arthritis Rheumatol. 2014;66:3160–9.

    Article  PubMed  Google Scholar 

  192. Ruscitti P, Cipriani P, Ciccia F, Masedu F, Liakouli V, Carubbi F, et al. Prognostic factors of macrophage activation syndrome, at the time of diagnosis, in adult patients affected by autoimmune disease: analysis of 41 cases collected in 2 rheumatologic centers. Autoimmun Rev. 2017;16:16–21.

    Article  CAS  PubMed  Google Scholar 

  193. Ferguson P, Chen S, Tayeh M, Ochoa L, Leal S, Pelet A, et al. Homozygous mutations in LPIN2 are responsible for the syndrome of chronic recurrent multifocal osteomyelitis and congenital dyserythropoietic anaemia (Majeed syndrome). J Med Genet. 2005;42:551–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. El-Shanti H, Ferguson P. Majeed Syndrome. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJ, Mefford HC, et al., editors. GeneReviews(®) [Internet]. Seattle (WA): University of Washington, Seattle; 1993 [cited 2017 Nov 23]. Available from: http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/books/NBK1974/

  195. Herlin T, Fiirgaard B, Bjerre M, Kerndrup G, Hasle H, Bing X, et al. Efficacy of anti-IL-1 treatment in Majeed syndrome. Ann Rheum Dis. 2013;72:410–3.

    Article  CAS  PubMed  Google Scholar 

  196. Lachmann HJ, Papa R, Gerhold K, Obici L, Touitou I, Cantarini L, et al. The phenotype of TNF receptor-associated autoinflammatory syndrome (TRAPS) at presentation: a series of 158 cases from the Eurofever/EUROTRAPS international registry. Ann Rheum Dis. 2014;73:2160–7.

    Article  CAS  PubMed  Google Scholar 

  197. Lainka E, Neudorf U, Lohse P, Timmann C, Stojanov S, Huss K, et al. Incidence of TNFRSF1A mutations in German children: epidemiological, clinical and genetic characteristics. Rheumatology. 2009;48:987–91.

    Article  CAS  PubMed  Google Scholar 

  198. Bachetti T, Chiesa S, Castagnola P, Bani D, Zanni ED, Omenetti A, et al. Autophagy contributes to inflammation in patients with TNFR-associated periodic syndrome (TRAPS). Ann Rheum Dis. 2013;72:1044–52.

    Article  CAS  PubMed  Google Scholar 

  199. Allenbach Y, Drouot L, Rigolet A, Charuel JL, Jouen F, Romero NB, et al. Anti-HMGCR Autoantibodies in European Patients With Autoimmune Necrotizing Myopathies. Medicine (Baltimore) [Internet]. 2014 [cited 2017 Nov 18];93. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4632910/

  200. Carroll MB, Newkirk MR, Sumner NS. Necrotizing autoimmune myopathy: a unique subset of idiopathic inflammatory myopathy. J Clin Rheumatol. 2016;22:376–80.

    Article  PubMed  Google Scholar 

  201. Targoff IN. Autoantibodies and their significance in myositis. Curr Rheumatol Rep. 2008;10:333–40.

    Article  CAS  PubMed  Google Scholar 

  202. Manole C, Inimioara Mihaela C, Bogdan C. New insights into antisynthetase syndrome. Maedica (Buchar). 2016;11:130–5.

    Google Scholar 

  203. Lega J-C, Fabien N, Reynaud Q, Durieu I, Durupt S, Dutertre M, et al. The clinical phenotype associated with myositis-specific and associated autoantibodies: a meta-analysis revisiting the so-called antisynthetase syndrome. Autoimmun Rev. 2014;13:883–91.

    Article  CAS  PubMed  Google Scholar 

  204. Cavagna L, Nuño L, Scirè CA, Govoni M, Longo FJL, Franceschini F, et al. Serum Jo-1 autoantibody and isolated arthritis in the antisynthetase syndrome: review of the literature and report of the experience of AENEAS Collaborative Group. Clinic Rev Allerg Immunol. 2017;52:71–80.

    Article  CAS  Google Scholar 

  205. Lehmann D, Motlagh L, Robaa D, Zierz S. Muscle Carnitine Palmitoyltransferase II Deficiency: A Review of Enzymatic Controversy and Clinical Features. Int J Mol Sci [Internet]. 2017 [cited 2017 Nov 18];18. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5297716/

  206. Sigauke E, Rakheja D, Kitson K, Bennett MJ. Carnitine palmitoyltransferase II deficiency: a clinical, biochemical, and molecular review. Lab Invest. 2003;83:1543–54.

    Article  CAS  PubMed  Google Scholar 

  207. Ohta A, Nagai M, Nishina M, Tomimitsu H, Kohsaka H. Prevalence and incidence of polymyositis and dermatomyositis in Japan. Mod Rheumatol. 2014;24:477–80.

    Article  PubMed  Google Scholar 

  208. Dobloug C, Garen T, Bitter H, Stjärne J, Stenseth G, Grøvle L, et al. Prevalence and clinical characteristics of adult polymyositis and dermatomyositis; data from a large and unselected Norwegian cohort. Ann Rheum Dis. 2015;74:1551–6.

    Article  PubMed  Google Scholar 

  209. Rosa J, Garrot LF, Navarta DA, Saucedo C, Scolnik M, Bedran Z, et al. Incidence and Prevalence of Polymyositis and Dermatomyositis in a Health Management Organization in Buenos Aires. J Clin Rheumatol. 2013;19:303–7.

    Article  PubMed  Google Scholar 

  210. Malik A, Hayat G, Kalia JS, Guzman MA. Idiopathic Inflammatory Myopathies: Clinical Approach and Management. Front Neurol [Internet]. 2016 [cited 2017 Nov 19];7. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4873503/

  211. Dobloug GC, Antal EA, Sveberg L, Garen T, Bitter H, Stjärne J, et al. High prevalence of inclusion body myositis in Norway; a population-based clinical epidemiology study. Eur J Neurol. 2015;22:672-e41.

    Article  CAS  PubMed  Google Scholar 

  212. Paltiel AD, Ingvarsson E, Lee DKK, Leff RL, Nowak RJ, Petschke KD, et al. Demographic and clinical features of inclusion body myositis in North America. Muscle Nerve. 2015;52:527–33.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Allen JA, Peterson A, Sufit R, Hinchcliff ME, Mahoney JM, Wood TA, et al. Post-epidemic eosinophilia myalgia syndrome associated with L-Tryptophan. Arthritis Rheum [Internet]. 2011 [cited 2017 Nov 9];63. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC3848710/

  214. Devic P, Gallay L, Streichenberger N, Petiot P. Focal myositis: a review. Neuromuscul Disord. 2016;26:725–33.

    Article  CAS  PubMed  Google Scholar 

  215. Auerbach A, Fanburg-smith JC, Wang G, Rushing EJ. Focal myositis: a clinicopathologic study of 115 cases of an intramuscular mass-like reactive process. Am J Surg Pathol. 2009;33:1016–24.

    Article  PubMed  Google Scholar 

  216. De Castro M, Johnston J, Biesecker L. Determining the prevalence of McArdle disease from gene frequency by analysis of next generation sequencing data. Genet Med. 2015;17:1002–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Santalla A, Nogales-Gadea G, Encinar AB, Vieitez I, González-Quintana A, Serrano-Lorenzo P, et al. Genotypic and phenotypic features of all Spanish patients with McArdle disease: a 2016 update. BMC Genomics. 2017;18:819.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  218. Nogales-Gadea G, Santalla A, Brull A, de Luna N, Lucia A, Pinós T. The pathogenomics of McArdle disease—genes, enzymes, models, and therapeutic implications. J Inherit Metab Dis. 2015;38:221–30.

    Article  CAS  PubMed  Google Scholar 

  219. Nogales-Gadea G, Pinós T, Andreu AL, Martín MA, Arenas J, Lucia A. Next-generation sequencing to estimate the prevalence of a great unknown: McArdle disease. Genetics in Medicine. 2015;17:gim201576.

  220. Musumeci O, Bruno C, Mongini T, Rodolico C, Aguennouz M, Barca E, et al. Clinical features and new molecular findings in muscle phosphofructokinase deficiency (GSD type VII). Neuromuscul Disord. 2012;22:325–30.

    Article  PubMed  Google Scholar 

  221. Toscano A, Musumeci O. Tarui disease and distal glycogenoses: clinical and genetic update. Acta Myol. 2007;26:105–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Kinoshita A, Saito T, Tomita H, Makita Y, Yoshida K, Ghadami M, et al. Domain-specific mutations in TGFB1 result in Camurati–Engelmann disease. Nat Genet. 2000;26:ng900_19.

    Article  CAS  Google Scholar 

  223. Simsek-Kiper PO, Dikoglu E, Campos-Xavier B, Utine GE, Bonafe L, Unger S, et al. Positive effects of an angiotensin II type 1 receptor antagonist in Camurati–Engelmann disease: a single case observation. Am J Med Genet. 2014;164:2667–71.

    Article  CAS  Google Scholar 

  224. Morales-Piga A, Bachiller-Corral J, Trujillo-Tiebas MJ, Villaverde-Hueso A, Gamir-Gamir ML, Alonso-Ferreira V, et al. Fibrodysplasia ossificans progressiva in Spain: epidemiological, clinical, and genetic aspects. Bone. 2012;51:748–55.

    Article  CAS  PubMed  Google Scholar 

  225. Baujat G, Choquet R, Bouée S, Jeanbat V, Courouve L, Ruel A, et al. Prevalence of fibrodysplasia ossificans progressiva (FOP) in France: an estimate based on a record linkage of two national databases. Orphanet J Rare Dis [Internet]. 2017 [cited 2017 Nov 17];12. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5493013/

  226. Heursen E-M, Partida M del CG, Expósito JP, Díaz FN. Osteomesopyknosis—a benign axial hyperostosis that can mimic metastatic disease. Skeletal Radiol. 2016;45:141–6.

  227. Meikle PJ, Hopwood JJ, Clague AE, Carey WF. Prevalence of lysosomal storage disorders. JAMA. 1999;281:249–54.

    Article  CAS  PubMed  Google Scholar 

  228. Biegstraaten M, Arngrímsson R, Barbey F, Boks L, Cecchi F, Deegan PB, et al. Recommendations for initiation and cessation of enzyme replacement therapy in patients with Fabry disease: the European Fabry Working Group consensus document. Orphanet J Rare Dis. 2015;10:36.

    Article  PubMed  PubMed Central  Google Scholar 

  229. Elliott PM, Germain DP, Hilz MJ, Spada M, Wanner C, Falissard B. Why systematic literature reviews in Fabry disease should include all published evidence. European Journal of Medical Genetics. 2019;62:103702.

  230. Ehlert K, Frosch M, Fehse N, Zander A, Roth J, Vormoor J. Farber disease: clinical presentation, pathogenesis and a new approach to treatment. Pediatr Rheumatol. 2007;5:15.

    Article  Google Scholar 

  231. Alves MQ, Le Trionnaire E, Ribeiro I, Carpentier S, Harzer K, Levade T, et al. Molecular basis of acid ceramidase deficiency in a neonatal form of Farber disease: Identification of the first large deletion in ASAH1 gene. Mol Genet Metab. 2013;109:276–81.

    Article  CAS  PubMed  Google Scholar 

  232. Orenstein M, Barbouth D, Bodamer OA, Weinreb NJ. Patients with type 1 Gaucher disease in South Florida, USA: demographics, genotypes, disease severity and treatment outcomes. Orphanet J Rare Dis. 2014;9:45.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Stirnemann J, Vigan M, Hamroun D, Heraoui D, Rossi-Semerano L, Berger MG, et al. The French Gaucher’s disease registry: clinical characteristics, complications and treatment of 562 patients. Orphanet J Rare Dis. 2012;7:77.

    Article  PubMed  PubMed Central  Google Scholar 

  234. Stirnemann J, Belmatoug N, Camou F, Serratrice C, Froissart R, Caillaud C, et al. A Review of Gaucher Disease Pathophysiology, Clinical Presentation and Treatments. Int J Mol Sci [Internet]. 2017 [cited 2017 Nov 24];18. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5343975/

  235. Nalysnyk L, Rotella P, Simeone JC, Hamed A, Weinreb N. Gaucher disease epidemiology and natural history: a comprehensive review of the literature. Hematology. 2017;22:65–73.

    Article  PubMed  Google Scholar 

  236. Gurevich E, Hershkovitz E, Yarza S, Landau D. High prevalence of hypophosphatasia in Southern Israel. Acta Paediatr. 2020;109:851–2.

    Article  PubMed  Google Scholar 

  237. Högler W, Langman C, Gomes da Silva H, Fang S, Linglart A, Ozono K, et al. Diagnostic delay is common among patients with hypophosphatasia: initial findings from a longitudinal, prospective, global registry. BMC Musculoskelet Disord [Internet]. 2019 [cited 2020 Sep 7];20. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC6376686/

  238. Mornet E, Yvard A, Taillandier A, Fauvert D, Simon-Bouy B. A Molecular-Based Estimation of the Prevalence of Hypophosphatasia in the European Population. Ann Hum Genet. 2011;75:439–45.

    Article  PubMed  Google Scholar 

  239. Montaño AM, Tomatsu S, Gottesman GS, Smith M, Orii T. International Morquio A Registry: clinical manifestation and natural course of Morquio A disease. J Inherit Metab Dis. 2007;30:165–74.

    Article  PubMed  Google Scholar 

  240. Leadley RM, Lang S, Misso K, Bekkering T, Ross J, Akiyama T, et al. A systematic review of the prevalence of Morquio A syndrome: challenges for study reporting in rare diseases. Orphanet J Rare Dis [Internet]. 2014 [cited 2017 Nov 24];9. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4251694/

  241. Hendriksz CJ, Berger KI, Giugliani R, Harmatz P, Kampmann C, Mackenzie WG, et al. International guidelines for the management and treatment of morquio a syndrome. Am J Med Genet A. 2015;167:11–25.

    Article  Google Scholar 

  242. Kotwal A, Clarke BL. Melorheostosis: a rare sclerosing bone dysplasia. Curr Osteoporos Rep. 2017;15:335–42.

    Article  PubMed  Google Scholar 

  243. Smith GC, Pingree MJ, Freeman LA, Matsumoto JM, Howe BM, Kannas SN, et al. Melorheostosis: a retrospective clinical analysis of 24 patients at the Mayo Clinic. PM&R. 2017;9:283–8.

    Article  Google Scholar 

  244. Freyschmidt J. Melorheostosis: a review of 23 cases. Eur Radiol. 2001;11:474–9.

    Article  CAS  PubMed  Google Scholar 

  245. Busch J, Frank V, Bachmann N, Otsuka A, Oji V, Metze D, et al. Mutations in the prostaglandin transporter SLCO2A1 cause primary hypertrophic osteoarthropathy with digital clubbing. J Investig Dermatol. 2012;132:2473–6.

    Article  CAS  PubMed  Google Scholar 

  246. Lee S, Park SY, Kwon HJ, Lee C-H, Kim O-H, Rhee Y. Identification of the mutations in the prostaglandin transporter gene, SLCO2A1 and clinical characterization in korean patients with pachydermoperiostosis. J Korean Med Sci. 2016;31:735–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Uppal S, Diggle CP, Carr IM, Fishwick CWG, Ahmed M, Ibrahim GH, et al. Mutations in 15-hydroxyprostaglandin dehydrogenase cause primary hypertrophic osteoarthropathy. Nat Genet. 2008;40:789–93.

    Article  CAS  PubMed  Google Scholar 

  248. Jurecka A, Ługowska A, Golda A, Czartoryska B, Tylki-Szymańska A. Prevalence rates of mucopolysaccharidoses in Poland. J Appl Genet. 2015;56:205–10.

    Article  PubMed  Google Scholar 

  249. Malm G, Lund AM, Månsson J-E, Heiberg A. Mucopolysaccharidoses in the Scandinavian countries: incidence and prevalence. Acta Pædiatrica. 2008;97:1577–81.

    Article  PubMed  Google Scholar 

  250. Guffon N, Heron B, Chabrol B, Feillet F, Montauban V, Valayannopoulos V. Diagnosis, quality of life, and treatment of patients with Hunter syndrome in the French healthcare system: a retrospective observational study. Orphanet J Rare Dis [Internet]. 2015 [cited 2017 Nov 24];10. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4407793/

  251. Kitaoka T, Miyoshi Y, Namba N, Miura K, Kubota T, Ohata Y, et al. Two Japanese familial cases of Caffey disease with and without the common COL1A1 mutation and normal bone density, and review of the literature. Eur J Pediatr. 2014;173:799–804.

    Article  PubMed  Google Scholar 

  252. Gensure RC, Mäkitie O, Barclay C, Chan C, DePalma SR, Bastepe M, et al. A novel COL1A1 mutation in infantile cortical hyperostosis (Caffey disease) expands the spectrum of collagen-related disorders. J Clin Invest. 2005;115:1250–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Guerin A, Dupuis L, Mendoza-Londono R. Caffey Disease. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJ, Mefford HC, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993 [cited 2017 Dec 5]. Available from: http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/books/NBK99168/

  254. Germain DP. Ehlers-Danlos syndrome type IV. Orphanet J Rare Dis. 2007;2:32.

    Article  PubMed  PubMed Central  Google Scholar 

  255. De Paepe A, Malfait F. The Ehlers–Danlos syndrome, a disorder with many faces. Clin Genet. 2012;82:1–11.

    Article  PubMed  CAS  Google Scholar 

  256. Malfait F, Francomano C, Byers P, Belmont J, Berglund B, Black J, et al. The 2017 international classification of the Ehlers–Danlos syndromes. Am J Med Genet. 2017;175:8–26.

    Article  PubMed  Google Scholar 

  257. Gorospe L, Ayala-Carbonero AM, Fernández-Méndez MÁ, Arrieta P, Muñoz-Molina GM, Cabañero-Sánchez A, et al. Idiopathic fibrosing mediastinitis: spectrum of imaging findings with emphasis on its association with IgG4-related disease. Clin Imaging. 2015;39:993–9.

    Article  PubMed  Google Scholar 

  258. Rossi SE, McAdams HP, Rosado-de-Christenson ML, Franks TJ, Galvin JR. Fibrosing mediastinitis. Radiographics. 2001;21:737–57.

    Article  CAS  PubMed  Google Scholar 

  259. Wu Z, Jarvis H, Howard LS, Wright C, Kon OM. Post-tuberculous fibrosing mediastinitis: a review of the literature. BMJ Open Respir Res [Internet]. 2017 [cited 2017 Dec 8];4. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5501238/

  260. Uchida K, Masamune A, Shimosegawa T, Okazaki K. Prevalence of IgG4-Related Disease in Japan Based on Nationwide Survey in 2009. Int J Rheumatol [Internet]. 2012 [cited 2020 Sep 7];2012. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC3415093/

  261. Umehara H, Okazaki K, Masaki Y, Kawano M, Yamamoto M, Saeki T, et al. A novel clinical entity, IgG4-related disease (IgG4RD): general concept and details. Mod Rheumatol. 2012;22:1–14.

    Article  CAS  PubMed  Google Scholar 

  262. Wallace ZS, Zhang Y, Perugino CA, Naden R, Choi HK, Stone JH. Clinical phenotypes of IgG4-related disease: an analysis of two international cross-sectional cohorts. Ann Rheum Dis. 2019;78:406–12.

    Article  CAS  PubMed  Google Scholar 

  263. Wallace ZS, Naden RP, Chari S, Choi H, Della-Torre E, Dicaire J-F, et al. The 2019 American College of Rheumatology/European League Against Rheumatism Classification Criteria for IgG4-Related Disease. Arthritis Rheumatol (Hoboken, NJ). 2020;72:7–19.

    Article  Google Scholar 

  264. Groth KA, Hove H, Kyhl K, Folkestad L, Gaustadnes M, Vejlstrup N, et al. Prevalence, incidence, and age at diagnosis in Marfan Syndrome. Orphanet J Rare Dis [Internet]. 2015 [cited 2017 Nov 23];10. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4668669/

  265. Loeys BL, Dietz HC, Braverman AC, Callewaert BL, Backer JD, Devereux RB, et al. The revised Ghent nosology for the Marfan syndrome. J Med Genet. 2010;47:476–85.

    Article  CAS  PubMed  Google Scholar 

  266. Judge DP, Dietz HC. Marfan’s syndrome. Lancet. 2005;366:1965–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  267. Pinal-Fernandez I, Selva-O’Callaghan A, Grau JM. Diagnosis and classification of eosinophilic fasciitis. Autoimmun Rev. 2014;13:379–82.

    Article  CAS  PubMed  Google Scholar 

  268. Mazori DR, Femia AN, Vleugels RA. Eosinophilic fasciitis: an updated review on diagnosis and treatment. Curr Rheumatol Rep. 2017;19:74.

    Article  PubMed  Google Scholar 

  269. Gunnarsson R, Molberg Ø, Gilboe I-M, Gran JT, Group PS. The prevalence and incidence of mixed connective tissue disease: a national multicentre survey of Norwegian patients. Ann Rheum Dis. 2011;70:1047–51.

    Article  PubMed  Google Scholar 

  270. Tani C, Carli L, Vagnani S, Talarico R, Baldini C, Mosca M, et al. The diagnosis and classification of mixed connective tissue disease. J Autoimmun. 2014;48–49:46–9.

    Article  PubMed  CAS  Google Scholar 

  271. Amigues JM, Cantagrel A, Abbal M, Mazieres B. Comparative study of 4 diagnosis criteria sets for mixed connective tissue disease in patients with anti-RNP antibodies. Autoimmunity Group of the Hospitals of Toulouse. J Rheumatol. 1996;23:2055–62.

    CAS  PubMed  Google Scholar 

  272. Valter I, Saretok S, Maricq HR. Prevalence of scleroderma spectrum disorders in the general population of Estonia. Scand J Rheumatol. 1997;26:419–25.

    Article  CAS  PubMed  Google Scholar 

  273. Ferri C, Valentini G, Cozzi F, Sebastiani M, Michelassi C, La Montagna G, et al. Systemic sclerosis: demographic, clinical, and serologic features and survival in 1,012 Italian Patients. Medicine. 2002;81:139.

    Article  PubMed  Google Scholar 

  274. van den Hoogen F, Khanna D, Fransen J, Johnson SR, Baron M, Tyndall A, et al. 2013 classification criteria for systemic sclerosis: an American college of rheumatology/European league against rheumatism collaborative initiative. Ann Rheum Dis. 2013;72:1747–55.

    Article  PubMed  Google Scholar 

  275. Mayes MD, Lacey JV, Beebe-Dimmer J, Gillespie BW, Cooper B, Laing TJ, et al. Prevalence, incidence, survival, and disease characteristics of systemic sclerosis in a large US population. Arthritis Rheum. 2003;48:2246–55.

    Article  PubMed  Google Scholar 

  276. Martinez-Lopez A, Blasco-Morente G, Perez-Lopez I, Herrera-Garcia J, Luque-Valenzuela M, Sanchez-Cano D, et al. CLOVES syndrome: review of a PIK3CA-related overgrowth spectrum (PROS). Clin Genet. 2017;91:14–21.

    Article  CAS  PubMed  Google Scholar 

  277. Keppler-Noreuil KM, Rios JJ, Parker VER, Semple RK, Lindhurst MJ, Sapp JC, et al. PIK3CA-related overgrowth spectrum (PROS): diagnostic and testing eligibility criteria differential diagnosis and evaluation. Am J Med Genet A. 2015;1:287–95.

    Article  CAS  Google Scholar 

  278. Kurek KC, Luks VL, Ayturk UM, Alomari AI, Fishman SJ, Spencer SA, et al. Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome. Am J Hum Genet. 2012;90:1108–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Keppler-Noreuil KM, Sapp JC, Lindhurst MJ, Parker VE, Blumhorst C, Darling T, et al. Clinical delineation and natural history of the PIK3CA-related overgrowth spectrum. Am J Med Genet A. 2014;164:1713–33.

    Article  CAS  PubMed Central  Google Scholar 

  280. Kihiczak GG, Meine JG, Schwartz RA, Janniger CK. Klippel-Trenaunay syndrome: a multisystem disorder possibly resulting from a pathogenic gene for vascular and tissue overgrowth. Int J Dermatol. 2006;45:883–90.

    Article  PubMed  Google Scholar 

  281. Wang SK, Drucker NA, Gupta AK, Marshalleck FE, Dalsing MC. Diagnosis and management of the venous malformations of Klippel–Trénaunay syndrome. J Vasc Surg Venous Lymphat Disord. 2017;5:587–95.

    Article  PubMed  Google Scholar 

  282. Dimopoulos A, Sicko RJ, Kay DM, Rigler SL, Fan R, Romitti PA, et al. Copy number variants in a population-based investigation of Klippel–Trenaunay syndrome. Am J Med Genet. 2017;173:352–9.

    Article  CAS  PubMed  Google Scholar 

  283. Zhang T, Yao Y, Wang J, Li Y, He P, Pasupuleti V, et al. Haploinsufficiency of Klippel–Trenaunay syndrome gene Aggf1 inhibits developmental and pathological angiogenesis by inactivating PI3K and AKT and disrupts vascular integrity by activating VE-cadherin. Hum Mol Genet. 2016;25:5094–110.

    CAS  PubMed  PubMed Central  Google Scholar 

  284. Lindhurst MJ, Sapp JC, Teer JK, Johnston JJ, Finn EM, Peters K, et al. A mosaic activating mutation in AKT1 associated with the proteus syndrome. N Engl J Med. 2011;365:611–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  285. Cohen MM. Proteus syndrome review: molecular, clinical, and pathologic features. Clin Genet. 2014;85:111–9.

    Article  CAS  PubMed  Google Scholar 

  286. Turner JT, Cohen MM, Biesecker LG. Reassessment of the proteus syndrome literature: application of diagnostic criteria to published cases. Am J Med Genet. 2004;130A:111–22.

    Article  PubMed  Google Scholar 

  287. Cives M, Simone V, Rizzo FM, Dicuonzo F, Cristallo Lacalamita M, Ingravallo G, et al. Erdheim–Chester disease: a systematic review. Crit Rev Oncol Hematol. 2015;95:1–11.

    Article  PubMed  Google Scholar 

  288. Diamond EL, Dagna L, Hyman DM, Cavalli G, Janku F, Estrada-Veras J, et al. Consensus guidelines for the diagnosis and clinical management of Erdheim–Chester disease. Blood. 2014;124:483–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Cavalli G, Guglielmi B, Berti A, Campochiaro C, Sabbadini MG, Dagna L. The multifaceted clinical presentations and manifestations of Erdheim–Chester disease: comprehensive review of the literature and of 10 new cases. Ann Rheum Dis. 2013;72:1691–5.

    Article  PubMed  Google Scholar 

  290. Mazor RD, Manevich-Mazor M, Shoenfeld Y. Erdheim–Chester disease: a comprehensive review of the literature. Orphanet J Rare Dis. 2013;8:137.

    Article  PubMed  PubMed Central  Google Scholar 

  291. Hanks S, Adams S, Douglas J, Arbour L, Atherton DJ, Balci S, et al. Mutations in the gene encoding capillary morphogenesis protein 2 cause juvenile hyaline fibromatosis and infantile systemic hyalinosis. Am J Hum Genet. 2003;73:791–800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Haidar Z, Temanni R, Chouery E, Jitesh P, Liu W, Al-Ali R, et al. Diagnosis implications of the whole genome sequencing in a large Lebanese family with hyaline fibromatosis syndrome. BMC Genet [Internet]. 2017 [cited 2017 Nov 21];18. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5244738/

  293. Shieh JTC, Swidler P, Martignetti JA, Ramirez MCM, Balboni I, Kaplan J, et al. Systemic hyalinosis: a distinctive early childhood-onset disorder characterized by mutations in the anthrax toxin receptor 2 gene (ANTRX2). Pediatrics. 2006;118:e1485-92.

    Article  PubMed  Google Scholar 

  294. Lindvall LE, Kormeili T, Chen E, Ramirez MCM, Grum-Tokars V, Glucksman MJ, et al. Infantile systemic hyalinosis: case report and review of the literature. J Am Acad Dermatol. 2008;58:303–7.

    Article  PubMed  Google Scholar 

  295. Marzano AV, Ishak RS, Saibeni S, Crosti C, Meroni PL, Cugno M. Autoinflammatory skin disorders in inflammatory bowel diseases, pyoderma gangrenosum and sweet’s syndrome: a comprehensive review and disease classification criteria. Clinic Rev Allerg Immunol. 2013;45:202–10.

    Article  CAS  Google Scholar 

  296. Ghoufi L, Ortonne N, Ingen-Housz-Oro S, Barhoumi W, Begon E, Haioun C, et al. Histiocytoid Sweet Syndrome Is More Frequently Associated With Myelodysplastic Syndromes Than the Classical Neutrophilic Variant. Medicine (Baltimore) [Internet]. 2016 [cited 2017 Oct 18];95. Available from: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4839791/

  297. Cohen PR. Sweet’s syndrome—a comprehensive review of an acute febrile neutrophilic dermatosis. Orphanet J Rare Dis. 2007;2:34.

    Article  PubMed  PubMed Central  Google Scholar 

  298. Su WP, Liu HN. Diagnostic criteria for Sweet’s syndrome. Cutis. 1986;37:167–74.

    CAS  PubMed  Google Scholar 

  299. Walker DC, Cohen PR. Trimethoprim-sulfamethoxazole-associated acute febrile neutrophilic dermatosis: Case report and review of drug-induced Sweet’s syndrome. J Am Acad Dermatol. 1996;34:918–23.

    Article  CAS  PubMed  Google Scholar 

  300. von den Driesch P. Sweet’s syndrome (acute febrile neutrophilic dermatosis). J Am Acad Dermatol. 1994;31:535–56.

    Article  PubMed  Google Scholar 

  301. Horváth A, Páll N, Molnár K, Kováts T, Surján G, Vicsek T, et al. A nationwide study of the epidemiology of relapsing polychondritis. Clin Epidemiol. 2016;8:211–30.

    Article  PubMed  PubMed Central  Google Scholar 

  302. Dion J, Costedoat-Chalumeau N, Sène D, Cohen-Bittan J, Leroux G, Dion C, et al. Relapsing polychondritis can be characterized by three different clinical phenotypes: analysis of a recent series of 142 patients. Arthritis Rheumatol. 2016;68:2992–3001.

    Article  PubMed  Google Scholar 

  303. Mathew SD, Battafarano DF, Morris MJ. Relapsing polychondritis in the department of defense population and review of the literature. Semin Arthritis Rheum. 2012;42:70–83.

    Article  PubMed  Google Scholar 

  304. Michet CJ. Relapsing polychondritis: survival and predictive role of early disease manifestations. Ann Intern Med. 1986;104:74.

    Article  PubMed  Google Scholar 

  305. McAdam LP, O’Hanlan MA, Bluestone R, Pearson CM. Relapsing polychondritis: prospective study of 23 patients and a review of the literature. Medicine (Baltimore). 1976;55:193–215.

    Article  CAS  Google Scholar 

  306. Damiani JM, Levine HL. Relapsing polychondritis–report of ten cases. Laryngoscope. 1979;89:929–46.

    CAS  PubMed  Google Scholar 

  307. Durtette C, Hachulla E, Resche-Rigon M, Papo T, Zénone T, Lioger B, et al. Cogan syndrome: characteristics, outcome and treatment in a French nationwide retrospective study and literature review. Autoimmun Rev. 2017;16:1219–23.

    Article  PubMed  Google Scholar 

  308. Gluth MB, Baratz KH, Matteson EL, Driscoll CLW. Cogan syndrome: a retrospective review of 60 patients throughout a half century. Mayo Clinic Proc. 2006;81:483–8.

    Article  Google Scholar 

  309. Tayer-Shifman OE, Ilan O, Tovi H, Tal Y. Cogan’s syndrome—clinical guidelines and novel therapeutic approaches. Clinic Rev Allerg Immunol. 2014;47:65–72.

    Article  CAS  Google Scholar 

  310. Wang Y, Zhao J, Ji L lan, Zhang S, Zhang Z. Weber–Christian disease present with lung nodules dramatically improved with corticosteroid therapy: one case report and literature review. Int J Rheum Dis. 2015;n/a-n/a.

  311. Verrilli S, Ciarnella A, Laganà B, Calafiore S, Guglielmelli F, Basile L, et al. Ocular inflammation: can it be a sign of activity of Weber–Christian disease? A case report and review of literature. Ocul Immunol Inflamm. 2016;24:223–6.

    PubMed  Google Scholar 

  312. White JW, Winkelmann RK. Weber–Christian panniculitis: a review of 30 cases with this diagnosis. J Am Acad Dermatol. 1998;39:56–62.

    Article  PubMed  Google Scholar 

  313. Mavrikakis I, Georgiadis T, Fragiadaki K, Sfikakis PP. Orbital lobular panniculitis in Weber–Christian disease: sustained response to anti-TNF treatment and review of the literature. Surv Ophthalmol. 2010;55:584–9.

  314. Cohen SS, Skovbo S, Vestergaard H, Kristensen T, Møller M, Bindslev-Jensen C, et al. Epidemiology of systemic mastocytosis in Denmark. Br J Haematol. 2014;166:521–8.

    Article  PubMed  Google Scholar 

  315. Lim K-H, Tefferi A, Lasho TL, Finke C, Patnaik M, Butterfield JH, et al. Systemic mastocytosis in 342 consecutive adults: survival studies and prognostic factors. Blood. 2009;113:5727–36.

    Article  CAS  PubMed  Google Scholar 

  316. Pardanani A. Systemic mastocytosis in adults: 2017 update on diagnosis, risk stratification and management. Am J Hematol. 2016;91:1146–59.

    Article  CAS  PubMed  Google Scholar 

  317. Pieri L, Bonadonna P, Elena C, Papayannidis C, Grifoni FI, Rondoni M, et al. Clinical presentation and management practice of systemic mastocytosis. A survey on 460 Italian patients. Am J Hematol. 2016;91:692–9.

    Article  PubMed  Google Scholar 

  318. Valent P, Akin C, Metcalfe DD. Mastocytosis: 2016 updated WHO classification and novel emerging treatment concepts. Blood. 2017;129:1420–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Dubrey S, Shah S, Hardman T, Sharma R. Sarcoidosis: the links between epidemiology and aetiology. Postgrad Med J. 2014;90:582–9.

    Article  PubMed  Google Scholar 

  320. Nicholson TT, Plant BJ, Henry MT. Sarcoidosis in Ireland: regional differences in prevalence and mortality from 1996–2005. Sarcoidosis Vasc Diffuse Lung Dis. 2010;27:111–20.

    CAS  PubMed  Google Scholar 

  321. Wessendorf TE, Bonella F, Costabel U. Diagnosis of sarcoidosis. Clinic Rev Allerg Immunol. 2015;49:54–62.

    Article  Google Scholar 

Download references

Acknowledgements

San Francisco edit for editing and proofreading the manuscript.

Funding

Open Access funding enabled and organized by Projekt DEAL. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

MM, CS and MS conceptualized the work and substantively revised it. JL, TB, and CD acquired and analyzed the data. JL, DK and MS analyzed and interpreted the data, and wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Matthias F. Seidel.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Appendix

Appendix

See Table 1, 2.

Table 1 Results from the literature analyses
Table 2 Overview of rare rheumatic diseases

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leyens, J., Bender, T.T.A., Mücke, M. et al. The combined prevalence of classified rare rheumatic diseases is almost double that of ankylosing spondylitis. Orphanet J Rare Dis 16, 326 (2021). https://0-doi-org.brum.beds.ac.uk/10.1186/s13023-021-01945-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13023-021-01945-8

Keywords