Familial Mediterranean Fever: Assessing the Overall Clinical Impact and Formulating Treatment Plans

Raffaele Manna1,2 and Donato Rigante3,2..

1 Institute of Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
2 Periodic Fevers Research Centre, Università Cattolica Sacro Cuore, Rome, Italy.
3 Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.

Correspondence to: Raffaele Manna, MD, PhD, Institute of Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Largo A. Gemelli 8, 00168 Rome, Italy. Tel. +39 06 30159433. Fax. +39 06 35502775. E-mail: raffaele.manna@policlinicogemelli.it

Published: May 1, 2019
Received: January 14, 2019
Accepted: March 7, 2019
Mediterr J Hematol Infect Dis 2019, 11(1): e2019027 DOI 10.4084/MJHID.2019.027

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Recurrent self-limited attacks of fever and short-lived inflammation in the serosal membranes, joints, and skin are the leading features of familial Mediterranean fever (FMF), the most common autoinflammatory disorder in the world, transmitted as autosomal recessive trait caused by MEFV gene mutations. Their consequence is an abnormal function of pyrin, a natural repressor of inflammation, apoptosis, and release of cytokines. FMF-related mutant pyrins are hypophosphorylated following RhoA GTPases’ impaired activity and show a propensity to relapsing uncontrolled systemic inflammation with inappropriate response to inflammatory stimuli and leukocyte spread to serosal membranes, joints or skin. Typical FMF phenotype 1 consists of brief episodes of inflammation and serositis, synovitis, and/or erysipelas-like eruption, whereas phenotype 2 is defined by reactive amyloid-associated (AA) amyloidosis, which is the most ominous complication of FMF, in otherwise asymptomatic individuals.
Furthermore, FMF phenotype 3 is referred to the presence of two 
MEFV mutations with neither clinical signs of FMF nor AA amyloidosis. The influence of epigenetic and/or environmental factors can contribute to the variable penetrance and phenotypic heterogeneity of FMF. Colchicine, a tricyclic alkaloid with anti-microtubule and anti-inflammatory properties, is the bedrock of FMF management: daily administration of colchicine prevents the recurrence of FMF attacks and the development of secondary AA amyloidosis. Many recent studies have also shown that anti-interleukin-1 treatment is the best therapeutic option for FMF patients nonresponsive or intolerant to colchicine. This review aims to catch readers’ attention to the clinical diversity of phenotypes, differential diagnosis, and management of patients with FMF.



Introduction

Primary dysfunction of the innate immune system is the architrave of autoinflammatory disorders, in which there is no evidence of adaptive immunity involvement, neither high-titer autoantibodies nor antigen-specific T cells, and no infectious triggers.[1] People with hereditary autoinflammatory disorders display periodically-recurring clinical features consisting of fever and inflammation with symptom-free intervals of different duration between febrile attacks.[2] In this group of diseases, familial Mediterranean fever (FMF) occupies a primary seat, as it is the most common autoinflammatory disorder worldwide,[3] although it has been historically associated with people living around the Mediterranean basin as Turks, Arabs, Sephardic Jews, and Armenians.[4]

The Ancient Heredity of Familial Mediterranean Fever

In 1945 the allergologist Sheppard Siegal described as “benign paroxysmal peritonitis” his own disease, which was similar to the characteristics presented by other five Jewish patients: they all had cutaneous signs, recurrent peritonitis, and periodic fever attacks.[5] In 1954 Hobart Reimann was the first clinician who talked about “FMF,” defining its “periodic” outstanding features, noting that the adjectives cyclic, rhythmic, episodic, relapsing, recurrent, paroxysmal and intermittent had been used interchangeably to highlight the disease.[6] In 1955 professor Harry Heller and his colleagues described the overall FMF clinical picture in detail, studied its inheritance, and found FMF-associated nephropathy deriving from amyloidosis as an ominous long-term complication of FMF.[7] FMF was the first autoinflammatory disorder for which the causing gene was identified in 1997 by two independent groups, American and French.[8-10] The gene was called MEFV (from Mediterranean FeVer) and is located on the short arm of chromosome 16 (p13.3) between the genes linked to polycystic kidney disease and Rubinstein-Taybi syndrome, spanning approximately 14 Kb of genomic DNA and comprising 10 exons: it encodes a 781 amino acid-protein with a molecular weight of 86 kD which was called “pyrin” (meaning “fever” in Greek) by the American group and “marenostrin” (using the ancient Latin name “mare nostrum” for the Mediterranean sea) by the French one.[11] This discovery represented the starting point to understand the pathogenesis not only for FMF but also for other autoinflammatory disorders. During the last years, there have been lots of studies demonstrating a growing interest of the scientific community in the search for autoinflammatory mechanisms in many other inflammatory conditions.[12,13]

The Size of a Borderless Disease

Although genetic research into FMF began around 1997, we still lack a complete picture of its genetic variation, carrier frequency, and penetrance.[14] Ethnic distribution of peculiar MEFV variants was initially considered a feature of FMF, and the disease was mostly recognized in people living around the Mediterranean basin and deriving from the ancient Mesopotamia or Phoenician lands: Armenians, non-Ashkenazi Jews (above all Sephardic Jews, who emigrated to the Iberian peninsula, in fact, Sephardi means “Spain” in Hebrew), Turks, Arabs (mostly North-Africans such as Maghrebins) and Druze population (an ethnoreligious Arabic-speaking group originating in Western Asia). The clinical diagnosis of FMF among these populations is enhanced by the widely-recognized higher prevalence of the disease. In addition, it is unclear whether all MEFV variants are true disease-causing mutations, as only five founder mutations (V726A, M694V, M694I, M680I, and E148Q) have been related to 74% of typical FMF cases in Armenians, Arabs, Jews and Turks.[15] However, because of so many migrations during the past centuries, the gene causing FMF has also been spread to Western Europe as well as to the Americas, China, Japan, Australia and New Zealand.[16] The higher frequencies of MEFV variants in some populations could be explained by an increased resistance against specific pathogens, such as Yersinia pestis or intracellular bacteria such as Mycobacterium tuberculosis, or by raised protection from asthma.[17] In Italy, FMF is erroneously considered a rare disease (“rare” means that its prevalence is less than 1 per 2000 inhabitants): various historical reasons account for the presence of the FMF gene in Italy, as Greek colonization of Sicily and Southern Italian regions in the VIII century B.C., the arrival of the first Christians in Rome under the Roman Empire in the I-II century A.D., and the Arab conquest of Sicily in the IX century A.D.[18,19] The observation of the same mutations and haplotypes in populations that have been isolated for centuries with high rates of consanguinity indicates that most cases of FMF are descended from a very ancient pool of founders. 

Pyrin, an Intracellular Sentinel against Infections

Before MEFV discovery, it was commonly known that FMF attacks were characterized by a massive sterile flux of polymorphonuclear leukocytes into some body regions, which were serosal membranes, joints, and skin.[20,21] A huge number of studies dedicated to pyrin is revealing new molecular details about the general processes of cellular defense against infections, inflammation and apoptosis. Indeed, pyrin is largely expressed in the white blood cells and has a pivotal role in the activation of inflammasome and processing of the potent pyrogenic cytokine interleukin (IL)-1ß: in fact, it is involved in the activation of caspase-1 and release of active IL-1, as a structural part of the inflammasome complex.[22] In particular, the “pyrin domain” of this protein is a member of the death-domain-fold superfamily and is involved in the apoptotic pathway modulation through caspase recruitment and production of IL-1.[23] IL-1 transcriptional pathways are also misregulated in FMF attack-free periods, supporting the presence of subclinical inflammation between attacks, though different studies have not confirmed this hypothesis.[24,25] Pyrin activity is at the level of cytoskeletal assembly, and specific microtubule assembly inhibitors prevent pyrin-mediated caspase-1 activation and secretion of IL-1 in peripheral blood mononuclear cells of FMF patients.[26] Recent experimental studies have suggested that pyrin function is mediated by RhoA (Ras homolog gene family-member A) GTPases, enzymes targeted to the plasma membrane by the addition of a geranylgeranyl lipid tail: more specifically, FMF mutations in the pyrin B30.2 domain decrease the threshold of activation of the pyrin inflammasome, generally activated by various RhoA-inhibiting toxins produced by both Gram-negative and Gram-positive bacteria. In a normal condition, the phosphorylated pyrin binds to regulatory proteins that block the pyrin inflammasome, but inhibition of RhoA effector kinases decreases the phosphorylation of pyrin, which in turn activates pyrin inflammasome.[27] Triggers stimulating at a cellular level the periodic occurrence of acute clinical attacks of FMF are not known, even if different chronic infections such as Helicobacter pylori infection or small bowel bacterial overgrowth might tune patients’ inflammatory responses.[28,29] Other current studies suggest that non-MEFV genetic systems, epigenetic, and environmental modifiers might interplay with pyrin, influencing the clinical expression of FMF. Colchicine, a major neutral alkaloid from Colchicum species, suppresses pyrin inflammasome activity through direct activation of RhoA, disabling host cell cytoskeletal organization and causing an anti-chemotactic effect on the polymorphonuclear cells.[30]

A Host of Genotype Studies

FMF is inherited in a recessive manner, although recent studies have suggested that some heterozygotes manifest a spectrum of findings from classic FMF to mild FMF.[31] The most frequent MEFV mutations are contained in the exons 10 and 2 and are heterogeneously distributed in different populations.[32] Four mutations represent more than 70% of the mutated alleles in FMF cases of Mediterranean ancestry: M694V, M694I, M680I and V726A, all in the exon 10. Genotypes including two mutations located within mutational MEFV “hot-spots” (codons 680 or 694) in the exon 10 have been associated with the most severe phenotypes of FMF.[33] An unsolved issue is the possibility of genotype-phenotype correlations: for instance, M694V homozygosis, M680I homozygosis and heterozygosis for M608I/M694V genotypes are usually associated with severe clinical pictures and more frequent incidence of amyloidosis, while V726A is associated with milder disease and less frequent amyloidosis.[34] Understanding the correlation between FMF phenotype and genotype is further obscured by the existence of complex alleles, modifier loci, and potential epigenetic factors. In the United States FMF is frequently encountered in Ashkenazi Jews and immigrants from the Middle East and Armenia. In Germany, most FMF patients are of Turkish origin. In France, there is a relatively large FMF population that originated from North Africa. Among Armenians, the carrier rate for FMF is approximately 1:7 with a disease rate of roughly 1:500.[35] The E148Q is the least penetrant mutation of FMF, frequently found in the less severe pictures so that it has been considered a polymorphism, not a true disease-causing mutation.[36] The R202Q mutation is also considered a genetic polymorphism, present in about 15% of unaffected populations.[37] Some genes have been tested to assess their possible modifying effect on the clinical features of FMF: for instance, the alpha/alpha genotype of the serum amyloid-A gene (SAA1) is associated with increased risk of amyloidosis in FMF patients, especially if homozygous for the M694V mutation.[38] Moreover, Berkun et al. showed that FMF patients carrying mutations in the NOD2/CARD15 gene, which is involved in Blau syndrome, an autoinflammatory disorder starting in the first years of life with noncaseating epithelioid granulomas affecting joints, skin and eye, variably associated with heterogeneous systemic features,[39] are prone to a more severe FMF course and higher development of colchicine resistance.[40]

A Kaleidoscopic Disease with Protean Faces

FMF is characterized by short-lived and self-resolving attacks of fever, abdominal, thoracic or joint pain and systemic inflammation which recur over time combined with intercritical periods of apparent wellness:[41] both clinical features and disease severity may vary among different ethnic groups.[42] General triggers of FMF attacks may be emotional stress, strenuous physical activity, menses, nutritional changes, use of contraceptives, hypovitaminosis D, etc. Bouts of fever with painful symptoms related to the abdomen, chest or one or multiple joints, singularly or in various combinations, are the most frequently declared symptoms by people living throughout many areas of the Mediterranean basin. Fever is present in 96% of inflammatory episodes: body temperature can peak over 40°C; it usually appears suddenly and lasts from 6 to 72 hours, preceded by chills in about ¼ of cases. Two phenotypically independent pictures of FMF can be recognized: [a] acute short-lasting painful febrile attacks of peritonitis, pleuritis, or arthritis (phenotype 1), and [b] nephropathic amyloidosis (phenotype 2), which can lead to terminal renal failure even at a young age, and no other clinical symptom. There is a third picture (phenotype 3) characterized by two MEFV mutations with neither clinical signs of FMF nor of amyloid-associated (AA) amyloidosis.[43] Attacks do not have a regular periodism: their frequency varies from once per week to once per decade. Over the course of this lifelong illness, an affected individual will probably have several forms of febrile and painful episodes, but the recurrence of one type over many years is most frequent.[44] Onset is usually during the first decade in about 50% of cases, and symptoms might also start in the first months of life. As clinical manifestations of FMF appear quite early; they might be confused with a variety of diseases occurring in the pediatric age: for instance, recurrent febrile tonsillitis may be a symptom of FMF attacks, especially in early childhood,[45] entering in a challenging differential diagnosis with periodic fever, aphthous stomatitis, pharyngitis, and adenitis (PFAPA) syndrome, a typical recurrent disorder occurring in children less than 5 years, but also reported in adults.[46-49]
Abdominal pain, due to peritoneum inflammation resembling acute abdominal disease like appendicitis, cholecystitis, ureteral stones or pelvic inflammatory disease, is present in about 90% of patients and is frequently associated with small amounts of ascites upon ultrasound investigation. Peritoneum inflammation may also occur in the form of massive ascites with myofibroblast proliferation in the mesenteric region as an initial presentation of FMF.[50] Sometimes (in 30-40% of cases) patients might undergo unnecessary surgery.[51] Unilateral or bilateral pleural effusion can be demonstrated in 50-60% of patients and is characterized by quick resolution, while recurrent pericarditis can be observed in a small percentage of FMF patients (1-2.5%) during febrile attacks.[52,53] Intense scrotal pain simulating a testicular torsion is frequent in children with FMF, but it usually requires a conservative management.[54] Joint involvement is reported in 45% of cases and might present as transient arthralgia or oligoarthritis: short-lasting attacks begin without prodromes, involve large joints and suddenly disappear after 24-48 hours with no sequelae. Rarely arthritis may last for more than one week or even develop destructive features.[55] Some patients have painful, swollen and self-limited erythematous skin lesions on the legs: these lesions resemble a skin infection called erysipelas and are quite specific for FMF.[56] Another symptom associated with FMF is muscle pain, which might be severe and paralyzing: this muscular involvement is not prevented by colchicine and does not respond to nonsteroidal anti-inflammatory drugs, but only to corticosteroids. Muscular manifestations can present with severe febrile myalgia having three different patterns: spontaneous, effort-induced (i.e., exertional leg pain) and prolonged with an overall duration of 6 weeks.[57,58]
In recent times the clinical spectrum of FMF has expanded and many non-canonical manifestations have been reported.[59] For instance, neurologic signs other than constitutional headache have been described in some FMF patients, such as recurrent aseptic meningitis, demyelinating disorders, recurrent peripheral facial palsy and even stroke.[60]
Amyloidosis is the most dreadful complication of late-diagnosed, untreated or neglected FMF, which results from the deposition in different organs of a fatty-like substance, which is a cleavage product of serum amyloid-A, an acute-phase reactant produced by the liver. Main organs involved by amyloid deposition are kidney, gut, spleen, liver, heart and endocrine glands. Renal amyloidosis culminating in renal failure, which occurs in as many as 60% of untreated patients with FMF, is the major cause of death in FMF. Recent studies have focused on the polymorphism of the SAA1 gene as a genetic contributor to the development of amyloidogenesis, but found no influence by the major histocompatibility complex.[61] The pathogenic role of environmental factors in FMF-related amyloidosis such as the country of origin is suggested by the lower incidence of amyloidosis in Jews living outside the Mediterranean basin: for instance, Armenians living in Armenia have a much higher incidence of amyloidosis than Armenian Americans, even before the introduction of colchicine. In addition, amyloidosis appears to be less common among Iraqis, Ashkenazi Jews and Arabs.[62] The association between amyloidosis and the mutation M694V is widely reported,[63] but non-amyloid glomerulopathies such as IgM or IgA nephropathy, focal and diffuse proliferative glomerulonephritis, and rapidly progressive glomerulonephritis have also been observed. Also, some non-granulomatous vasculitides, such as Henoch-Schönlein purpura (in 2.6-5% of cases), polyarteritis nodosa (0.8-1%) and Behçet’s disease (0.5%) have been associated with FMF.[64-66] Over the years, an increased rate of MEFV carriers has also been found in complex multifactorial diseases such as ankylosing spondylitis and multiple sclerosis, implicating this gene and its pathways in the development of such disorders.[67] A few patients with FMF might also have attacks characterized by macrophage activation syndrome, in which well-differentiated mononuclear cells exhibiting hemophagocytic activity have swarmed different organs and systems, giving rise to a dramatic clinical picture consisting of persistent fever, multi-organ damage, cytopenia, hyperferritinemia, hypertriglyceridemia, and hypofibrinogenemia with high mortality rates.[68,69] Lastly, a retrospective study related to over 8.000 Jewish patients with FMF registered at the Tel Hashomer Hospital (with a mean age of 43.74 ± 14.7 years) revealed that there is a significantly lower incidence of cancer in FMF patients than in the general population of Israel, and which might be attributed to a direct physiologic effect of FMF or the antimitotic effects of colchicine administered on the long run.[70] Table 1 shows the main clinical features reported by 373 patients managed in the Periodic Fevers Research Center of our University.

Table 1 Table 1. List of the main clinical features of familial Mediterranean fever in the cohort of patients managed in our Centre.  

Searching for Universal Diagnostic Criteria of Familial Mediterranean Fever

Traditionally, diagnosis of FMF has been based on the typical clinical manifestations and physician's insight, though a purely clinical diagnosis is complicated if there are febrile episodes without serositis or if amyloidosis is initially demonstrated without a clear history of fever and serositis. There is no specific test for FMF diagnosis, and some diagnostic criteria have been suggested, all based on clinical data and supported by the patient’s ethnic origin or family history. During past years many authors have tried to develop different diagnostic flow-charts and several sets of criteria have been proposed since 1967, when Sohar et al. deduced that periodically-recurrent fevers and self-limited painful manifestations in various sites (abdomen, chest, joints, or skin), not explained by any other causative factor, were a mandatory FMF feature. They also recognized the importance of AA amyloidosis and the patient’s origin from the areas around the Mediterranean sea. The Tel Hashomer Hospital criteria by Sohar’s group (listed in Table 2) were promulgated starting from clinical observations in adult Israeli population, and are the most widely used for diagnosis of FMF in most clinical settings. If 2 major Tel Hashomer criteria or 1 major criterion and 2 minor ones are satisfied the diagnosis of FMF can be confirmed.[71]

Table 2 Table 2. Classification Criteria for the Clinical Diagnosis of Familial Mediterranean Fever (FMF) according to the Tel Hashomer criteria. Diagnosis is made when 2 major criteria or 1 major and 2 minor criteria are satisfied, while diagnosis is probable if 1 major and 1 minor criterion are present. 

In 1997 new criteria were validated by Livneh et al. to corroborate the clinical elements included in the Tel Hashomer criteria, but excluding other manifestations, like amyloidosis not explained by any other causative factor, less common at the onset of FMF. Two versions of these criteria, one more conservative and extensive than the other, were created, both with a sensitivity and specificity above 95%: the presence of at least 1 of 4 major criteria related to “typical” disease attacks or 2 of 5 minor criteria or 1 minor criterion plus 5 of 10 supportive criteria should suggest the diagnosis of FMF (see Table 3).[72]

Table 3 Table 3. Classification Criteria for the Clinical Diagnosis of Familial Mediterranean Fever (FMF) according to Livneh et al. Diagnosis of FMF requires 1 major criteria, or  2 minor criteria, or 1 minor criterion plus ≥5 supportive criteria (family history of FMF, appropriate ethnic origin, age less than 20 years at disease onset, severity of attacks requiring bed rest, spontaneous remission of symptoms, presence of symptom-free intervals, transient elevation of inflammatory markers, episodic proteinuria or hematuria, nonproductive laparotomy with removal of a “white” appendix, consanguinity of parents) or 1 minor criterion plus ≥ 4 of the “first” five supportive criteria. “Incomplete” attacks are defined as painful and recurrent flares that differ from typical attacks in 1 or 2 features, as follows: 1) normal temperature or lower than 38°C; 2) attacks longer than 1 week or shorter than 6 hours; 3) no signs of peritonitis recorded during acute abdominal complaint.

Diagnosis of FMF in children has been established using the same clinical criteria created for adults. However, a frequent delay in the appearance of a complete clinical picture in very young children, the presence of atypical signs, and absence of a suggestive family history may cause additional difficulty. Moreover, the evidence that some criteria were of poor relevance for children with FMF and differences of some clinical manifestations in younger children (who might have shorter attacks, infrequent chest pain, or even fever alone) prompted a Turkish group of clinicians in 2009 to formulate new FMF criteria for the pediatric population (the so-called Turkish FMF Pediatric criteria), listed in the Table 4.[73] The pediatric cohorts where these criteria were evaluated included only cases genetically ascertained to have two MEFV mutations, regardless of their phenotypes.

Table 4 Table 4. Classification Criteria for the Clinical Diagnosis of Familial Mediterranean Fever (FMF) according to Yalçinkaya and Ozen (Turkish FMF Pediatric Criteria). Diagnosis of FMF requires the presence of 2 out of 5 criteria (in Turkish children).

These Turkish criteria for the diagnosis of FMF yielded a better sensitivity in an international cohort of children from either European or Eastern Mediterranean regions, though their specificity was lower than in previous criteria.[74] Anyway, all used diagnostic criteria should need further improvements, such as validation on broader cohorts of probands, and the inclusion of genetic data. Federici et al. developed new classification criteria for the diagnosis of different autoinflammatory disorders through the identification of ‘positive’ and ‘negative’ criteria correlated with each disease, suggesting that the presence of aphthous stomatitis, urticaria-like rashes, cervical lymph node enlargement and febrile episodes lasting more than 6 days should exclude the diagnosis of FMF.[75]
There is no specific laboratory examination to support the diagnosis of FMF, except for MEFV analysis. During typical acute attacks, blood tests show a generalized increase of the inflammatory parameters (erythrosedimentation rate, C-reactive protein, serum amyloid-A, immunoglobulins) with a parallel neutrophil leukocytosis (until and over 20.000/mm3).[76]
The role of genetics in supporting the diagnosis of FMF is essential, but the genetic analysis should never substitute both clinical process and clinician’s judgment. Following MEFV cloning, the genetic test for FMF has become available in many countries, providing a new exciting tool for diagnostic confirmation. Genetic diagnosis is definite when two mutations, also nonidentical, are present in the two MEFV alleles; if only one or no pathogenic mutation is found the clinical diagnosis of FMF is still possible due to the potential occurrence of occult mutations. Of course, for patients with typical clinical pictures and appropriate ethnic origin, FMF diagnosis can be made without a genetic confirmation, which is vice versa contributory for cases with atypical presentation, the absence of family history, or unusual ethnic origin. In particular, genetic testing might reveal no known mutation in 30-40% of patients: in this case if clinical manifestations are convincingly those of FMF the patient is put on an open trial with colchicine for 3-6 months. If there is a positive response and symptoms reappear after stopping colchicine, it is assumed that the patient has FMF.[77]
Furthermore, diagnosis of FMF in very young heterozygous children should be made with caution, as FMF heterozygous children can present with an FMF-like disease in early childhood, which does not differ from that seen in patients carrying two mutated alleles, although it may disappear with age. Therefore, a careful follow-up of heterozygous children is of paramount importance before establishing a definite diagnosis.[78] For individuals with two FMF-related mutations who do not report symptoms, if there are risk factors for amyloidosis (such as the country, family history, and persistently elevated inflammatory markers, particularly serum amyloid-A), a close follow-up should be started and at least colchicine prophylaxis considered.

Ancient and Modern Treatments

Since 1972 colchicine is the anchor drug used in patients with FMF. Lifelong prophylaxis with colchicine is safe and effective in preventing attacks of FMF and also the well-known complication of amyloidosis:[79] a therapeutic trial with colchicine for at least three-six months is useful to ascertain whether there is a decrease in the severity or in the frequency of FMF flares in every patient suspected to have FMF.[80] Continuative colchicine prophylaxis, even if started during childhood, has no effects on the normal final expected growth of children. By contrast, colchicine is not effective if administered during acute flares of FMF. The minimal daily dose in adults is 1 mg/day, but in children there is no definitely suggested dose, and management should be driven by the occurrence of clinical symptoms and inflammation on laboratory tests. Other factors, such as the genotype or body surface area may help to manage colchicine dose in a more individualized fashion.[81] The initial dose in children is usually 0.5-1 mg/day regardless of age and body weight, with little increases until disease control is achieved. The rate of adverse events for colchicine, mainly gastrointestinal effects, is low. Neutropenia, hair loss and allergies are infrequent side effects. A colchicine-related myopathy combined with the mild-to-severe elevation of creatine kinase may appear two weeks to several months after initiation of treatment. Moreover, since in vitro high doses of colchicine stop mitosis and colchicine displays its effect by fixating the intracellular microtubules, arresting their polymerization and finally disrupting mitosis and motility systems within the cells, in theory, this drug could interfere with patients’ fertility, though many studies have proved no clearly adverse effects on gametogenesis.[82] However, if untreated, FMF itself can lead to amyloid deposits in the testis and ovary, resulting in infertility: therefore, patients with FMF may safely continue to use colchicine throughout the reproductive phase of their life.[83]
Complete remission of FMF under colchicine is achieved in 65% of patients and partial remission in 30%, while around 5% of patients remain nonresponsive.[84] True resistance to colchicine is a rare event, mostly observed in patients displaying peculiar MEFV genotypes and occurring despite the regular use of colchicine at the maximal doses. Low adherence to colchicine administration may be a key component of resistance and should be assessed.[85] Since long-term colchicine prophylaxis may be complicated by gastrointestinal side effects, by our experience, we usually recommend lactose-free diet and treatment of intestinal bacterial overgrowth to improve colchicine tolerance.[86] There are also FMF patients with low disease activity who might become utterly free of attacks for a long time and even stop colchicine prophylaxis: long-term remission of FMF, characterized by a time interval of at least 3 years without FMF clinical manifestations and off-colchicine, after a period of FMF activity, is rather infrequent. The prevalence of disease remission in the FMF population is estimated at 3.3%, based on a study by Ben-Zvi et al. from the largest center for diagnosis and treatment of FMF in Israel.[87] These patients seem to have distinct clinical, demographic and molecular characteristics, allocating them to the mildest end of the disease severity spectrum of FMF. This phenotype is comparable to that of patients with late-onset FMF.[88]
FMF protracted arthritis, mostly affecting hip or knees, has shown results following treatment with tumor necrosis factor inhibitors.[89] Characterization and early identification of FMF patients with uncontrolled inflammatory activity have become more important after the availability of new treatment options for the prevention of disease-associated complications and permanent damages of FMF. After the demonstration of inflammasome dysregulation as the dominating pathogenetic mechanism in different autoinflammatory disorders including FMF, IL-1β has been shown as the most intriguing target to attack.[90,91] An increasing experience of IL-1 blockade has been matured over recent times and, after showing its dramatic efficacy in cryopyrin-associated periodic syndrome, a rare and totally IL-1-mediated hereditary autoinflammatory disorder,[92] several case reports and case series dedicated to FMF have documented both efficacy and safety of anti-IL-1 agents, such as anakinra, canakinumab and rilonacept in patients inadequately responding to colchicine.[93-96] All IL-1 inhibitors are effective for controlling attacks and inflammatory activity in patients with refractory FMF and even in those complicated with AA amyloidosis, reducing or stabilizing the amount of proteinuria and preserving renal function in short-term follow-up studies. Structured scores rating FMF activity or severity by the use of attack parameters (site, duration and frequency) have been used to classify and settle disease diversity, while response to treatment has been evaluated by patients’/parents'/physicians' global assessment of disease severity, laboratory parameters performed every 6 months, and different scores related to symptoms and organ damages.[97-102]

How to Put Patients in Differential Diagnosis

Diagnosis of FMF is mainly a clinical process based on the fever recurrence combined with different symptoms: the final phenotype is not determined by the MEFV genotype alone, but a combination with other modifier genes and environmental factors. In addition, many diseases share recurrent fever as a common presenting feature. The problem remains relevant at whatever age, but mostly in children who frequently may show recurrent infections requiring assessment and even hospitalization.[103] A frequent and misdiagnosed cause of recurrent fevers in the pediatric age is PFAPA syndrome, but a detailed history-taking combined with a mindful collection of physical findings during flares is crucial to identify FMF. It is important to be aware that colchicine prophylaxis has been adopted by different research groups on PFAPA patients, although complete responses have not been observed.[104] The discrimination between PFAPA syndrome during childhood, early onset-Behçet’s disease, and FMF may be challenging: in particular, Behçet’s disease involves primarily the oral and genital mucosa, but also skin and eyes.[105] An Israelian large-scale population-based study has shown that FMF is more frequently diagnosed in female patients with Behçet’s disease of Arab descent.[106] Quite similar to FMF is mevalonate kinase deficiency/hyperimmunoglobulin D syndrome, though the median age at the onset of this disease is 0.5 years, and these children have concurrent clinical symptoms involving the gastrointestinal, mucocutaneous and musculoskeletal system lasting more than three days:[107] increased urinary levels of mevalonate during febrile flares can consent to identify patients with mevalonate kinase deficiency.[108] Another autoinflammatory disorder to differentiate is tumor necrosis factor receptor-associated periodic syndrome: its presenting features include a high-grade fever of much longer duration (in comparison with FMF), abdominal pain, arthralgia, myalgia caused by monocytic fasciitis, conjunctivitis and periorbital edema, though the genetic diagnosis remains discriminating.[109] The quite characteristic recurrence of abdominal pain in FMF patients should also require to consider alternative diagnosis, such as inflammatory bowel disease, biliary and renal lithiasis, cholecystitis, pancreatitis, hemolytic syndromes, Behçet’s disease and acute hepatic porphyrias, which are rare inborn errors of heme biosynthesis characterized by acute neurovisceral attacks heralded by severe abdominal pain. Porphyria is commonly diagnosed during acute attacks by the demonstration of a striking urinary elevation of the neurotoxic porphyrin precursors aminolevulinic acid and porphobilinogen, that correlate with severity of abdominal symptoms.[110

General Conclusions

In our modern era, FMF cannot be considered a rare condition confined to the Eastern Mediterranean countries as in the more recent past: indeed, this disease is scattered throughout the world showing a dynamic face with genetic and environmental aspects which are diversely tangled to determine its clinical phenotype.[111] Colchicine prophylaxis allows all age-patients with FMF to live a normal life with no restriction or substantial risk of sequelae. However, the undiagnosed disease has a negative influence on patients’ and their families’ quality of life because of frequent hospitalizations, potential surgical interventions caused by a wrong interpretation of FMF clinical signs or simply due to the recurrence of febrile attacks, which reduce school or work attendance and limit any social attitudes for patients.[112] Diagnosis of FMF is mainly made on the basis of the typical clinical findings in association with the peculiar ethnicity, family history, and response to colchicine.[113,114] Despite the great steps in our understanding of FMF, we still have a number of hanging questions: for instance, what is the exact role of additional genes in the definition of the final FMF phenotype, what is the pathophysiology of the disease in patients with only one MEFV mutation or in those without any MEFV mutation, which are further unexplored inflammatory pathways which might be involved in the disease expression or progression to amyloidosis, and so on. The progress in the knowledge of genetic determinants of FMF could constitute a significant step towards the understanding of the human genome power and general mechanisms of inflammation with future relevant therapeutic implications. Wider awareness of FMF will probably reduce the diagnostic delay in recognition of the disease and positively affect the quality of life of patients who will have a lower risk of long-term morbidity and complications.

References   

  1. Stojanov S, Kastner DL. Familial autoinflammatory diseases: genetics, pathogenesis and treatment. Curr Opin Rheumatol 2005;17:586-99. https://doi.org/10.1097/bor.0000174210.78449.6b  PMid:16093838
  2. Ozen S, Demir S. Monogenic periodic fever syndromes: treatment options for the pediatric patient. Paediatr Drugs 2017;19:303-1. https://doi.org/10.1007/s40272-017-0232-6
  3. Rigante D. The broad-ranging panorama of systemic autoinflammatory disorders with specific focus on acute painful symptoms and hematologic manifestations in children. Mediterr J Hematol Infect Dis 2018;10 https://doi.org/10.4084/mjhid.2018.067
  4. Rigante D. A developing portrait of hereditary periodic fevers in childhood. Expert Opin Orphan Drugs 2018;6:47-55. https://doi.org/10.1080/21678707.2018.1406797
  5. Siegal S. Benign paroxysmal peritonitis. Gastroenterology 1949;12:234-47. PMid:18124924
  6. Reimann HA, Moadie J, Semerdjian S, Sahyoun PF. Periodic peritonitis; heredity and pathology: report of seventy-two cases. J Am Med Assoc 1954;154:1254-9. https://doi.org/10.1001/jama.1954.02940490018005  PMid:13151833
  7. Heller H, Sohar E, Sherf L. Familial Mediterranean fever. AMA Arch Int Med 1958;102:50-71. https://doi.org/10.1001/archinte.1958.00260190052007  PMid:13558745
  8. Babior BM, Matzner Y. The familial Mediterranean fever gene - cloned at last. N Engl J Med 1997;337:1548-9. https://doi.org/10.1056/NEJM199711203372112  PMid:9366590
  9. French FMF Consortium. A candidate gene for familial Mediterranean fever. Nat Genet 2017;17:25-31. https://doi.org/10.1038/ng0997-25  PMid:9288094
  10. 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. https://doi.org/10.1016/S0092-8674(00)80539-5  PMid:9288758
  11. Booth DR, Gillmore JD, Booth SE, et al. Pyrin/marenostrin mutations in familial Mediterranean fever. QJM 1998;91:603-6. https://doi.org/10.1093/qjmed/91.9.603  PMid:10024914
  12. Rigante D. The fresco of autoinflammatory diseases from the pediatric perspective. Autoimmun Rev 2012;11:348-56. https://doi.org/10.1016/j.autrev.2011.10.008  PMid:22024500
  13. Rigante D. The protean visage of systemic autoinflammatory syndromes: a challenge for inter-professional collaboration. Eur Rev Med Pharmacol Sci 2010;14:1-18. PMid:20184084
  14. Fujikura K. Global epidemiology of familial Mediterranean fever mutations using population exome sequences. Mol Genet Genomic Med 2015;3:272-82. https://doi.org/10.1002/mgg3.140 PMid: PMC4521964
  15. Touitou I. The spectrum of familial Mediterranean fever mutations. Eur J Hum Genet 2001;9:473-83. https://doi.org/10.1038/sj.ejhg.5200658n  PMid:11464238
  16. Gershoni-Baruch R, Shinawi M, Leah K, et al. Familial Mediterranean fever: prevalence, penetrance and genetic drift. Eur J Hum Genet 2001;9:634-7. https://doi.org/10.1038/sj.ejhg.5200672  PMid:11528510
  17. Lidar M, Livneh A. Familial Mediterranean fever: clinical, molecular and management advancements. Neth J Med 2007;65:318-24. PMid:17954950
  18. Manna R, Cerquaglia C, Curigliano V, et al. Clinical features of familial Mediterranean fever: an Italian overview. Eur Rev Med Pharmacol Sci 2009;13 Suppl 1:51-3. PMid:19530512
  19. Rigante D, Frediani B, Galeazzi M, et al. From the Mediterranean to the sea of Japan: the transcontinental odyssey of autoinflammatory diseases. Biomed Res Int 2013;2013:485103. https://doi.org/10.1155/2013/485103  PMid:23971037
  20. Balci-Peynircioglu B, Akkaya-Ulum YZ, Avci E, et al. Potential role of pyrin, the protein mutated in familial Mediterranean fever, during inflammatory cell migration. Clin Exp Rheumatol 2018;36 (6 Suppl 115):116-24. PMid:30582517
  21. Tunca M, Akar S, Onen F, et al. Familial Mediterranean fever in Turkey: results of a nationwide multicenter study. Medicine 2005 84:1-11. https://doi.org/10.1097/01.md.0000152370.84628.0c  PMid:15643295
  22. Lucherini OM, Rigante D, Sota J, et al. Updated overview of molecular pathways involved in the most common monogenic autoinflammatory diseases. Clin Exp Rheumatol 2018;36 Suppl 110(1):3-9. PMid: 29742053
  23. Alghamdi M. Familial Mediterranean fever, review of the literature. Clin Rheumatol 2017;36:1707-13. https://doi.org/10.1007/s10067-017-3715-5  PMid: 28624931
  24. Bagci S, Toy B, Tuzun A, et al. Continuity of cytokine activation in patients with familial Mediterranean fever. Clin Rheumatol 2004;23:333-7. https://doi.org/10.1007/s10067-004-0925-4   PMid:15293095
  25. Kelesoglu FM, Aygun E, Okumus NK, et al. Evaluation of subclinical inflammation in familial Mediterranean fever patients: relations with mutation types and attack status: a retrospective study. Clin Rheumatol 2016;35:2757-63. https://doi.org/10.1007/s10067-016-3275-0 PMid:27106545
  26. Van Gorp H, Saavedra PH, de Vasconcelos NM, et al. Familial Mediterranean fever mutations lift the obligatory requirement for microtubules in pyrin inflammasome activation. Proc Natl Acad Sci USA 2016;113:14384-9. https://doi.org/10.1073/pnas.1613156113 PMid:27911804
  27. Park YH, Wood G, Kastner DL, et al. Pyrin inflammasome activation and RhoA signaling in the autoinflammatory diseases FMF and HIDS. Nat Immunol 2016;17:914-21. https://doi.org/10.1038/ni.3457 PMid:27270401
  28. Demirtürk L, Ozel AM, Cekem K, et al. Co-existence of Helicobacter pylori infection in patients with familial Mediterranean fever (FMF) and the effect of Helicobacter pylori on the frequency and severity of FMF attacks. Dig Liver Dis 2005;37:153-8. https://doi.org/10.1016/j.dld.2004.09.027 PMid:15888278
  29. Verrecchia E, Sicignano LL, La Regina M, et al. Small intestinal bacterial overgrowth affects the responsiveness to colchicine in familial Mediterranean fever. Mediators Inflamm 2017;2017:7461426. https://doi.org/10.1155/2017/7461426 PMid:29379228
  30. Rigante D, La Torraca I, Avallone L, et al. The pharmacological basis of treatment with colchicine in children with familial Mediterranean fever. Eur Rev Med Pharmacol Sci 2006;10:173-8. PMid:16910346
  31. Rigante D. A systematic approach to autoinflammatory syndromes: a spelling booklet for the beginner. Expert Rev Clin Immunol 2017;13:571-97. https://doi.org/10.1080/1744666X.2017.1280396  PMid:28064547
  32. Dodé C, Pecheux C, Cazeneuve C, et al. Mutations in the MEFV gene in a large series of patients with a clinical diagnosis of familial Mediterranean fever. Am J Med Genet 2000;92:241-6. PMid: 10842288 https://doi.org/10.1002/(SICI)1096-8628(20000605)92:4<241::AID-AJMG3>3.0.CO;2-G
  33. Shinar Y, Livneh A, Langevitz P, et al. Genotype-phenotype assessment of common genotypes among patients with familial Mediterranean fever. J Rheumatol 2000; 27:1703-7. PMid:10914855
  34. Pasa S, Altintas A, Devecioglu B, et al. Familial Mediterranean fever gene mutations in the Southeastern region of Turkey and their phenotypical features. Amyloid 2008;15:49-53. https://doi.org/10.1080/13506120701815456  PMid:18266121
  35. Ben-Chetrit E, Touitou I. Familial Mediterranean fever in the world. Arthritis Rheum 2009;61:1447-53. https://doi.org/10.1002/art.24458 PMid:19790133
  36. Marek-Yagel D, Bar-Joseph I, Pras E, et al. Is E148Q a benign polymorphism or a disease-causing mutation? J Rheumatol 2009;36:2372.https://doi.org/10.3899/jrheum.090250 PMid:19820229
  37. Comak E, Akman S, Koyun M, et al. Clinical evaluation of R202Q alteration of MEFV genes in Turkish children. Clin Rheumatol 2014;33:1765-71. https://doi.org/10.1007/s10067-014-2602-6 PMid:24718488
  38. Gershoni-Baruch R, Brik R, Zacks N, et al. The contribution of genotypes at the MEFV and SAA1 loci to amyloidosis and disease severity in patients with familial Mediterranean fever. Arthritis Rheum 2003;48:1149-55. https://doi.org/10.1002/art.10944 PMid:12687559
  39. Caso F, Costa L, Rigante D, et al. Caveats and truths in genetic, clinical, autoimmune and autoinflammatory issues in Blau syndrome and early onset sarcoidosis. Autoimmun Rev 2014;13:1220-9. https://doi.org/10.1016/j.autrev.2014.08.010 PMid:25182201
  40. Berkun Y, Karban A, Padeh S, et al. NOD2/CARD15 gene mutations in patients with familial Mediterranean fever. Semin Arthritis Rheum 2012;42:84-8. https://doi.org/10.1016/j.semarthrit.2011.12.002 PMid:22244368
  41. Rigante D. New mosaic tiles in childhood hereditary autoinflammatory disorders. Immunol Lett 2018;193:67-76. https://doi.org/10.1016/j.imlet.2017.11.013 PMid:29198619
  42. Rigante D, La Torraca I, Ansuini V, et al. The multi-face expression of familial Mediterranean fever in the child. Eur Rev Med Pharmacol Sci 200;10:163-71. PMid:16910345
  43. Soriano A, Manna R. Familial Mediterranean fever: new phenotypes. Autoimmun Rev 2012;12:31-7. https://doi.org/10.1016/j.autrev.2012.07.019 PMid:22878273
  44. El-Shanti H, Majeed HA, El-Khateeb M. Familial mediterranean fever in Arabs. Lancet 2006 Mar 25;367(9515):1016-24. https://doi.org/10.1016/S0140-6736(06)68430-4 PMid:16564365
  45. Adrovic A, Sahin S, Barut K, et al. Familial Mediterranean fever and periodic fever, aphthous stomatitis, pharyngitis, and adenitis (PFAPA) syndrome: shared features and main differences. Rheumatol Int 2019;39:29-36. https://doi.org/10.1007/s00296-018-4105-2 PMid:30019226
  46. Gentileschi S, Vitale A, Frediani B, et al. Challenges and new horizons in the periodic fever, aphthous stomatitis, pharingitis and adenitis (PFAPA) syndrome. Expert Opin Orphan Drugs 2017;5:165-71. https://doi.org/10.1080/21678707.2017.1279049
  47. Gaggiano C, Rigante D, Sota J, et al. Treatment options for periodic fever, aphthous stomatitis, pharyngitis, and cervical adenitis (PFAPA) syndrome in children and adults: a narrative review. Clin Rheumatol 2019; 38: 11-17. https://doi.org/10.1007/s10067-018-4361-2 PMid:30488366
  48. Cantarini L, Vitale A, Bartolomei B, et al. Diagnosis of PFAPA syndrome applied to a cohort of 17 adults with unexplained recurrent fevers. Clin Exp Rheumatol 2012;30:269-71. PMid:22325152
  49. Rigante D, Vitale A, Natale MF, et al. A comprehensive comparison between pediatric and adult patients with periodic fever, aphthous stomatitis, pharyngitis, and cervical adenopathy (PFAPA) syndrome. Clin Rheumatol 2017;36:463-8. https://doi.org/10.1007/s10067-016-3317-7 PMid:27251674
  50. Cakir M, Ozgenc F, Baran M, et al. A rare cause of refractory ascites in a child: familial Mediterranean fever. Rheumatol Int 2010;30:531-4. https://doi.org/10.1007/s00296-009-0957-9 PMid:19466424
  51. Reissman P, Durst Al, Rivkind A, et al. Elective laparoscopic appendectomy in patients with familial Mediterranean fever. World J Surg 1994;18:139-42. https://doi.org/10.1007/BF00348205 PMid:8197770 PMid:8197770
  52. Kees S, Langevitz P, Zemer D, et al. Attacks of pericarditis as a manifestation of familial Mediterranean fever. QJM 1997;90:643-7. PMid: 9415347 https://doi.org/10.1093/qjmed/90.10.643   PMid:9415347
  53. Rigante D, Cantarini L, Imazio M, et al. Autoinflammatory diseases and cardiovascular manifestations. Ann Med 2011;43:341-6. https://doi.org/10.3109/07853890.2010.547212  PMid:21284530
  54. Eshel G, Vinograd I, Barr J, et al. Acute scrotal pain complicating familial Mediterranean fever in children. Br J Surg 1994; 81: 894-6. https://doi.org/10.1002/bjs.1800810633  PMid:8044614
  55. Jarjour RA, Dodaki R. Arthritis patterns in familial Mediterranean fever patients and association with M694V mutation. Mol Biol Rep 2011;38:2033-6. https://doi.org/10.1007/s11033-010-0326-5  PMid:20845072
  56. Rigante D, Cantarini L. Monogenic autoinflammatory syndromes at a dermatological level. Arch Dermatol Res 2011;303:375-80. https://doi.org/10.1007/s00403-011-1134-z  PMid:21340744
  57. Kotevoglu N, Sahin F, Ozkiris SO, et al. Protracted febrile myalgia of familial Mediterranean fever. Clin Exp Rheumatol 2004;22:S69-S70. PMid:15515790
  58. Tufan G, Demir S. Uncommon clinical pattern of FMF: protracted febrile myalgia syndrome. Rheumatol Int 2010;30:1089-90. https://doi.org/10.1007/s00296-009-1024-2  PMid:19590876
  59. Rigante D, Lopalco G, Tarantino G, et al. Non-canonical manifestations of familial Mediterranean fever: a changing paradigm. Clin Rheumatol 2015;34:1503-11. https://doi.org/10.1007/s10067-015-2916-z PMid:25761640
  60. Feld O, Yahalom G, Livneh A. Neurologic and other systemic manifestations in FMF: published and own experience. Best Pract Res Clin Rheumatol 2012;26:119-33. https://doi.org/10.1016/j.berh.2012.01.004 PMid:22424198
  61. Medlej-Hashim M, Delague V, Chouery E, et al. Amyloidosis in familial Mediterranean fever patients: correlation with MEFV genotype and SAA1 and MICA polymorphisms effects. BMC Med Genet 2004;5:4. https://doi.org/10.1186/1471-2350-5-4  PMid:15018633 PMCid:PMC356915
  62. Touitou I, Sarkisian T, Medlej-Hashim M, et al. Country as the primary risk factor for renal amyloidosis in familial Mediterranean fever. Arthritis Rheum 2007;56:1706-12. https://doi.org/10.1002/art.22507  PMid:17469185
  63. Rigante D, Frediani B, Cantarini L. A comprehensive overview of the hereditary periodic fever syndromes. Clin Rev Allergy Immunol 2018;54:446-53. https://doi.org/10.1007/s12016-016-8537-8 PMid:27068928
  64. Lange-Sperandio B, Möhring K, Gutzler F, et al. Variable expression of vasculitis in siblings with familial Mediterranean fever. Pediatr Nephrol 2004;19:539-43. https://doi.org/10.1007/s00467-004-1440-1 PMid:15015067
  65. Alghamdi M. Autoinflammatory disease-associated vasculitis/vasculopathy. Curr Rheumatol Rep 2018;20:87. doi: 10.1007/s11926-018-0788-3 https://doi.org/10.1007/s11926-018-0788-3  PMid:30446874
  66. Yazici A, Cefle A, Savli H. The frequency of MEFV gene mutations in Behçet's disease and their relation with clinical findings. Rheumatol Int 2012;32:3025-30. https://doi.org/10.1007/s00296-011-2011-y  PMid:21901355
  67. Soriano A, Pras E. Familial Mediterranean fever: genetic update. Isr Med Assoc J 2014;16:274-6. PMid: 24979829
  68. Stabile A, Bertoni B, Ansuini V, et al. The clinical spectrum and treatment options of macrophage activation syndrome in the pediatric age. Eur Rev Med Pharmacol Sci 2006;10:53-9. PMid:16705949
  69. Rigante D, Emmi G, Fastiggi M, et al. Macrophage activation syndrome in the course of monogenic autoinflammatory disorders. Clin Rheumatol 2015;34:1333-9. https://doi.org/10.1007/s10067-015-2923-0 PMid:25846831
  70. Brenner R, Ben-Zvi I, Shinar Y, et al. Familial Mediterranean fever and incidence of cancer: an analysis of 8,534 Israeli patients with 258,803 person-years. Arthritis Rheumatol 2018;70:127-33. https://doi.org/10.1002/art.40344  PMid:28992365
  71. Sohar E, Gafni J, Pras M, et al. Familial Mediterranean fever. A survey of 470 cases and review of the literature. Am J Med 1967;43:227-53. https://doi.org/10.1016/0002-9343(67)90167-2  PMid:5340644
  72. Livneh A, Langevitz P, Zemer D, et al. Criteria for the diagnosis of familial Mediterranean fever. Arthritis Rheum 1997;40:1879-85. https://doi.org/10.1002/1529-0131(199710)40  PMid:9336425
  73. Yalçinkaya F, Ozen S, Ozçakar ZB, et al. A new set of criteria for the diagnosis of familial Mediterranean fever in childhood. Rheumatology (Oxford) 2009;48:395-8. https://doi.org/10.1093/rheumatology/ken509 PMid:19193696
  74. Demirkaya E, Saglam C, Turker T, et al. Performance of different diagnostic criteria for Familial Mediterranean fever in children with periodic fevers: results from a multicenter international registry. J Rheumatol 2016;43:154-60. https://doi.org/10.3899/jrheum.141249 PMid:26568587
  75. Federici S, Sormani MP, Ozen S, et al. Evidence-based provisional clinical classification criteria for autoinflammatory periodic fevers. Ann Rheum Dis 2015;74:799-805. https://doi.org/10.1136/annrheumdis-2014-206580  PMid:25637003
  76. Yepiskoposyan L, Harutyunyan A. Population genetics of familial Mediterranean fever: a review. Eur J Hum Genet 2007; 15: 911-6. https://doi.org/10.1038/sj.ejhg.5201869  PMid:17568393
  77. Caso F, Cantarini L, Lucherini OM, et al. Working the endless puzzle of hereditary autoinflammatory disorders. Mod Rheumatol 2014;24:381-9. https://doi.org/10.3109/14397595.2013.843755  PMid:24251993
  78. Hentgen V, Grateau G, Stankovic-Stojanovic K, et al. Familial Mediterranean fever in heterozygotes: are we able to accurately diagnose the disease in very young children? Arthritis Rheum 2013;65:1654-62. https://doi.org/10.1002/art.37935  PMid:23508419
  79. Zemer D, Pras M, Sohar E, et al. Colchicine in the prevention and treatment of the amyloidosis of familial Mediterranean fever. N Engl J Med 1986;314:1001-5. https://doi.org/10.1056/NEJM198604173141601  PMid:3515182
  80. Rigante D, Vitale A, Lucherini OM, et al. The hereditary autoinflammatory disorders uncovered. Autoimmun Rev 2014;13:892-900. https://doi.org/10.1016/j.autrev.2014.08.001  PMid:25149390
  81. Knieper AM, Klotsche J, Lainka E, et al. Familial Mediterranean fever in children and adolescents: factors for colchicine dosage and predicting parameters for dose increase. Rheumatology (Oxford) 2017;56:1597-606. https://doi.org/10.1093/rheumatology/kex222  PMid:2885939
  82. Yanmaz MN, Özcan AJ, Savan K. The impact of familial Mediterranean fever on reproductive system. Clin Rheumatol 2014;33:1385-8. https://doi.org/10.1007/s10067-014-2709-9  PMid:24924605
  83. Ozturk MA, Kanbay M, Kasapoglu B, et al. Therapeutic approach to familial Mediterranean fever: a review update. Clin Exp Rheumatol 2011;29(4 Suppl 67):S77-86. PMid:21968242
  84. Eroglu FK, Beşbaş N, Topaloglu R, et al. Treatment of colchicine-resistant Familial Mediterranean fever in children and adolescents. Rheumatol Int 2015;35:1733-7. https://doi.org/10.1007/s00296-015-3293-2  PMid:26001859
  85. Corsia A, Georgin-Lavialle S, Hentgen V, et al. A survey of resistance to colchicine treatment for French patients with familial Mediterranean fever. Orphanet J Rare Dis 2017;12:54. https://doi.org/10.1186/s13023-017-0609-1  PMid:28302131
  86. Cerquaglia C, Diaco M, Nucera G, et al. Pharmacological and clinical basis of treatment of familial Mediterranean fever (FMF) with colchicine or analogues: an update. Curr Drug Targets Inflamm Allergy 2005;4:117-24. https://doi.org/10.2174/1568010053622984 PMid:15720245  PMid:15720245
  87. Ben-Zvi I, Krichely-Vachdi T, Feld O, et al. Colchicine-free remission in familial Mediterranean fever: featuring a unique subset of the disease-a case control study. Orphanet J Rare Dis 2014;9:3. doi: 10.1186/1750-1172-9-3 https://doi.org/10.1186/1750-1172-9-3  PMid:24401676
  88. Tamir N, Langevitz P, Zemer D, et al. Late-onset familial Mediterranean fever (FMF): a subset with distinct clinical, demographic, and molecular genetic characteristics. Am J Med Genet 1999;87:30-5. PMid: 10528243 https://doi.org/10.1002/(SICI)1096-8628(19991105)87:1<30::AID-AJMG6>3.0.CO;2-B
  89. Rigante D, Manna R. A position for tumor necrosis factor inhibitors in the management of colchicine-resistant familial Mediterranean fever? Immunol Lett 2016;180:77-8. https://doi.org/10.1016/j.imlet.2016.10.007 PMid:27984066
  90. Lopalco G, Cantarini L, Vitale A, et al. Interleukin-1 as a common denominator from autoinflammatory to autoimmune disorders: premises, perils, and perspectives. Mediators Inflamm 2015;2015:194864. https://doi.org/10.1155/2015/194864  PMid:25784780
  91. Cantarini L, Lopalco G, Cattalini M, et al. Interleukin-1: Ariadne's thread in autoinflammatory and autoimmune disorders. Isr Med Assoc J 2015;17:93-7. PMid:26223084
  92. Cantarini L, Lucherini OM, Frediani B, et al. Bridging the gap between the clinician and the patient with cryopyrin-associated periodic syndromes. Int J Immunopathol Pharmacol 2011;24:827-36. https://doi.org/10.1177/039463201102400402  PMid:22230390
  93. Varan Ö, Kucuk H, Babaoglu H, et al. Efficacy and safety of interleukin-1 inhibitors in familial Mediterranean fever patients complicated with amyloidosis. Mod Rheumatol 2018 Apr 27:1-4. https://doi.org/10.1080/14397595.2018.1457469  PMid:29578360
  94. De Benedetti F, Gattorno M, Anton J, et al. Canakinumab for the treatment of autoinflammatory recurrent fever syndromes. N Engl J Med 2018;378:1908-19. https://doi.org/10.1056/NEJMoa1706314  PMid:29768139
  95. Vitale A, Insalaco A, Sfriso P, et al. A snapshot on the on-label and off-label use of the interleukin-1 inhibitors in Italy among rheumatologists and pediatric rheumatologists: a nationwide multi-center retrospective observational study. Front Pharmacol 2016;7:380. https://doi.org/10.3389/fphar.2016.00380  PMid:27822185
  96. Hashkes PJ, Spalding SJ, Hajj-Ali R, et al. The effect of rilonacept versus placebo on health-related quality of life in patients with poorly controlled familial Mediterranean fever. Biomed Res Int 2014;2014:854842. https://doi.org/10.1155/2014/854842,  PMid:25147819
  97. Mor A, Shinar Y, Zaks N, et al. Evaluation of disease severity in familial Mediterranean fever. Semin Arthritis Rheum 2005;35:57-64. https://doi.org/10.1016/j.semarthrit.2005.02.002  PMid:16084225
  98. Cantarini L, Lucherini OM, Iacoponi F, et al. Development and preliminary validation of a diagnostic score for identifying patients affected with adult-onset autoinflammatory disorders. Int J Immunopathol Pharmacol 2010;23:1133-4. https://doi.org/10.1177/039463201002300417  PMid:21244762
  99. Cantarini L, Iacoponi F, Lucherini OM, et al. Validation of a diagnostic score for the diagnosis of autoinflammatory diseases in adults. Int J Immunopathol Pharmacol 2011;24:695-702. https://doi.org/10.1177/039463201102400315  PMid:21978701
  100. Piram M, Koné Paut I, Lachmann H, et al. Validation of the auto-inflammatory diseases activity index (AIDAI) for hereditary recurrent fever syndromes. Ann Rheum Dis 2014;73:2168-73. https://doi.org/10.1136/annrheumdis-2013-203666  PMid:24026675
  101. Ter Haar NM, Annink KV, Al-Mayouf SM, et al. Development of the autoinflammatory disease damage index (ADDI). Ann Rheum Dis 2017;76:821-30. https://doi.org/10.1136/annrheumdis-2016-210092  PMid:27811147
  102. Ter Haar NM, van Delft ALJ, Annink KV, et al. In silico validation of the Autoinflammatory Disease Damage Index. Ann Rheum Dis 2018;77:1599-1605. https://doi.org/10.1136/annrheumdis-2018-213725  PMid:30077992
  103. Rigante D. Autoinflammatory syndromes behind the scenes of recurrent fevers in children. Med Sci Monit 2009;15:RA179-87. PMid:19644432
  104. Dusser P, Hentgen V, Neven B, et al. Is colchicine an effective treatment in periodic fever, aphthous stomatitis, pharyngitis, cervical adenitis (PFAPA) syndrome? Joint Bone Spine 2016;83:406-11. https://doi.org/10.1016/j.jbspin.2015.08.017  PMid:27068612
  105. Caso F, Costa L, Rigante D, et al. Biological treatments in Behçet's disease: beyond anti-TNF-therapy. Mediators Inflamm 2014;2014:107421. https://doi.org/10.1155/2014/107421  PMid:25061259
  106. Watad A, Tiosano S, Yahav D, et al. Behçet's disease‬ and familial Mediterranean fever: Two sides of the same coin or just an association? A cross-sectional study‬. Eur J Intern Med 2017;39:75-8. https://doi.org/10.1016/j.ejim.2016.10.011  PMid:27776949
  107. Ter Haar NM, Jeyaratnam J, Lachmann HJ, et al. The phenotype and genotype of mevalonate kinase deficiency: a series of 114 cases from the Eurofever Registry. Arthritis Rheumatol 2016;68:2795-805. https://doi.org/10.1002/art.39763 PMid:27213830
  108. Esposito S, Ascolese B, Senatore L, et al. Current advances in the understanding and treatment of mevalonate kinase deficiency. Int J Immunopathol Pharmacol 2014;27:491-8. https://doi.org/10.1177/039463201402700404 PMid:25572728
  109. Rigante D, Lopalco G, Vitale A, et al. Key facts and hot spots on tumor necrosis factor receptor-associated periodic syndrome. Clin Rheumatol 2014;33:1197-207. https://doi.org/10.1007/s10067-014-2722-z PMid:24935411
  110. Naik H, Stoecker M, Sanderson SC, et al. Experiences and concerns of patients with recurrent attacks of acute hepatic porphyria: A qualitative study. Mol Genet Metab 2016;119:278-83. https://doi.org/10.1016/j.ymgme.2016.08.006  PMid:27595545
  111. Cantarini L, Rigante D, Brizi MG, et al. Clinical and biochemical landmarks in systemic autoinflammatory diseases. Ann Med 2012;44:664-73. https://doi.org/10.3109/07853890.2011.598546  PMid:21972825
  112. Cantarini L, Vitale A, Lucherini OM, et al. Childhood versus adulthood-onset autoinflammatory disorders: myths and truths intertwined. Reumatismo 2013;65:55-62. https://doi.org/10.4081/reumatismo.2013.55  PMid:23877409
  113. Rigante D, Vitale A, Natale MF, et al. Lights and shadows in autoinflammatory syndromes from the childhood and adulthood perspective. Clin Rheumatol 2016;35:565-72. https://doi.org/10.1007/s10067-015-3132-6 PMid: 6631101
  114. Vitale A, Rigante D, Lucherini OM, et al. The diagnostic evaluation of patients with a suspected hereditary periodic fever syndrome: experience from a referral center in Italy. Intern Emerg Med 2017;12:605-11. https://doi.org/10.1007/s11739-017-1622-z PMid:28194697

[TOP]