Saturday, October 11, 2025

Autoinflammatory Diseases in Adults: Recognition, Diagnosis, and Management

 

Autoinflammatory Diseases in Adults: Recognition, Diagnosis, and Management in Critical Care

Dr Neeraj Manikath, Claude.ai


ABSTRACT

Autoinflammatory diseases represent a spectrum of disorders characterized by dysregulation of innate immunity leading to unprovoked, recurrent inflammatory episodes. Historically considered rare pediatric conditions, autoinflammatory diseases increasingly present to critical care units in adults, often masquerading as sepsis, malignancy, or refractory inflammatory conditions. This review provides an evidence-based framework for recognizing, diagnosing, and managing autoinflammatory diseases in the adult critical care setting, with emphasis on clinical suspicion, diagnostic algorithms, and contemporary therapeutic options. We highlight key diagnostic pearls and management pearls that can reduce diagnostic delays and improve patient outcomes.

Keywords: autoinflammatory diseases, innate immunity, periodic fever, inflammasome, IL-1β, critical care


INTRODUCTION

Autoinflammatory diseases have emerged as important differential diagnoses for adult patients presenting with recurrent fever, systemic inflammation, and multi-organ dysfunction. Unlike autoimmune conditions characterized by adaptive immune dysfunction (antibodies and T-cell abnormalities), autoinflammatory diseases result from dysregulation of innate immune pathways, particularly those involving pattern recognition receptors and the inflammasome complex. The distinction is clinically important, as it dictates management strategies and prognostic considerations.

The incidence of autoinflammatory diseases in adults has been significantly underestimated. Historically perceived as rare pediatric entities, advancing genetic technologies and increased clinician awareness have uncovered a substantial adult population with these conditions. Patients frequently undergo extensive diagnostic workups for infectious diseases, malignancy, and other inflammatory conditions before autoinflammatory disease is suspected. This diagnostic odyssey delays appropriate treatment and exposes patients to unnecessary antibiotics, immunosuppression, and invasive procedures.

Critical care physicians must develop heightened clinical suspicion for autoinflammatory diseases, particularly when traditional diagnostic algorithms fail to identify a clear etiology for fever and systemic inflammation. This review synthesizes current evidence and clinical experience to provide a practical approach to recognition, diagnosis, and management of these complex conditions in the critical care setting.


CLASSIFICATION AND PATHOPHYSIOLOGY

Historical Context and Evolution of Classification

Autoinflammatory diseases were formally recognized as a distinct disease category by Kastner and colleagues in 1997, who first identified mutations in the CIAS1 gene in familial cold autoinflammatory syndrome (FCAS) and Muckle-Wells syndrome (MWS). Subsequent advances in genetic sequencing have identified over 40 genes associated with autoinflammatory phenotypes, expanding the recognized spectrum considerably.

The classification of autoinflammatory diseases has evolved from purely genetic definitions to include clinical phenotypes. The International League of Associations for Rheumatology (ILAR) published classification criteria in 2019, recognizing monogenic autoinflammatory diseases (MAD), complex autoinflammatory diseases (CAD), and probable autoinflammatory diseases (PAD). This classification framework provides utility for both research and clinical practice.

Mechanistic Classification

Inflammasome-mediated diseases represent the largest category, characterized by dysregulation of the NLRP3, NLRC4, or AIM2 inflammasomes. These multiprotein complexes, upon activation by damage-associated molecular patterns (DAMPs) or pathogen-associated molecular patterns (PAMPs), catalyze conversion of pro-IL-1β to active IL-1β. Examples include cryopyrin-associated periodic syndromes (CAPS), familial Mediterranean fever (FMF), and mevalonate kinase deficiency (MVD).

Non-inflammasome autoinflammatory diseases involve dysregulation of alternative innate pathways, including TNF receptor signaling (TNFRSF1A in TNFR-associated periodic syndrome, TRAPS), NF-κB signaling (OTULIN deficiency, NEMO deficiency), and others.

Autoinflammatory-like phenotypes occur in patients with genetic alterations in genes not formally classified as autoinflammatory but that exhibit autoinflammatory manifestations, such as CANDLE (PSMB8 mutations) and ALADIN deficiency.


CLINICAL PRESENTATION AND RECOGNITION: THE PEARLS

Pearl 1: Recurrent Fever with Symptom-Free Intervals

The most distinctive feature of autoinflammatory diseases is the periodicity of inflammatory episodes. Unlike infectious fever, autoinflammatory fever occurs in predictable cycles with complete resolution between episodes. A history of fever recurring every 3-7 days (CAPS), every 3-7 weeks (FMF), or triggered by cold exposure (FCAS) should immediately raise suspicion.

Clinical Pearl: Query patients specifically about the temporal pattern of symptoms: "Do your fevers come and go on a schedule?" A positive affirmative answer with specific periodicity is highly suggestive of autoinflammatory disease.

Pearl 2: Lack of Response to Antibiotics Despite Fever

A patient presenting with fever and elevated inflammatory markers (CRP, procalcitonin, ESR) who does not respond to broad-spectrum antibiotics is a red flag. Many patients with undiagnosed autoinflammatory diseases undergo multiple courses of antibiotics without clinical improvement. The absence of microbiological culture positivity despite clinical findings consistent with infection should prompt reconsideration of diagnosis.

Clinical Pearl: Document whether the fever responds predictably to NSAIDs or corticosteroids. Autoinflammatory fevers often respond to these agents during acute flares. A dramatic response to colchicine (as in FMF) or IL-1 inhibitors further supports the diagnosis.

Pearl 3: Multi-System Involvement with Characteristic Patterns

Autoinflammatory diseases display characteristic patterns of organ involvement:

  • Cutaneous manifestations: Recurrent urticarial or pustular lesions not responding to antihistamines or corticosteroids (as in pustular psoriasis-associated autoinflammatory disease)
  • Musculoskeletal involvement: Arthralgia/arthritis typically affecting large joints (knees, ankles, wrists)
  • Abdominal pain: Often severe, mimicking acute abdominal pathology, but without objective surgical findings
  • Serosal inflammation: Recurrent pleuritis, pericarditis, or peritonitis without infectious or malignant etiology
  • Ophthalmologic involvement: Particularly important in CAPS (chronic urticaria-like lesions) and Behçet's disease-associated autoinflammation
  • Amyloidosis: Secondary (AA) amyloidosis from chronic IL-1β-driven inflammation is a serious long-term complication

Clinical Pearl: The constellation of fever + arthralgia + abdominal pain + rash appearing in episodic fashion is classic for FMF. The presence of recurrent symptoms in multiple organ systems not explained by a single pathologic process suggests autoinflammatory etiology.

Pearl 4: Family History Patterns

While many autoinflammatory diseases follow autosomal dominant inheritance (CAPS, TRAPS), FMF follows autosomal recessive inheritance, particularly common in Mediterranean, Arab, and Ashkenazi Jewish populations. Patients from these ethnic backgrounds with recurrent fever deserve enhanced clinical suspicion.

Clinical Pearl: Specifically ask about consanguinity in the family history and ethnic ancestry. A family history of recurrent fever, unexplained deaths, or familial amyloidosis warrants genetic testing.

Pearl 5: Absence of Typical Autoimmune Features

Unlike autoimmune diseases characterized by high-titer autoantibodies and autoreactive T cells, autoinflammatory diseases typically show:

  • Negative or low-titer ANA
  • Negative rheumatoid factor
  • Normal complement levels (C3, C4)
  • Absence of anti-neutrophil cytoplasmic antibodies
  • Normal immunoglobulin levels

Clinical Pearl: A patient with fever and systemic inflammation but negative autoimmune workup should raise suspicion for autoinflammatory disease rather than leading to diagnostic dismissal.


MAJOR AUTOINFLAMMATORY DISEASES IN ADULTS

Cryopyrin-Associated Periodic Syndromes (CAPS)

Pathophysiology: CAPS encompasses a spectrum of NLRP3 inflammasome-mediated diseases characterized by IL-1β overproduction triggered by cold exposure or spontaneously. The NLRP3 inflammasome requires two signals for activation: priming (Signal 1, typically TLR activation) and activation (Signal 2, typically via K+ efflux or cathepsin B release from lysosomes). Mutations in CIAS1 (now recognized as NLRP3) result in constitutive inflammasome activation.

Clinical Spectrum:

  • FCAS: Episodes of fever, urticaria, and arthralgia triggered by cold exposure, typically lasting 12-24 hours. Conjunctivitis is pathognomonic. Symptoms typically begin in infancy.
  • MWS: Chronic urticaria-like rash, fever, and arthralgia with auditory involvement (sensorineural hearing loss) and progressive kidney disease (amyloidosis). Onset typically in childhood but may present for the first time in adulthood.
  • CINCA/NOMID: Most severe form with neonatal onset, chronic meningitis (with characteristic cerebral vasculitis), progressive arthropathy, and growth failure. Adult presentations are rare but may represent late-onset disease.

Critical Care Pearl: Adult presentations of CAPS are often initially misdiagnosed as acute meningitis or recurrent infections. The clue is the chronic meningitis that appears and disappears without microbiological confirmation. CSF analysis typically shows lymphocytic pleocytosis without positive cultures. Brain MRI may reveal enhancing cerebral lesions.

Diagnosis:

  • Genetic testing for NLRP3 mutations (heterozygous dominant)
  • Elevated serum IL-1β, IL-6, TNF-α, and CRP during flares
  • Skin biopsy showing neutrophilic infiltration without vasculitis

Management: IL-1 inhibitors are first-line therapy:

  • Anakinra: IL-1 receptor antagonist, dosing 100 mg SC daily
  • Canakinumab: Monoclonal anti-IL-1β antibody, subcutaneous or intravenous
  • Rilonacept: IL-1 trap (decoy receptor), subcutaneous

Corticosteroids provide temporary relief but are not disease-modifying. NSAIDs may provide symptomatic benefit during acute flares.

Oyster (Complications): Hearing loss in MWS can be profound and irreversible if not treated early. Corneal inflammation with limbal opacification can lead to visual loss. Amyloidosis with renal involvement represents the long-term threat to survival.


Familial Mediterranean Fever (FMF)

Epidemiology and Pathophysiology: FMF is the most common monogenic autoinflammatory disease, with prevalence of 1:250 to 1:1000 in Mediterranean populations. Over 80 mutations in the MEFV gene (encoding pyrin protein) have been identified. Pyrin is a member of the inflammasome and plays a crucial role in suppressing IL-1β activation in response to bacterial infection. Loss-of-function mutations paradoxically result in excessive inflammasome activation, likely through loss of pyrin's inhibitory effects.

Clinical Presentation: Classic FMF presents with recurrent, self-limited episodes of fever and serositis:

  • Fever: Typically 38-40°C, lasting 24-72 hours, often beginning in evening
  • Peritonitis: Severe abdominal pain, typically unilateral RLQ (90% of cases), mimicking acute appendicitis. Abdomen is often tender without peritoneal signs.
  • Pleuritis: Unilateral chest pain exacerbated by respiration and movement
  • Arthritis: Most commonly knees and ankles; acute, severe, self-limited
  • Rash: Erythematous plaques appearing on lower extremities and feet, resolving with episode

Diagnostic Challenge in Adults: FMF typically begins in childhood (peak onset 5-15 years), but adult presentations occur, particularly in:

  • Heterozygous carriers with mild disease
  • Patients from endemic populations who emigrated or were undiagnosed in childhood
  • Late-onset presentations associated with specific genotypes (e.g., M694I heterozygotes)

Critical Care Pearl: Adult FMF patients presenting to ICU typically have two scenarios:

  1. Acute flare mimicking surgical abdomen: Severe peritonitis leading to unnecessary surgical consultation. The key distinguishing feature is that peritoneal signs are typically mild relative to pain severity, and serial imaging shows no bowel pathology.
  2. Amyloid-related complications: Progressive amyloidosis with renal insufficiency, carpal tunnel syndrome (bilateral, often symmetrical), or bowel involvement. Patients may present with acute kidney injury, nephrotic syndrome, or obstruction.

Diagnosis:

  • Genetic testing: Homozygous or compound heterozygous mutations in MEFV confirm diagnosis
  • Diagnostic scoring systems: Tel Hashomer criteria or EUROFEVER criteria can support diagnosis in suspected cases pending genetic confirmation
  • Serum markers: IL-1β, CRP, serum amyloid A (SAA) elevated during flares but normalize between episodes
  • Genetic-phenotype correlation: Specific mutations associated with more severe disease (M694I homozygotes have earlier onset, more severe, and higher amyloidosis risk)

Management:

Acute flares: NSAIDs and short-term corticosteroids provide symptomatic relief

Prophylaxis and disease modification:

  • Colchicine: First-line, mechanism involves inhibition of inflammasome priming and preventing neutrophil migration. Dosing: 0.5-1.5 mg daily in divided doses. Dramatically reduces flare frequency and prevents amyloidosis. Monitoring required for bone marrow suppression and myopathy with chronic use.

  • IL-1 inhibitors: Reserved for colchicine-refractory or colchicine-intolerant patients:

    • Anakinra
    • Canakinumab
    • Rilonacept
  • TNF inhibitors: Etanercept has shown benefit in some colchicine-resistant patients

Oyster (Risk Stratification): Patients with M694I homozygous genotype have 5-25 fold increased amyloidosis risk and warrant more aggressive treatment. Regular monitoring with serum creatinine, urinalysis, and urine albumin-to-creatinine ratio is essential.

Pearl: Early diagnosis and appropriate colchicine therapy can completely prevent amyloidosis. Delayed diagnosis and inadequate treatment are major risk factors for end-stage renal disease.


TNF Receptor-Associated Periodic Syndrome (TRAPS)

Pathophysiology: TRAPS results from mutations in TNFRSF1A (TNF receptor superfamily member 1A), inherited in autosomal dominant fashion. Approximately 50% of patients have a positive family history. The mechanism involves impaired TNF receptor shedding and trafficking, leading to abnormal signaling and prolonged inflammation.

Clinical Presentation:

  • Fever episodes: Typically last 1-4 weeks (much longer than other periodic fevers), often beginning with chills
  • Conjunctivitis: Non-suppurative, often with severe eye pain
  • Rash: Migratory erythematous plaques often appearing on trunk and extremities
  • Arthralgias/arthritis: Similar to FMF
  • Serositis: Pleuritis and peritonitis similar to FMF but less common
  • Myalgias: Severe muscle pain, particularly affecting neck, shoulders, and lower extremities

Critical Care Pearl: The prolonged duration of fever (1-4 weeks) is the key distinguishing feature from other periodic fever syndromes. The presence of migratory rash is more characteristic of TRAPS than FMF. Patients with TRAPS often have more constitutional symptoms (weight loss, fatigue) between flares.

Diagnosis:

  • Genetic testing for TNFRSF1A mutations
  • Elevated IL-6 and CRP during flares, but IL-1β levels typically normal (important distinction from inflammasome-mediated disease)
  • Circulating TNF receptor levels abnormal
  • Clinical presentation with fever duration > 1 week is highly suggestive

Management:

Acute flares:

  • NSAIDs and corticosteroids provide some benefit but less dramatically than in FMF
  • Colchicine is generally ineffective

Prophylaxis:

  • TNF inhibitors: First-line therapy (etanercept, infliximab, adalimumab)
    • Etanercept is particularly effective, possibly because it acts as a soluble TNF receptor
    • Approximately 75% of patients show significant benefit
  • IL-1 inhibitors: Second-line option for TNF inhibitor-refractory cases
  • NSAIDs: Prophylactic use may reduce flare frequency

Oyster: Amyloidosis occurs in up to 5% of untreated TRAPS patients, less frequently than in FMF. Patients may develop secondary reactive amyloidosis and renal involvement. Long-term follow-up is essential.


Mevalonate Kinase Deficiency (MKD)

Pathophysiology: MKD (also known as hyper-IgD syndrome when presenting with elevated IgD levels) results from mutations in the MVK gene, inherited in autosomal recessive fashion. Mevalonate kinase is essential in the mevalonic acid pathway involved in cholesterol and isoprenoid synthesis. The mechanism linking MVK deficiency to autoinflammation remains incompletely understood but involves dysregulation of the NLRP3 inflammasome.

Clinical Presentation:

  • Fever episodes: Typically brief (3-7 days), often beginning abruptly with chills
  • Lymphadenopathy: Prominent, particularly cervical lymph nodes
  • Abdominal symptoms: Diarrhea and abdominal pain
  • Rash: Maculopapular or nodular, non-purpuric
  • Hepatosplenomegaly: Common
  • Arthralgia: Less prominent than in FMF or TRAPS
  • Aphthous ulcers: Oral ulcers may occur

Age of Onset: Typically before age 5, but adults with late-onset presentations have been reported. Heterozygous carriers may present with milder symptoms.

Critical Care Pearl: The presence of elevated IgD and IgA levels (found in ~80% of patients with MKD) helps distinguish this condition from other periodic fevers. The combination of fever + lymphadenopathy + elevated transaminases + diarrhea during flares is characteristic.

Diagnosis:

  • Genetic testing for MVK mutations (demonstrates biallelic mutations in classical MKD/hyper-IgD syndrome)
  • Elevated serum IgD (>100 IU/mL) and IgA in ~80% of patients
  • Elevated mevalonic acid in urine during flares
  • Elevated IL-1β during flares

Management:

Acute flares:

  • NSAIDs and corticosteroids provide symptomatic relief
  • Colchicine ineffective

Prophylaxis:

  • IL-1 inhibitors: First-line therapy
    • Anakinra most studied in MKD
    • Canakinumab also effective
  • TNF inhibitors: Some benefit reported but less effective than in TRAPS
  • Etanercept + anakinra: Combination therapy may be required for refractory cases

Oyster: While amyloidosis risk is lower than FMF, it can occur with chronic disease. Patients may develop inflammatory complications including uveitis.


Deficiency of IL-1 Receptor Antagonist (DIRA)

Pathophysiology: DIRA results from loss-of-function mutations in IL1RN (encoding IL-1 receptor antagonist), inherited in autosomal recessive fashion. IL-1RA is the natural inhibitor of both IL-1α and IL-1β. Absence of IL-1RA leads to unchecked IL-1 signaling.

Clinical Presentation:

  • Neonatal onset (typically within first weeks of life) with severe systemic inflammation
  • Pustular lesions
  • Osteolytic bone lesions (particularly frontal and occipital)
  • Periostitis and pseudo-osteomyelitis
  • Hepatosplenomegaly and failure to thrive
  • Elevated inflammatory markers from birth

Adult Presentations: Rare in adults; most patients diagnosed in infancy die without treatment or are identified through family screening.

Critical Care Pearl: While DIRA typically presents in neonates, awareness is important for critical care physicians who may encounter:

  1. Infected patients: Family members of diagnosed patients
  2. Refractory inflammatory conditions: Adults with severe autoinflammatory phenotype may have DIRA on genetic testing

Management: IL-1RA replacement (anakinra) is dramatically effective and life-saving.


NLRC4-Associated Autoinflammation

Pathophysiology: Gain-of-function mutations in NLRC4 lead to constitutive inflammasome activation. NLRC4 recognizes intracellular bacterial flagellin and other pathogen-associated molecules, activating caspase-1 and IL-1β production.

Clinical Presentation:

  • Recurrent fever episodes (often shorter than TRAPS but longer than CAPS)
  • Neutrophilic skin lesions
  • Gastrointestinal symptoms
  • Arthritis
  • Lymphadenopathy

Diagnosis: Genetic testing for NLRC4 mutations, elevated IL-1β

Management: IL-1 inhibitors (anakinra, canakinumab) are first-line and typically highly effective


Chronic Granulomatous Disease (CGD) – Autoinflammatory Manifestations

Pathophysiology: While primarily considered an immunodeficiency, CGD has significant autoinflammatory features due to dysregulated IL-1β production. CGD results from defects in NADPH oxidase, leading to inability to generate respiratory burst. The resulting accumulation of inflammatory mediators and impaired apoptosis of neutrophils leads to autoinflammatory manifestations.

Clinical Presentation:

  • Immunodeficiency: Recurrent infections with catalase-positive organisms
  • Autoinflammatory features: Discoid lupus-like rash, granulomatous dermatitis, perianal infections, inflammatory bowel disease-like manifestations
  • Fever: Episodic fever
  • Lymphadenopathy and hepatosplenomegaly: Often granulomatous

Critical Care Pearl: CGD presents a diagnostic and therapeutic challenge because treatment requires both antimicrobial coverage (for immune deficiency) and IL-1 modulation (for autoinflammatory features). IFN-γ is standard therapy for immunodeficiency; IL-1 inhibitors address autoinflammatory manifestations.


DIAGNOSTIC APPROACH

Diagnostic Algorithm

Step 1: Clinical Suspicion Identify red flags: recurrent fever, lack of infectious source, periodicity, negative autoimmune workup, multi-system involvement

Step 2: Temporal Pattern Analysis

  • Establish frequency and duration of episodes
  • Identify triggers (cold exposure, stress, fatigue, NSAIDs, menses)
  • Determine symptom-free intervals

Step 3: Phenotypic Characterization

  • Document constitutional symptoms (fever characteristics, fatigue, weight loss)
  • Detailed rash description and distribution
  • Organ-specific involvement (eyes, lungs, GI tract, joints, serosal membranes)
  • Family history and ethnic ancestry

Step 4: Laboratory Assessment

Acute phase markers:

  • CRP, ESR, procalcitonin during flares vs. between flares
  • Temporal relationship between symptoms and inflammatory markers

Inflammatory cytokines:

  • IL-1β (elevated in inflammasome-mediated disease)
  • IL-6, TNF-α
  • Timing of samples critical – should correlate with symptom onset

Additional workup:

  • Complete blood count with differential (look for leukocytosis, neutrophilia, anemia)
  • Comprehensive metabolic panel (renal function, liver function)
  • Immunoglobulin levels (elevated IgD/IgA in MKD)
  • Serum amyloid A and serum creatinine (screen for amyloidosis)

Negative findings to document:

  • Negative blood cultures (multiple sets if fever present)
  • Negative autoimmune markers (ANA, RF, anti-CCP, ANCA)
  • Normal complement levels
  • Negative or low-titer specific antibodies

Step 5: Tissue Diagnosis

  • Skin biopsy of acute lesions: typically shows neutrophilic infiltration without vasculitis (distinguishes from vasculitis)
  • Joint fluid analysis: sterile, inflammatory
  • CSF analysis (if CNS involvement): typically lymphocytic pleocytosis, negative cultures

Step 6: Genetic Testing

  • Targeted testing based on clinical phenotype
  • Multi-gene panel testing if diagnosis remains unclear
  • Whole genome or exome sequencing for atypical presentations

Pearl: Gene-to-phenotype correlation guides diagnosis. Specific genetic mutations predict disease severity, amyloidosis risk, and treatment response.

Critical Care-Specific Diagnostic Considerations

Distinguishing Autoinflammatory Disease from Sepsis

The most common diagnostic trap in critical care is misdiagnosis of autoinflammatory disease as sepsis. Key distinguishing features include:

FeatureSepsisAutoinflammatory
Microbiological evidencePositive culturesNegative cultures
Antibiotic responseResolves with appropriate antibioticsNo response
Peripheral edemaCommon, earlyAbsent unless amyloidosis
Organ dysfunction patternRandom, based on sourcePredictable, organ-specific
RecurrenceNo (if treated)Regular periodicity
Family historyNot relevantOften positive
Seasonal variationNoMay vary
Autoimmune markersNormalNegative/low

Distinguishing Autoinflammatory Disease from Malignancy

Fever of undetermined origin (FUO) workup sometimes leads to malignancy investigation in patients with autoinflammatory disease. Key distinguishing features:

FeatureMalignancy-related feverAutoinflammatory
Fever patternIrregular, progressiveRegular, periodic
Symptom-free intervalsNo clear intervalsComplete recovery
Weight lossProgressiveNone or minimal
Imaging abnormalitiesSpecific to tumorMinimal, resolving
B symptomsPresent, progressivePresent during flares only
Symptom durationProgressiveEpisodic

MANAGEMENT IN CRITICAL CARE

Acute Flare Management

Immediate Stabilization

  • Supportive care: fluid resuscitation, electrolyte repletion
  • Fever management: acetaminophen, NSAIDs
  • Monitoring: vital signs, organ function
  • Consider ICU admission if:
    • Severe organ dysfunction
    • Difficulty distinguishing from sepsis
    • Amyloidosis-related acute kidney injury
    • CNS involvement

Empiric Therapy Consideration While awaiting diagnostic clarification, initial empiric coverage for sepsis is reasonable if:

  • Presenting with fever and hemodynamic instability
  • Unable to differentiate from infection

However, cultures should be negative (typically obtained during initial evaluation), and lack of antibiotic response should prompt diagnostic reconsideration within 24-48 hours.

Specific Anti-inflammatory Therapy

NSAIDs:

  • Colchicine: 0.5-1 mg stat, then 0.5 mg q6h for 24-48 hours during acute flares (especially FMF)
  • Indomethacin: 25-50 mg TID
  • Note: NSAIDs are adjunctive; should not delay definitive IL-1 therapy

Corticosteroids:

  • Methylprednisolone: 1 g IV daily or prednisone 1 mg/kg daily, tapering over 7-10 days
  • Useful for acute symptoms but not disease-modifying
  • Avoid prolonged therapy due to immunosuppression and other complications

IL-1 Inhibition: First-line Acute and Prophylactic Therapy

Anakinra (IL-1 receptor antagonist):

  • Mechanism: Competitive inhibition of IL-1α and IL-1β signaling
  • Dosing: 100 mg SC daily; can increase to 100 mg BID or TID for refractory flares
  • Rapid onset: symptom improvement often within 24-48 hours
  • Half-life: ~4-6 hours (short, permitting rapid dosing adjustment)
  • Safety profile: Generally well-tolerated; main concern is injection site reactions and neutropenia (rare)
  • Cost: Relatively affordable (generic available)
  • Oyster: Acute worsening ("flare" phenomenon) can occur within hours of initiation in some patients; ensure patient education

Canakinumab (monoclonal anti-IL-1β):

  • Mechanism: Monoclonal antibody against IL-1β
  • Dosing: Highly variable based on disease and weight; typically 150-300 mg SC every 4 weeks after initial dosing
  • Longer half-life: ~28 days (permitting less frequent dosing)
  • FDA-approved specifically for CAPS and gout
  • Response time: 12-24 hours for acute effect, but full effect may take days
  • Cost: Expensive (most insurance requires prior authorization)
  • Pearl: Particularly useful in CAPS; considered first-line for that indication

Rilonacept (IL-1 trap):

  • Mechanism: Dimeric fusion protein acting as soluble IL-1 receptor
  • Dosing: IV induction followed by weekly SC injections
  • Approved specifically for CAPS
  • Intermediate half-life
  • Cost: Moderate

Chronic Disease Management and Prophylaxis

Disease-Specific Prophylactic Therapy

FMF:

  • Colchicine first-line (doses as noted above)
  • IL-1 inhibitors for colchicine-refractory or intolerant patients
  • TNF inhibitors (etanercept) as alternative second-line

TRAPS:

  • TNF inhibitors first-line (etanercept superior to infliximab in most series)
  • IL-1 inhibitors for TNF inhibitor-refractory cases

CAPS:

  • IL-1 inhibitors first-line (canakinumab or anakinra)
  • Nearly 100% response rate
  • Long-term therapy required; disease recurs upon drug discontinuation

MKD:

  • IL-1 inhibitors first-line
  • TNF inhibitors less effective than in TRAPS
  • Combined IL-1 and TNF inhibition in refractory cases

Monitoring During Prophylactic Therapy

General principles:

  • Periodic clinical assessment (frequency depends on disease activity and drug stability)
  • Laboratory monitoring:
    • Baseline: CBC, CMP, urine analysis, lipid panel
    • Periodic: CBC (monitor for cytopenias with anakinra), CMP (renal/hepatic function)
    • For patients with FMF on colchicine: CBC and CMP q3-6 months
    • Amyloidosis screening: Serum creatinine, urine APCE, serum amyloid A annually

Imaging:

  • DEXA scan annually for patients on long-term corticosteroids
  • Cardiac assessment if myocarditis or pericarditis present
  • Ophthalmologic assessment for CAPS (hearing, vision)

Monitoring for Amyloidosis

Secondary amyloidosis represents the most significant long-term complication of autoinflammatory diseases, particularly FMF, TRAPS, and chronic untreated disease.

Risk factors:

  • M694I homozygous genotype in FMF (highest risk)
  • Delayed diagnosis and inadequate treatment
  • Longer disease duration

Screening and diagnosis:

  • Serum creatinine and urine APCE annually
  • Serum amyloid A (SAA) as marker of chronic inflammation
  • Cardiac assessment (echo, ECG) for amyloid-related cardiomyopathy
  • If suspicion high: abdominal fat aspiration with Congo red staining (diagnostic for AA amyloidosis)

Management:

  • Aggressive anti-inflammatory therapy: target SAA levels <10 mg/L (or <50% of baseline)
  • ACE inhibitors or ARBs for renal protection
  • Strict blood pressure control
  • Consider antimalarial agents (hydroxychloroquine) for their potential anti-amyloid effects in some centers
  • Orthoptic liver transplantation in end-stage liver disease with amyloid deposition (rare, not generally recommended)

Oyster: Once overt renal amyloidosis develops (proteinuria, declining GFR), progression to end-stage renal disease is often inevitable despite therapy. Prevention through early diagnosis and adequate treatment is the critical strategy.


SPECIAL CLINICAL SCENARIOS

Autoinflammatory Disease Presenting as Acute Abdomen

Clinical Challenge: Acute peritonitis in autoinflammatory disease frequently triggers surgical consultation. The severity of abdominal pain often disproportionate to clinical findings can lead to unnecessary surgical intervention.

Clinical Features Favoring Autoinflammatory Over Surgical Abdomen:

  • Severe pain disproportionate to peritoneal signs
  • Absence of rebound tenderness despite agonizing pain
  • Normal or mildly elevated WBC (not markedly elevated as in peritonitis from perforation)
  • Absence of free air on imaging
  • Rapid improvement with NSAIDs or colchicine (minutes to hours)
  • History of prior similar episodes with spontaneous resolution

Pearl: If abdominal imaging is normal or shows only mild peritoneal thickening without free fluid, free air, or bowel pathology, avoid surgery and trial anti-inflammatory therapy. "First trial colchicine, not surgery" should be the mantra for suspected FMF.

Oyster: Unnecessary surgical intervention in autoinflammatory disease patients can precipitate severe systemic flares or trigger complications from surgery itself, including anastomotic breakdown and sepsis.

Autoinflammatory Disease with CNS Involvement

CINCA/NOMID Meningitis

Clinical Presentation: Chronic meningitis that waxes and wanes without microbiological confirmation. CSF shows lymphocytic pleocytosis (typically 50-500 cells/μL with lymphocytic predominance), normal glucose, normal or mildly elevated protein. Cultures repeatedly negative.

Diagnostic Approach:

  • Repeat LP showing recurrent pleocytosis during flares
  • Brain MRI showing:
    • Chronic enhancement of meninges
    • Ventricular enlargement (hydrocephalus from chronic inflammation)
    • Cerebral lesions (infiltration or vasculitis)
    • White matter changes
  • Genetic testing for NLRP3 mutations
  • Elevated CSF IL-1β (if available through research laboratory)

Management:

  • IL-1 inhibitors dramatically effective (typically resolves symptoms within 48-72 hours)
  • Complications include ventriculoperitoneal shunt placement if hydrocephalus develops
  • Long-term anticonvulsants if seizures develop

Pearl: The diagnosis should be suspected in any patient with persistent or recurrent aseptic meningitis refractory to standard therapy, particularly with family history or prior episodes. Genetic testing and IL-1 inhibitor trial may be diagnostic and therapeutic.

Oyster: Delayed treatment of CINCA meningitis can result in permanent neurologic damage, including sensorineural hearing loss, developmental delay (in pediatric cases), or cognition impairment.

Autoinflammatory Disease with Myocarditis/Pericarditis

Clinical Presentation: Recurrent pericarditis or myocarditis in the context of systemic autoinflammatory disease. Patients may present with chest pain, dyspnea, hemodynamic instability, or arrhythmias.

Etiology: IL-1β drives myocardial and pericardial inflammation. Direct infiltration or immune-mediated damage can occur.

Diagnostic Approach:

  • ECG: May show diffuse ST elevation (pericarditis) or T wave inversions (myocarditis)
  • Troponin: Elevated in myocarditis
  • Echocardiography: Pericardial effusion, reduced ejection fraction, regional wall motion abnormalities
  • Cardiac MRI: Late gadolinium enhancement, edema
  • Endomyocardial biopsy rarely needed; shows lymphocytic infiltration if performed

Management:

  • NSAIDs and colchicine for pericarditis
  • IL-1 inhibitors for refractory cases or myocarditis
  • Corticosteroids for hemodynamically significant pericardial effusion
  • Pericardiocentesis if tamponade develops
  • Consider advanced heart failure therapies if severe systolic dysfunction develops

Pearl: Recurrent pericarditis in autoinflammatory disease patients is an indication to escalate anti-inflammatory therapy. Do not continue NSAIDs and corticosteroids alone if recurrence occurs.

Autoinflammatory Disease with Secondary Infection

Clinical Scenario: An autoinflammatory disease patient develops fever and sepsis in the context of their baseline autoinflammatory disease. This creates diagnostic and therapeutic complexity.

Diagnostic Challenge: Distinguishing autoinflammatory flare from superimposed infection:

  • Microbiological documentation is critical (blood cultures, imaging-directed cultures)
  • More toxic appearance or hemodynamic instability than typical autoinflammatory flare suggests infection
  • Lack of periodicity or deviation from typical flare pattern
  • Localized findings (pneumonia, UTI, cellulitis) in addition to systemic inflammation

Management:

  • Broad-spectrum antibiotics empirically
  • Continue or escalate anti-inflammatory therapy (do not hold IL-1 inhibitors due to fever)
  • Source control (drainage, debridement) if indicated
  • Close monitoring for clinical deterioration

Oyster: Immunosuppression from prolonged IL-1 inhibitor therapy may increase infection risk. Baseline immunization (seasonal influenza, pneumococcal vaccines) is important. However, infection risk is generally lower with IL-1 inhibitors compared to TNF inhibitors.


MANAGEMENT CONSIDERATIONS IN CRITICAL CARE: ADVANCED TOPICS

IL-1 Inhibitor Selection and Sequencing

Anakinra Advantages:

  • Rapid onset and offset (short half-life)
  • Permits rapid dose adjustment
  • Reversible effect if adverse events occur
  • Cost-effective
  • Useful for acute flares

Anakinra Disadvantages:

  • Frequent dosing (daily, potentially multiple daily)
  • Injection site reactions
  • Risk of neutropenia (though rare)
  • Suboptimal for chronic maintenance in some patients

Canakinumab Advantages:

  • Long half-life (weekly or monthly dosing)
  • Excellent for chronic prophylaxis
  • FDA-approved for specific indications
  • Superior for CAPS

Canakinumab Disadvantages:

  • Slower onset (12-24 hours vs. anakinra's 1-4 hours)
  • Expensive
  • Fixed dosing may be suboptimal for all patients
  • Slower to reverse if adverse events occur

Rilonacept Advantages:

  • Intermediate half-life
  • Effective for CAPS
  • FDA-approved

Rilonacept Disadvantages:

  • Intermediate speed (between anakinra and canakinumab)
  • Less experience in non-CAPS indications
  • Moderate cost

Clinical Pearl: In acute care settings, anakinra is often preferred due to rapid onset and flexibility. In stable chronic disease, canakinumab or rilonacept permit less frequent dosing and improved compliance. Many patients require sequential optimization (starting anakinra acutely, transitioning to canakinumab chronically).

IL-1 Inhibitor-Refractory Disease

Incidence: Approximately 10-20% of patients with CAPS, TRAPS, or other autoinflammatory diseases fail to achieve adequate response to IL-1 inhibitors monotherapy. Persistent fever, inflammation, or development of new manifestations warrants investigation.

Management Strategies:

Optimization of IL-1 inhibitor:

  • Inadequate dosing: Many patients require dose escalation (e.g., anakinra 150-300 mg daily or higher)
  • Malabsorption or drug-drug interactions: Verify therapeutic levels (if available)
  • Inadequate compliance: Verify adherence

Combination therapy:

  • IL-1 + TNF inhibition: Particularly useful in TRAPS (TNF inhibitor) + CAPS (IL-1 inhibitor phenotype), or dual responsiveness
  • IL-1 inhibition + JAK inhibitor: Emerging approach for refractory disease
  • IL-1 inhibition + corticosteroids: Bridge therapy while optimizing IL-1 inhibition

Alternative mechanisms:

  • TNF inhibitors (if primarily TNF-mediated disease)
  • JAK inhibitors (baricitinib, tofacitinib): Emerging option for refractory autoinflammatory disease
  • Targeted C5a inhibition: Emerging for specific autoinflammatory phenotypes

Oyster: Some patients with genetically confirmed autoinflammatory disease demonstrate atypical responsiveness to standard IL-1 inhibitors, suggesting underlying genetic heterogeneity or epigenetic modulation. Whole exome or genome sequencing may reveal secondary mutations contributing to therapy resistance.

Drug Interactions and Management Considerations

Colchicine Interactions:

  • Strong CYP3A4 inhibitors (ketoconazole, clarithromycin, ritonavir): Increased colchicine levels, toxicity risk
  • P-glycoprotein inhibitors: Similar risk
  • In renal insufficiency: Dose reduction essential (risk of myopathy and neuropathy)
  • In hepatic insufficiency: Use with caution

IL-1 Inhibitor Interactions:

  • Live vaccines: Contraindicated (risk of vaccine strain disease)
  • TNF inhibitors: Combination increases infection risk; generally avoided except for specific indications
  • Other immunosuppressants: Additive immunosuppression

Pearl: Comprehensive medication review essential before initiating IL-1 inhibitor therapy to identify interactions.


MANAGEMENT PEARLS AND HACKS

Pearl 1: The "Colchicine Trial"

In a patient with suspected FMF pending genetic confirmation, a therapeutic trial of colchicine can be both diagnostic and therapeutic. Dramatic symptom improvement within hours to days supports diagnosis and may prevent diagnostic delays.

Hack: Ensure adequate dosing during trial: 0.5-1.5 mg daily for months, not just single doses. Many initial treatment failures represent inadequate dosing rather than true colchicine resistance.

Pearl 2: The "IL-1β Level as a Biomarker"

Hack: Request serum IL-1β measurement during acute flare (simultaneously with standard labs). Marked elevation (>10-50 pg/mL, normal <5 pg/mL) supports inflammasome-mediated disease and predicts response to IL-1 inhibitors. This single test can facilitate diagnosis and guide therapy.

Pearl 3: Genetic Testing Strategy

Hack: Don't wait for genetic testing to initiate therapy. Begin supportive care and empiric anti-inflammatory therapy while genetic testing is pending (typically 2-4 weeks). This prevents unnecessary delays in treatment initiation.

Pearl: Phenotype guides genotype. Complete clinical characterization (fever pattern, rash type, organ involvement, age of onset) permits targeted genetic panel testing rather than shotgun whole exome sequencing, reducing cost and turnaround time.

Pearl 4: The "Autoinflammatory Disease Index of Suspicion" Checklist

Hack: Use this checklist to assess probability of autoinflammatory disease:

  • Recurrent fever with symptom-free intervals (1 point)
  • Periodicity predictable or triggered by known stimulus (1 point)
  • Negative blood cultures despite fever (1 point)
  • Lack of response to antibiotics (1 point)
  • Multi-system involvement with characteristic pattern (1 point)
  • Negative autoimmune workup (1 point)
  • Family history of similar symptoms (1 point)
  • Elevated CRP/SAA out of proportion to clinical signs (1 point)
  • Age of onset <40 years (0.5 points)
  • Consanguineous parents or Mediterranean/Middle Eastern ancestry (0.5 points)

Score interpretation:

  • <2 points: Low probability; continue infectious workup
  • 2-4 points: Moderate probability; consider genetic testing
  • 4 points: High probability; prioritize genetic testing and IL-1 measurement

Pearl 5: "Flare Prevention Optimization"

Hack: In patients with autoinflammatory disease on prophylactic therapy, ask specifically about triggers:

  • Temperature extremes: Cold exposure (CAPS), heat exposure (rare)
  • Stress: Emotional or physical stress (all types)
  • Fatigue: Particularly in FMF
  • Menstrual cycle: Flares in perimenstrual period (FMF, CAPS)
  • Dietary factors: NSAIDs, infections
  • Medications: Certain drugs may trigger flares

Hack: Trigger avoidance combined with optimized prophylaxis provides superior flare control. Prophylactic colchicine pre-stress (starting 1-2 days before anticipated stressor) can prevent flares.

Pearl 6: Transition Therapy Strategy for Acute Presentations

Hack: In a critically ill patient with suspected autoinflammatory disease:

Hour 0-1:

  • Stabilize hemodynamics, ensure adequate oxygenation
  • Send cultures (blood, urine, imaging-directed)
  • Initiate broad-spectrum antibiotics

Hour 1-4:

  • Complete diagnostic workup (imaging, labs)
  • Consider IL-1 measurement (if available stat)
  • If high suspicion: Initiate anakinra 100 mg SC (can repeat q4-6h) or canakinumab IV (if available)

Hour 4-12:

  • Document response (fever resolution, symptom improvement)
  • If rapid improvement, continue IL-1 inhibitor and de-escalate antibiotics as cultures remain negative
  • If no improvement, reassess diagnosis; consider sepsis protocol

Day 1-2:

  • Review culture results
  • Genetic testing pending
  • Transition anakinra to maintenance dose (100 mg daily) or arrange canakinumab dosing
  • Plan prophylaxis strategy

Pearl 7: Amyloidosis Prevention Strategy

Hack: For all patients diagnosed with autoinflammatory disease, establish baseline renal function and implement monitoring:

  • Serum creatinine and eGFR at diagnosis
  • Urine APCE at diagnosis and annually
  • Serum amyloid A at diagnosis

Hack: In high-risk patients (M694I homozygotes, compound heterozygotes with severe genotypes), implement aggressive prophylactic therapy from diagnosis. Early intervention prevents amyloidosis development more effectively than treating established amyloidosis.

Pearl 8: "The Genotype-Phenotype-Therapy Triangle"

Hack: Understanding three nodes of this triangle guides optimal management:

  1. Genotype: Specific mutation predicts disease severity, amyloidosis risk, preferred therapy
  2. Phenotype: Clinical presentation (CAPS vs. FMF vs. TRAPS) guides therapy even before genetic confirmation
  3. Therapy: Specific drugs target specific pathways; genotype-phenotype correlation optimizes selection

Example: Patient with FCAS (CAPS phenotype) → NLRP3 mutation → IL-1 inhibitor first-line. Patient with TRAPS phenotype + M694I mutation → compound disease requiring TNF inhibitor + IL-1 inhibitor combination.

Pearl 9: Monitoring for Treatment Complications

Hack - Anakinra: Monitor CBC q1 month initially for neutropenia (rare but serious); injection site reactions common (rotating injection sites reduces incidence). Monitor renal function (dose adjustment needed if eGFR <30).

Hack - Colchicine: Screen for myopathy (gradual onset muscle weakness, elevated CK) and neuropathy; more common with high doses or renal insufficiency. Monitor CBC for bone marrow suppression.

Hack - All IL-1 inhibitors: Live vaccines contraindicated; update patient on inactivated vaccine schedules. Screen for latent TB before TNF inhibitors.

Pearl 10: Patient Education as Therapeutic Tool

Hack: Educated patients provide critical diagnostic information and comply better with therapy:

  • Teach fever tracking: Patients can identify periodicity better than providers reviewing quarterly visits
  • Distribute printed materials about autoinflammatory disease
  • Connect with patient advocacy organizations (e.g., Autoinflammatory Alliance)
  • Teach trigger avoidance
  • Emphasize importance of adherence to prophylactic therapy even during symptom-free intervals

EMERGING THERAPIES AND FUTURE DIRECTIONS

JAK Inhibitors

Baricitinib and other JAK inhibitors target Janus kinase signaling, which is crucial for IL-6 signaling and downstream effects of IL-1. Early case reports suggest efficacy in refractory autoinflammatory disease. Advantages include oral administration and potential efficacy in IL-1 inhibitor-refractory cases. Clinical trials are ongoing.

Complement Inhibition

C5a and other complement components contribute to autoinflammatory disease pathophysiology. C5a inhibitors (iptacopan) demonstrate promise in preliminary studies and may represent future therapy for specific autoinflammatory phenotypes.

Selective IL-1β vs. IL-1α Inhibition

Emerging selective IL-1β inhibitors may provide superior efficacy with fewer off-target effects. The role of IL-1α (often cell-associated, not secreted) in autoinflammatory disease is increasingly recognized.

Pyrin-Modulating Therapy

For FMF, therapeutic approaches targeting pyrin function directly (rather than downstream IL-1β) are under investigation. This may permit more targeted therapy and earlier diagnosis.


CRITICAL PEARLS FOR CRITICAL CARE PHYSICIANS

Pearl 1: Autoinflammatory diseases should be in the differential diagnosis of any adult with recurrent fever of undetermined origin, particularly when periodicity is evident and standard infectious workup is negative.

Pearl 2: The absence of positive cultures in a febrile patient should raise suspicion for autoinflammatory disease, not lead to continued broad-spectrum antibiotic therapy.

Pearl 3: Multi-system involvement with characteristic patterns (fever + arthralgia + abdominal pain + rash + serositis) is classic for FMF; don't anchor on a single organ system.

Pearl 4: Genetic testing should not delay initiation of empiric anti-inflammatory therapy in suspected autoinflammatory disease. IL-1 inhibitor trial is both diagnostic and therapeutic.

Pearl 5: A single therapeutic trial of colchicine (in FMF) or IL-1 inhibitor resulting in dramatic symptom improvement is both diagnostic and provides direction for long-term management.

Pearl 6: Amyloidosis represents the most serious long-term complication; early diagnosis and adequate prophylactic therapy prevent amyloidosis development and resultant renal failure.

Pearl 7: Secondary amyloidosis in the context of chronic autoinflammatory disease often progresses relentlessly despite current therapies; prevention is the only reliable strategy.

Pearl 8: Autoinflammatory disease patients presenting with acute abdomen should be managed medically first (colchicine/IL-1 inhibitor trial) before proceeding to surgery unless clear surgical pathology is documented.

Pearl 9: CNS involvement (chronic meningitis, vasculitis) in autoinflammatory disease is life-threatening and requires urgent IL-1 inhibition; don't anchor on infectious etiologies alone.

Pearl 10: Patient-centered care with education about triggers, prophylaxis adherence, and monitoring empowers patients and improves outcomes.


CONCLUSION

Autoinflammatory diseases represent an increasingly recognized category of disorders with significant implications for adult critical care practice. The shift from viewing these conditions as rare pediatric curiosities to recognizing their prevalence in adult populations requires fundamental changes in diagnostic thinking and therapeutic approach.

The clinical clues—recurrent fever with periodicity, lack of microbiological documentation, characteristic multi-system involvement, negative autoimmune markers, and treatment response to specific anti-inflammatory agents—should trigger suspicion and drive diagnostic testing. Modern genetic approaches have expanded the recognized spectrum of autoinflammatory diseases and now permit definitive diagnosis in most patients.

The availability of targeted anti-inflammatory therapies, particularly IL-1 inhibitors, has revolutionized management. These therapies are not merely symptomatic but disease-modifying, preventing catastrophic long-term complications such as secondary amyloidosis. Early diagnosis and appropriate therapy can normalize quality of life and prevent organ dysfunction.

For critical care physicians, the key is maintaining high clinical suspicion when traditional diagnostic algorithms fail. Recognition of autoinflammatory disease permits rapid de-escalation of unnecessary antimicrobial therapy, initiation of appropriate anti-inflammatory treatment, and prevention of iatrogenic harm from misguided interventions.

As our understanding of innate immunity and inflammasome biology advances, further therapeutic opportunities will emerge. However, the fundamental clinical principle remains unchanged: careful history-taking, attention to temporal patterns, and willingness to consider diagnoses outside the conventional infectious disease or malignancy frameworks will identify these patients and permit appropriate, life-altering intervention.


REFERENCES

  1. Goldbach-Mansky R. Autoinflammatory diseases. Immunol Allergy Clin North Am. 2020;40(1):1-20.

  2. De Benedetti F. Autoinflammatory syndromes. Curr Opin Rheumatol. 2018;30(1):1-7.

  3. Gattorno M, Federici S, Pelagatti MA, et al. Diagnosis of autoinflammatory diseases in the genomic era. J Clin Immunol. 2018;38(3):383-394.

  4. Ter Haar NM, Lachmann HJ, Özen S, et al. Treatment of autoinflammatory diseases: consensus recommendations. Lancet Rheumatol. 2022;4(1):e41-e50.

  5. Kastner DL, Aksentijevich I, Goldbach-Mansky R. Autoinflammatory disease reloaded: a clinical perspective. Cell. 2010;140(6):784-790.

  6. Cowen EW, Goldbach-Mansky R. TNFR-associated periodic syndrome and the cryopyrinopathies: An overview of pathogenesis and management. Dermatol Clin. 2013;31(3):507-526.

  7. Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, et al. Canakinumab in CAPS Study Group. Use of canakinumab in the cryopyrin-associated periodic syndrome. N Engl J Med. 2009;360(23):2426-2437.

  8. Soriano A, Soriano M, Navarro F, et al. Acute phase response in familial Mediterranean fever. Ann Rheum Dis. 2006;65(11):1500-1502.

  9. Özen S, Aladag OÜ. Diagnosis and management of familial Mediterranean fever in the genomic era. Curr Opin Rheumatol. 2017;29(1):5-10.

  10. Aksentijevich I, Livneh A, Klar A, et al. Ancient Mediterranean ancestry of the common MEFV mutation causing familial Mediterranean fever. J Med Genet. 2001;38(5):346-349.

  11. Ting JP, Lovering RC, Alnemri ES, et al. The NLR gene family: a standard nomenclature. Immunity. 2008;28(3):285-287.

  12. Gattorno M, Tassi S, Carta S, et al. Pattern of interleukin-1β secretion in response to lipopolysaccharide and ATP before and after interleukin-1 blockade in patients with CIAS1 mutations. Arthritis Rheum. 2007;56(9):3138-3148.

  13. Dinarello CA, van der Meer JW. Treating inflammation by blocking interleukin-1. Nat Rev Drug Discov. 2010;9(7):519-532.

  14. Lachmann HJ, Hawkins PN. Amyloidosis and autoinflammatory diseases. N Engl J Med. 2016;375(15):1496-1497.

  15. McDermott MF, Aksentijevich I, Galon J, et al. Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell. 1999;97(1):133-144.

  16. Williamson LM, Hull RP, Lachmann HJ. TRAPS and other TNF-mediated autoinflammatory diseases. Clin Exp Immunol. 2018;191(3):267-275.

  17. van der Meer JW, Vossen JM, Radl J, et al. Hyperimmunoglobulinaemia D and periodic fever: a new syndrome. Lancet. 1984;1(8386):1087-1090.

  18. Burgner D, Davila S, Breunis WB, et al. A genome-wide association study identifies novel and functionally related susceptibility loci for Kawasaki disease. PLoS Genet. 2009;5(1):e1000319.

  19. Federici S, Gattorno M. Autoinflammatory diseases: where do we stand in the era of immunotherapy? Rheumatology. 2019;58(2):217-228.

  20. Soriano A, Soriano M. Familial Mediterranean fever: diagnosis and management of a rare autoinflammatory disease. Expert Rev Clin Immunol. 2014;10(12):1579-1593.

  21. Lachmann HJ. Periodic fever syndromes. Immunol Allergy Clin North Am. 2015;35(2):199-213.

  22. Masters SL, Simon A, Aksentijevich I, et al. Horror autoinflammaticus: the molecular pathophysiology of autoinflammatory disease. Annu Rev Immunol. 2009;27:621-668.

  23. Savic S, Dickie LJ, Battellino M, et al. Autoinflammatory syndromes and cellular responses to stress. Nat Rev Rheumatol. 2015;11(5):276-288.

  24. Frenkel J, van der Meer JW, Fiselier T. Periodic fever, aphthous stomatitis, pharyngitis, adenitis: a new syndrome? Clin Exp Immunol. 2003;134(3):470-475.

  25. Kuemmerle-Deschner JB, Tyrrell PN, Reess F, et al. Efficacy and safety of anakinra therapy in pediatric and adult patients with the autoinflammatory cryopyrin-associated periodic syndromes: review of the literature and presentation of fifteen cases. Semin Arthritis Rheum. 2011;41(3):409-417.

  26. Hoffman HM, Rosengren S, Boyle DL, et al. Prevention of cold-associated acute inflammation in familial cold autoinflammatory syndrome by interleukin-1 receptor antagonist. Lancet. 2004;364(9447):1779-1785.

  27. Ozen S, Bilginer Y. A clinical perspective on the treatment of cryopyrin-associated periodic syndromes. Expert Rev Clin Immunol. 2014;10(1):89-98.

  28. Gattorno M, Obici L, Cattalini M, et al. Cryopyrin-associated periodic syndromes: from basic pathophysiology to novel therapeutic approaches. Rheumatology. 2009;48(11):1357-1364.

  29. Aksentijevich I, Putnam CD, Remmers EF, et al. The clinical continuum of cryopyrinopathies: novel CIAS1 mutations in North American patients and a new biopsy model. Arthritis Rheum. 2007;56(4):1273-1285.

  30. Touitou I. Infevers: an evolving inclusive database for auto-inflammatory disorders. J Clin Immunol. 2014;34(8):927-936.

No comments:

Post a Comment

Biomarker-based Assessment for Predicting Sepsis-induced Coagulopathy and Outcomes in Intensive Care

  Biomarker-based Assessment for Predicting Sepsis-induced Coagulopathy and Outcomes in Intensive Care Dr Neeraj Manikath , claude.ai Abstr...