Juvenile Dermatomyositis and Other Inflammatory Myopathies in Children




Keywords

Juvenile idiopathic inflammatory myopathy (JIIM), overlap connective tissue disease, muscle enzymes in JIIM, inclusion body myositis, orbital myositis, myositis specific antibodies, immunogenetics of JIIM, therapy of JIIM

 




Introduction


Patients with idiopathic inflammatory myopathies (IIM) manifest a diverse group of syndromes that have in common the loss of muscle function as a disease symptom. The symptoms of inflammatory muscle disease in children can be characterized as either acute (reviewed in Chapter 36 , [Case Example 36.3] , Chapter 1 ) or chronic in nature. Throughout the world, acute muscle complaints are usually associated with bacterial or parasitic infectious agents. In contrast, in North America, acute inflammatory myopathies are more often of viral etiology. With respect to chronic myositis in children, there is considerable variation in disease onset, presentation, and response to therapy. This chapter reviews the spectrum of disease manifestations of the chronic inflammatory myopathies of childhood and their association with specific autoantibodies and immunopathology, as well as our current approaches to medical therapy for the major subgroups of these disorders.


The most common pediatric presentation of the juvenile IIM (JIIM) is juvenile dermatomyositis (JDM). The primary clinical feature of both juvenile dermatomyositis and juvenile polymyositis (JPM) is chronic and progressive weakness of proximal muscles. In JDM, the distinctive skin manifestations ( Figure 42.1 ) and vasculopathy ( Figure 42.2 ) are commonly associated with muscle involvement. In children with JPM, there is no evidence of skin involvement. The child with definite JDM, in addition to the typical rash, has three of the four criteria: elevated serum levels of muscle-derived enzymes, electromyographic (EMG) evidence of inflammatory myopathy, positive muscle biopsy, and proximal muscle weakness ( Table 42.1 ). These criteria for diagnosis rely on the selection of the involved inflammatory site for two (EMG, muscle biopsy) of the four criteria. Because the inflammation is quite focal, it is essential to use magnetic resonance imaging (MRI), or other imaging to guide the biopsy site selection. A compatible MRI image is considered by some to be a critical diagnostic criteria. The use of ultrasound for biopsy site selection may also be fruitful. Recent information about serologic characteristics of patients with inflammatory myopathy has led to the recognition of a growing panel of myositis-specific antibodies (MSAs) associated with characteristic patterns of disease onset and course.




Figure 42.1


Cutaneous features of juvenile dermatomyositis. ( A ) The rash of JDM showing the periorbital edema, erythema crossing the bridge of the nose, and swelling of the right cheek. ( B ) Severe diffuse involvement with poikilodermatous changes on the chest. ( C ) Rash over the buttocks can be painful, making sitting uncomfortable. ( D ) The hands of an adolescent with JDM showing severe Gottron’s papules, periungual thickening, and dilated nailfold capillary loops. ( E ) Inflammation and secondary infection of an area of dystrophic calcification in a newly diagnosed child with JDM.



Figure 42.2


Vascular features of juvenile dermatomyositis. ( A ) Characteristic capillary dilation on margin of the eyelid of a child with active JDM. Capillary thrombosis is seen, reflecting active intravascular coagulation. ( B ) Telangiectasia (arrow) on the eyelid of a child with chronic severe JDM. ( C1 C3 ) The spectrum of changes in the nailfold capillaries in children with untreated JDM. Dropout of nailfold capillary loops are seen with increased periungual avascularity. The dilated capillaries often assume bizarre bush forms. ( D ) Improvement in the number of nailfold capillary end row loops (ERL) over time in a subset of JDM. ( E ) Muscle biopsy of a child with longstanding JDM showing vascular occlusion by smooth muscle proliferation. The lumen of the capillary is further compromised by adhesion of mononuclear cells to the high endothelial lining.


Table 42.1

The Modified Diagnostic Criteria for Inflammatory Myopathies

















































1 Symmetric, often progressive, proximal muscle weakness
2 Characteristic electromyographic (EMG) triad seen in myositis Short duration, small, low amplitude potentials
Fibrillation potentials, seen even at rest
Bizarre high-frequency repetitive discharges
3 Elevations of serum levels of muscle-associated enzymes Creatine kinase (CK)
Aldolase
Lactate dehydrogenase (LHD)
Transaminases (ALT/SGPT and AST/SGOT)
4 Evidence of chronic inflammation in muscle biopsy Necrosis of type I and type II muscle fibers
Degeneration and regeneration of myofibers with variation in myofiber size
Interstitial or perivascular mononuclear cells
5 Characteristic rashes of dermatomyositis Scaly erythematous eruptions over the metacarpal phalangeal or interphalangeal joints (Gottron’s papules)
Periorbital purplish discoloration (heliotrope rash)
Erythematous scaly rashes over the face, neck (V sign), upper back and arms (shawl sign), and extensor tendons (linear extensor erythema) and other extensor surfaces
6 Other potential criteria MRI compatible with diagnosis
Upregulation of class I antigen expression in muscle
Microvascular damage

Definite Disease: All criteria 1–4, except for dermatomyositis, in which any three of the first four criteria plus the rash are adequate.

Probable Disease: Any 3 of the first 4 criteria, except for dermatomyositis, in which any 2 of the first 4 criteria plus the rash are adequate.

Possible Disease: Any 2 of the first 4 criteria, except for dermatomyositis, in which any one of the first 4 criteria plus the rash is adequate.

Modified with permission from Bohan and Peter (1975). (#6 includes other potential criteria currently under discussion.)


The most common of the inflammatory myopathies in children are JDM, JPM, and overlap connective tissue disease syndromes. Children with JDM comprise 85% of this inflammatory myopathy group, compared with about 37% in adults; JPM occurs in 4–8% or less of the total chronic inflammatory myopathies in children, compared with 27% in adults. The third major group of children, 6–12%, fall into the category of connective tissue diseases or overlap syndromes; these children often meet criteria for another autoimmune disease, most commonly systemic sclerosis, with antibody to PM/Scl or Scl-70, or Sjögren syndrome with antibody to SSA/Ro, or SSB/La antigen. There are far fewer cases in which there is highly localized inflammation in muscle (focal or nodular myositis, or orbital or ocular myositis). Finally, there is limited evidence of significant variations in the inflammatory response that can occur in the muscle of the child (proliferative myositis, eosinophilic myositis, granulomatous myositis, cancer-associated myositis), or a necrotizing inflammatory response in response to statins reported in adults. In addition, children may have “dermatomyositis sine myositis” where there is no apparent muscle involvement at all.




The Epidemiology of the Major Pediatric Inflammatory Myopathies


There is a bimodal age distribution for JPM and JDM (at 5 to 9 years of age, with an estimated incidence of 3.7 cases per million per year and at 10 to 14 years of age, with 4.3 pediatric cases per million per year). There is a separate adult peak at 45 to 64 years of age, with 10 cases per million per year) first reported in 1970. In the United States, a National Institutes of Health-sponsored registry of new-onset JDM documented an incidence of JDM of 3.2 cases per million children per year, as determined by the capture and recapture method. Most of the 325 cases occurred in white children (71%), compared with the Hispanic (12%) and African American (9%) populations. In this population, the female-to-male ratio was 2:1, and the mean age of the child at diagnosis was 6.7 for girls, but 7.3 years for boys; 25% of the children were ≤4 years old at disease onset ( Figure 42.3 ). There is controversy concerning the impact of a very young age at disease onset on JDM disease course and severity, as well as tendency to develop calcification. Table 42.2 lists some of the major symptoms observed at the diagnosis of JDM in white children and minorities. As seen in Box 42.1 , delay in diagnosis and institution of therapy is associated with increased frequency of soft tissue calcification. Table 42.3 displays the first definite symptom of JDM: rash or weakness. Table 42.4 presents some of the systemic manifestations commonly seen in children with definite JDM. The mortality rate from JDM has decreased in the past 70 years. Before corticosteroids, there was a 33% mortality rate and a 33% morbidity rate, but both of those outcomes have decreased, with earlier diagnosis and more aggressive therapy (see section on therapy). Factors associated with increased mortality include the diagnosis of overlap syndrome>JPM>JDM, the presence of aminoacyl-tRNA synthetase autoantibody (particularly anti-alanyl-tRNA synthetase autoantibodies) and accompanying interstitial lung disease (ILD) as well as other autoantibodies: anti-Ku, anti-La, and anti-Sm. Other elements contributing to premature death are older age at illness onset and at diagnosis, dysphagia, and abdominal perforation as the first signs of illness. Cardiac disease has also been identified in the pediatric patients with IIM, exhibiting features similar to those described in studies of adult myositis, encompassing rhythm disturbances, conduction abnormalities, pericardial effusion, left ventricular dysfunction, myocarditis, cardiomyopathy and congestive failure, but they do not appear to be associated with early death.




Figure 42.3


The age at JDM diagnosis in children with JDM, in panel A for girls and in panel B for boys. NIAMS National Registry (N=286).

Reproduced with permission from Pachman et al. (2005).


Table 42.2

Symptoms at the Diagnosis of JDM

































Rash 79 (100) a
Weakness 79 (100)
Muscle pain 58 (73)
Fever 51 (65)
Dysphagia 35 (44)
Hoarseness 34 (43)
Abdominal pain 29 (37)
Arthritis 28 (35)
Calcifications 18 (23)
Melena 10 (13)

From Pachman et al. (1998).

a Numbers in parentheses indicate percentage of untreated children.



Box 42.1





  • Calcinosis at diagnosis




    • Total population: 18/79 (23%)



    • Caucasian: 13/59 (22%)



    • Non-Caucasian: 5/20 (25%)




  • Median interval between first symptom and diagnosis




    • Without calcinosis: 3.0 months



    • With calcinosis: 4.5 months (p=.04)




Duration of Untreated Disease: Association with Dystrophic Calcification

Data from Pachman et al. (1998). Used with permission.


Table 42.3

The First Symptom (Rash and/or Weakness) of JDM and the Interval of Time (Median in Months) before the Second Symptom




































N Rash, 1 st Rash-Weakness Interval Median (range) * Weakness, 1 st Rash-Weakness Interval Median (range) Rash-Weakness Occurred at Same Time
White 59 30 2.0 (0.2–12.9) 15 2.0 (0.7–9.6) 14
Minority 20 12 3.5 (1.0–9.4) 4 6.0 (4.6–20.0) 4
All 79 42 2.0 (0.2–12.9) 19 3.2 (0.7–20.0) 18

From Pachman et al. (1998). Used with permission.

* Median in months.



Table 42.4

Systemic Manifestations of Untreated Children with JDM





















































































1. General symptoms Fatigue
Malaise
Weight loss
Raynaud’s syndrome
2. Musculoskeletal features Muscle weakness, pain, and tenderness
Arthralgia, usually symmetric and involving the hand joints
Non-erosive polyarthritis, usually symmetric, mild and responsive to corticosteroids
Deforming arthropathy of hands is rare, but is associated with subluxation of the thumb interphalangeal joints (floppy thumb sign)
Carpal tunnel syndrome
3. Cutaneous findings Dematomyositis-associated rashes
Gottron’s papules
Heliotrope rash
V-sign and shawl sign rashes
Linear extensor erythema
Other rashes
Roughening, scaling, and erythematous fissuring of the palmar and lateral aspects of the fingers (mechanic’s hands)
Photosensitivity
Periungual abnormalities including telangiectasias and cuticular overgrowth
Irregular indurated plaques over the fingers with mucin accumulation in the dermis
Subcutaneous and intradermal calcification, which may ulcerate with secondary infection
Vasculitis with infarcts and digital ulcers
4. Pulmonary manifestations Ventilatory insufficiency caused by respiratory muscle weakness
Atelectasis
Aspiration pneumonia in patients with dysphagia
Interstitial lung disease
Drug-related pneumonitis
Opportunistic infections in immunocompromised patients
5. Cardiac involvement Myocarditis with arrhythmias and congestive failure
Cor pulmonale
Rare pericarditis
6. Gastrointestinal manifestations Abnormal pharyngeal and cricopharyngeal function
Esophageal dysphagia with occasional nasal regurgitation
Delayed gastric emptying and reflux
Dysmotility of the small and large intestines
Vasculitis with infarcts and necrosis of the bowel

Adapted from Pachman and Miller (2001).


There is controversy about the presence of a seasonal pattern in disease onset of JDM and adults with polymyositis/dermatomyositis (PM/DM), but the data appear to vary by geographic region and by year, defying reproducibility. In the north central region of the United States, children with definite JDM (diagnosed within 4 months of onset) were more likely to have their first symptoms between January and June than at other times in each of 7 years (1974–1980). An investigation of 79 newly diagnosed cases of JDM from all regions of the United States (1989–1992) identified an increased frequency of disease onset in the spring and summer of some years, but a survey of US JDM did not confirm that finding. In the United Kingdom and Ireland, several clusters of children with new symptoms of JDM were identified, the largest of which was in April and May 1992, but the timing of other clusters varied from year to year. When season of birth was investigated, some subgroups of patients with IIM had seasonal birth distributions. Hispanic patients with juvenile-onset IIM had a seasonal birth pattern (mean birth date October 1) significantly different from that of Hispanic controls (p=0.002), who had a uniform birth distribution, and from that of non-Hispanic patients with juvenile-onset IIM (p<0.001), who had a mean birth date of May 2. Juvenile dermatomyositis patients with p155 autoantibody had a birth distribution that differed significantly from that of p155 antibody-negative juvenile dermatomyositis patients (p=0.003). Juvenile IIM patients with the HLA risk factor allele DRB1*0301 had a birth distribution significantly different from those without the allele (p=0.021). Similar results were observed for juvenile and adult IIM patients with the linked allele DQA1*0501, versus juvenile and adult IIM patients without DQA1*0501, respectively. These data are of importance for they suggest that pre- and perinatal influences may play a role in disease susceptibility.


Other prenatal factors include studies of maternal chimerism in muscle tissue from males with JDM which may contribute to the development of the inflammatory response. Of note, monozygotic twins are equally chimeric because of the shared placenta, but it is still evident that they can be either concordant or discordant for development of IIM.




Pathogenesis/Genetic Data


Overview


Intensive study of children with newly diagnosed JDM suggests that the symptoms of the disease might follow exposure to infectious agent(s) in genetically susceptible individuals. Disease expression and chronicity appear to be facilitated by genetic risk factors located on chromosome 6, including the tumor necrosis factor alpha (TNFα)-308A allele, which may be exacerbated by exposure to ultraviolet B (UVB) light. Antecedent infection also appears to play a role in initiation of JDM symptoms. A case control study of over 320 US children documented a significant increase in antecedent symptoms in JDM children in the 3 months before diagnosis; the symptoms are primarily respiratory, often with concurrent gastrointestinal symptoms ; these observations were supported by those of Canadian investigators.


Infectious Agents and JDM


In addition to temporal and seasonal differences, there may be regional differences in infectious agents identified in association with the first symptoms of JDM. Agents associated with the onset (and on occasion, flare) of JDM include group A beta hemolytic streptococci and the RNA picornaviruses. Other infections that have been implicated in the pathogenesis of JDM include parvovirus, influenza, parainfluenza, and Borellia , as well as parasites, such as Toxoplasmosis gondii , but no evidence of related DNA or RNA has been identified in affected JDM muscle. Enteroviral agents are the most tantalizing, when antibody studies are considered. Newly diagnosed children (1974–1980) from the Chicago area were tested and displayed an increased frequency of antibody—both neutralizing (N) and complement fixing (CF)—to the coxsackievirus B antigen. There may be wide variation in different years, because these data were not reproduced by the same laboratory in a study of 20 children with dermatomyositis from the same region with onset of disease in the years 1987 to 1992. In studies searching for genetic evidence of infection, enteroviral RNA was identified in the muscle of UK patients with PM/DM, which was not confirmed in a similar patient population living in Sweden. Echovirus has been linked to the development of inflammatory myopathy in children with hypogammaglobulinemia and growth hormone deficiency, which was exacerbated by administration of growth hormone. In contrast, evidence of viral RNA has not been identified in the muscle biopsies from patients with myositis living in the United States, either in Jo-1 positive adults or in newly diagnosed, untreated children with active JDM, who had the specific area of the inflamed muscle identified for biopsy by MRI scan. There is also a possible role for molecular mimicry. Evidence supporting this hypothesis is based on a report of a protein present in skeletal myosin (which has homology with the streptococcal type 5M protein), which stimulated peripheral blood mononuclear cells in vitro . Blood from children who had “flares” of their dermatomyositis symptoms following streptococcal infections show a higher proliferative response than blood from children without a known antigen exposure. In parallel studies, an epitope specific for rheumatic fever did not stimulate the peripheral blood mononuclear cells from children with JDM.


Early reports of JDM suggested that exposure to T. gondii or hepatitis B might initiate the inflammatory myopathy. However, antibody titers to these agents were not increased either in a case control regional study, which tested sera from children with JDM in the mid-1980s or in a national case control study of new onset JDM. Noninfectious agents and exposures currently posited as causative agents in JDM include vaccines (hepatitis B, measles-mumps-rubella vaccine, typhoid, cholera) which a recent thoughtful appraisal could not refute. These data contributed to the current ban on the administration of live viral vaccines to children with JDM, irrespective of the level of their disease activity. In addition, drugs such as d -penicillamine, growth hormone, and bone marrow transplants in which graft versus host myositis occurs as a component of immune activation have been invoked as contributing factors to the display of IIM symptoms. Of note, influenza B, which typically causes a short episode of pain in the calf muscles of young children has not been associated with the development of juvenile chronic myositis. Taken together, the above data suggest that the etiology of JDM appears to be multifactorial, and may encompass a specific epitope(s) and environmental conditions in individuals who have a genetic predisposition to develop IIM.


Genetic Data


Several lines of evidence indicate that there is a strong genetic component that contributes to JDM display of symptoms. Although JDM may occur sporadically in more than one family member, the disease has been reported in monozygotic twins in whom muscle-related abnormalities developed 2 weeks after an upper respiratory infection. Proteomic studies of monozygotic twins with autoimmune disease, including JDM, suggests that autoimmune diseases share common pathways. Specific genetic associations have focused on the region controlling autoimmunity on chromosome 6 and previous data have identified DR3*0301 and TNFα in linkage disequilibrium with DQA1*0501 and DQA*0301 in US white patients. The strong association of JDM/DM and JPM/PM with this specific region on chromosome 6 was confirmed by a Genome Wide Association Study (GWAS) of a large population of Western European adult and pediatric patients with these diseases ( Figure 42.4 ). However, this association appears to be modified by race. When 151 white American adults with idiopathic inflammatory myopathy (IIM) were compared with 50 Korean adults with IIM, the linked allele DQA1*0501 and DRB1 alleles sharing the first hypervariable region motif 9EYSTS13 were major genetic risk factors for the development of myositis in Americans. Although both the white and Korean patients had similar distributions of clinical characteristics, autoantibody profiles, and clinical groups, none of the HLA-DRB1 or DQA1 alleles or motifs was a risk factor for IIM in the Korean patients. These data support the currently held assumption that the genetic risk factors for the inflammatory myopathies are multifactorial and appear to be associated with the patient’s ethnicity.




Figure 42.4


Genome Wide Association Study (GWAS). Manhattan plot of 1178 patients (European) with dermatomyositis (matched controls=4724) showing p values for 242,876 SNPs successfully genotyped. The orange line=7.3 (p=5×10 −8 ) and the blue line=4 (p=1×10 −4 ). SNP, single nucleotide polymorphism (2).

Reproduced with permission from Miller et al. (2013).


When muscle from untreated children with definite JDM was examined, Wt-1 (Wilms Tumor 1) was found to be massively hypomethylated, in contrast to the age-matched healthy controls. Of note, this hypomethylation was also true for children with JIIM who had partial therapy or those with overlap or JPM, suggesting that less muscle wasting may occur in children because they still have the capacity to repair cell damage—controlled by Wt-1. Studies of microRNA (mi-RNA) arrays in untreated JDM muscle disclosed that the mi-12 was downregulated and controlled the inflammatory cytokine cascade via NFkB. In addition, downregulation of mi-12 was associated with increased von Willebrand Factor-antigen (vWF:Ag) in JDM sera, suggesting that this might be one of the inflammatory pathways that contribute to the widespread microvascular vascular damage seen in JDM.


Maternal factors also might be associated with susceptibility to JDM. Boys with JDM appear to be chimeric because 73% of the boys had peripheral blood mononuclear cells (PBMCs) that contained female (XX) cells. These cells were positive for HLA-DQA1*0501 and were activated by the host male cells, which suggests that this antigen (or related MHC antigen) may be an important indicator of predisposing factor(s) for JDM (see the following).


Clinical Features at Diagnosis


The most common physical findings of JDM children at diagnosis are presented in Table 42.2 . Less commonly, hepatosplenomegaly and generalized lymphadenopathy are present at diagnosis in about 5% of children, indicating reticuloendothelial system activation. Box 42.1 shows that delay in diagnosis and therapy is associated with the development of dystrophic calcifications. Table 42.3 presents the first definite symptom of JDM: weakness or rash. The systemic symptoms that may be identified in the various idiopathic inflammatory myopathies are listed in Table 42.4 .


Cutaneous Findings


One of the early symptoms of JDM is an intermittent periorbital edema, which may be the only presenting sign. Periorbital erythema and edema ( Figure 42.1A ), and eyelid capillary vessel dilation may persist long after other signs and symptoms of disease activity have resolved; these are present in the majority of affected children. We have observed a flare in several children indicated only by erythema of the pinna of the ears, heralding the development of full-blown involvement of muscle and skin. As in children with systemic lupus erythematosus (SLE), the erythematous rash may cross the bridge of the nose and is malar in distribution. Sun exposure can precipitate exacerbation of symptoms (the children may sunburn easily) either initially, or once the disease has been diagnosed and treated. When rash is the first definite symptom of JDM, it is first observed in the summer months in 41% of newly diagnosed cases of JDM. Other areas of capillary involvement can be seen on inspection of the roof of the mouth, which shows dilated erythematous capillaries at the junction of the hard and soft palate, as well as swollen and often bleeding erythematous gums.


Proceeding distally, other cutaneous features include the neck, which may have a rash extending to the upper torso (the shawl sign) ( Figure 42.1B ). The examiner may find involvement of the extensor surfaces of the arms and legs, medial malleoli of the ankles, as well as the buttocks ( Figure 42.1C ), all of which may occur in the absence of elevated serum concentrations of muscle-derived enzymes. Skin over the knuckles (metacarpophalangeal joints and proximal interphalangeal joints) is often either hypertrophic or pale red (Gottron’s sign) and evolves into colorless bands of atrophic skin, which, during active disease, may have a papular, “alligator skin” type of appearance (Gottron’s papules) ( Figure 42.1D ). Pathologic calcifications occur at pressure points, which, if they break through the skin, offer a portal for infection (see Figure 42.1E and C ). Partial baldness may result as a consequence of chronic scalp inflammation ( Figure 42.5B ). The cuticle becomes dry and children frequently try to bite away the thickened skin at the base of the nailbeds. Other cutaneous signs found in JDM include paronychiae, often located on the lateral aspects of the nail of the great toe, which improve with symptomatic care and return of sufficient peripheral vascular blood flow. Livedo reticularis (lacy patterning on the skin) is frequent in children with Raynaud’s phenomenon and may indicate the presence of a hypercoagulable state as well as the presence of an antiphospholipid antibody. An excellent catalog of cutaneous manifestations in JDM is readily available. Newly diagnosed JDM parents, who were interviewed within 6 months of diagnosis, identified the rash of JDM before proximal muscle weakness in more than 50% of the children. The parents recognized the rash at the same time as the weakness in another 23% of children, and, in the remainder of the JDM children (27%), weakness was the first symptom reported by parents ( Table 42.4 ). Many children present with the amyopathic form of JDM (rash only) much later in their disease course. It is now recognized that anti-MDA5 antibody may be associated with malignancy in adults with amyopathic disease.




Figure 42.5


Features of chronic long-standing disease in JDM. ( A ) Lipoatrophy showing loss of fat over hand/fingers. ( B ) Scarring of the scalp following chronic inflammation with extensive hair loss. ( C ) Extension of pathological calcifications as seen on radiograph of the arm of a child with long-standing active disease.


The vascular features are one of the hallmarks of JDM. The upper eyelid margins frequently have a fringe of dilated capillaries ( Figure 42.2A ), some of which become thrombosed in active disease. The rash has a violaceous or heliotrope hue and is often most prominent on the eyelid where small infarctions can occur, as well as in the area of the medial canthi near the eyes in children who have a prominent component of necrotizing vasculopathy. Rarely, children with a chronic disease course can present with eyelid telangiectasia ( Figure 42.2B ). The spectrum of capillary changes in the nailfold is shown in Figure 42.2C1, 2, and 3 . The association with the number of nailfold capillary end row loops and the severity of the rash but not the muscle involvement is graphed in Figure 42.2D and the occlusion of capillaries and arterioles is demonstrated in the muscle biopsy ( Figure 42.2E ). If the active symptoms of JDM have been present for more than 1 year before treatment, there is greater likelihood that the lipodystrophy will be easy to detect ( Figure 42.5A ) (see information on chronic course later in this section).


Musculoskeletal Symptoms


Weakness is the first symptom in about 25% of cases ( Table 42.3 ) and is routinely evaluated using the Childhood Myositis Assessment Scale (CMAS). The evaluation of muscle strength is challenging in children, because it requires voluntary cooperation (see later section “Evaluation Tools for Disease Activity and Chronicity”). Proximal muscle weakness may be detected when the child has difficulty with climbing stairs, getting up from a chair, combing hair, or when using their hands to push off from the body in an attempt to stand up from the floor (Gowers’ sign). Weakness of the neck flexors, a particularly sensitive indicator of muscular impairment, is often the first area of impairment, and is frequently the last sign of weakness to normalize. Truncal weakness is common and can be demonstrated by asking the child to perform a sit-up (without a counterbalance to the feet) with the arms held in three positions: extended, crossed on the chest, and behind the head. However, even a healthy child younger than 5 years is often not able to clear the scapula; the endurance test of holding first the right arm and then the right leg off the examining table was not achievable by a good proportion of healthy children. More than 60% of children with JDM complain of pain on proximal, but not distal, compression of muscles, which is seen in PM and overlap syndromes, but it is less severe than the muscle pain in bacterial myopathies. Children with spinal muscular atrophy do not usually have muscle pain on compression. Fatigue is a common symptom of many dystrophies (especially Duchenne’s and Becker’s) and is not useful in differentiating children with JDM. Usually the child with an inflammatory myopathy is more comfortable when the limbs are held in the flexed position, promoting the development of flexion contractures, which may be difficult to reverse.


Cardiorespiratory Abnormalities


In more than half the children with definite JDM the electrocardiogram is abnormal at diagnosis. Asymptomatic conduction abnormalities predominate, with an occasional complete block of the right bundle branch, which usually resolves with control of the inflammatory process. Dilated cardiomyopathy can be a rare presenting complaint and usually resolves when all the indicators of immunologic activation have normalized. Tachycardia is common at diagnosis and when it continues during the disease course it reflects cardiac compromise, which can be detected by an increase in cardiac-specific indicators, such as troponin levels. Because the respiratory musculature in the trunk is almost always involved, most cases of untreated JDM have a restrictive ventilatory defect, accompanied by a decrease in ventilatory capacity. The decrease in ventilatory capacity can also be associated with diminished speech volume. A husky voice or vocal cord nodules, presumably as a result of the stress of trying to be heard, are present in approximately 3% of the over 600 children with JDM followed at our center. A study of long-term follow-up identified restrictive pulmonary impairment, associated with long-term JDM damage in 8% of patients, indicating a need for repetitive pulmonary follow-up in JDM patients.


Pulmonary Fibrosis


Pulmonary fibrosis is more commonly found in individuals who carry an antibody to the t-RNA synthetases, of which anti-Jo-1 is the most common. Children with JDM who have an antibody to Ro, or patients with an overlap syndrome with antibody to U1 RNP, or those with antibody to signal recognition particle (SRP) or anti-alanyl-tRNA synthetase (Pl-12), have a higher frequency of pulmonary fibrosis. They may develop a honeycomb appearance at the base of the lung, identified by high resolution computed tomography scan of the chest, and decreased diffusion of CO 2 and FEV1 on pulmonary function testing. It is critical to identify ILD early in the disease course, for not only is ILD a cause of death in older patients with JDM, but if recognized and treated promptly with compliance in childhood, the fibrosis can be reversed.


Gastrointestinal Involvement


The most severe prognostic indicator is impairment of the flow of secretions associated with decreased esophageal motility. This can be demonstrated by a rehabilitation cookie swallow with radiographic contrast, which shows retention of barium in a widened atonic, pyriform sinus, which can be associated with a “stacked coin” appearance. This anatomic change could allow swallowed liquids to penetrate the adjacent unprotected airway. Esophageal reflux may result in aspiration pneumonia, and appropriate precautions should be taken (e.g. using thickened foods, raising the head of the bed, and giving attention to clearing the respiratory tree). Involvement of the masseter muscles ( Figure 42.1A ) may be associated with difficulty in chewing as well as with swelling of the cheek(s) and the gradual development (over weeks/months) of buccal fat atrophy characteristic of partial lipodystrophy.


Boys and girls with untreated JDM were shorter and lighter than national norms (p>0.0005 for both), perhaps reflecting decreased GI absorption secondary to inflammation, and nonwhite children were weaker than white children (p>0.0005). Older children had more dysphagia (p=0.017) and arthritis (p>0.001). Vasculopathy can affect any part of the gastrointestinal tract. In severe disease, in addition to weight loss, mucosal ulceration with melena can occur with the possibility of life-threatening perforation. Pneumatosis intestinalis can occur as well, requiring bowel rest and aggressive immunosuppression. In the young child, development of normal speech patterns can be disturbed; soft palate involvement is often revealed by nasal, high-pitched speech (e.g. identified by having the child say the letter “E”) and usually resolves with a decrease in the inflammatory component of the myositis. A dilated atonic esophagus can be associated not only with delayed gastric emptying but also with infectious or neoplastic diseases. Smooth muscle dysfunction can also result in decreased lower gastrointestinal motility, making constipation an early and easily ascertainable symptom that responds to a stool modifier, such as Marilax, although chronic intermittent diarrhea has also been reported by the children or their parents as well. If there is a family history of celiac disease or the physician has an increased index of suspicion, then a celiac panel should be obtained, which if positive, should be confirmed by gastrointestinal biopsy, before the children embark on a gluten-free diet because they are thought to have celiac disease.


Genitourinary Function


Severe inflammation can result in massive breakdown of muscle elements as well as primary compromise of the renal parenchyma itself, which may culminate in acute renal failure. This requires prompt hydration with monitoring of renal function. Necrosis of the ureter with calcifications has been reported, involving the middle (iliac) segment, because of the relatively sparse blood supply to this region compared with the upper (lumbar) or lower (pelvic) segments.


Reproductive function may be altered in acute or chronic disease: menses may cease during severe disease, only to resume after the active inflammation is controlled. One study identified delayed menarche with normal cycles and low follicular reserve in JDM patients. The girls with JDM had decreased progesterone levels suggesting an underlying subclinical corpus luteum dysfunction in this disease. Lipodystrophy can result in impairment of fertility in female patients, especially if severe disease onset is before puberty. In addition, JDM girls may have abnormal levels of LH and FSH.


With respect to pregnancy, if the JDM symptoms in the mother are relatively quiescent, then both mother and infant will do well, but a JDM disease flare during pregnancy can be problematic. Active JDM symptoms during pregnancy—muscle weakness, rash flare—can usually be managed with the use of modest doses of corticosteroids (both IV and PO), given with periodic IVIg without harm to the fetus. However, pregnancy in a mother with JDM may be associated with increased fetal wastage secondary to antibodies, such as anticardiolipin antibodies, which alter coagulation. Anticardiolipin antibodies have been identified in children with JDM (often with a negative lupus anticoagulant) and can be associated with fetal death, as well as vascular thrombosis. Birth control measures, such as oral contraceptives, should take into account the possibility of anticardiolipin antibodies. The type of birth control used should be discussed with the patient and her OB/GYN physician to diminish the possible risk of increased intravascular thrombosis.


Ophthalmologic Findings


Children with a more severe vasculopathy may display ulceration and infarction of tissue, often in the region of the medial canthus of the eye, which heal and leave a depressed scar. Within the eye, in active inflammation of JDM, transient retinal exudates and “cotton wool” spots may occur after the occlusion of small vessels, leading to intraretinal edema with injury to retinal nerve fibers, optic atrophy, and sustained visual loss. Neovascularization of the retina with spontaneous regression has also been reported. Central retinal vessel occlusion has been associated with anticardiolipin antibody. Disease of conjunctivae vessels can lead to an avascular zone with a potential for infarction. If there is a family history of red-green color blindness, the use of hydroxychloroquine should be avoided. Orbital myositis can be documented by MRI, either targeting a single fast-moving muscle, such as the superior oblique or as part of the spectrum of inflammatory myopathy which responds to corticosteroid administration or other immunosuppressant and may be part of the IgG4 syndrome (see later text). Children treated with steroids should be monitored for glaucoma (especially if there is a positive family history!) and for the development of sublenticular cataracts, which often resolve as the child improves.


Other Disease Manifestations


Vasculopathy involving the central nervous system may be associated with depression or wide mood swings, which may be exacerbated by steroid therapy (see “Psychological Aspects of Juvenile Myositis” below). Very few children with JDM present with Raynaud’s phenomenon; these symptoms are more frequently found as isolated phenomena in children with overlap syndromes or scleroderma.


Juvenile Dermatomyositis with a Chronic Course


Lipodystrophy


Three stages of lipodystrophy are now recognized: (1) localized lipodystrophy, occasionally following panniculitis that may be painful; (2) partial lipodystrophy targeting the areas over intense proximal muscle inflammation; and (3) generalized lipodystrophy, in which fat is lost over a larger part of the body, muscle atrophy is prominent, and joint contractures may be present. Several clinical features can provide clues to the duration of disease, for lipodystrophy occurs later in the disease course, reported 4.6 years after first symptom. Loss of subcutaneous fat over the masseter area and over the arms and legs, proximal<distal, gives a ropey appearance to the musculature, with clear definition of muscle groups that are not hypertrophied ( Figure 42.5A ). In contrast, abdominal muscle tone is diminished, so that the child or young adult has difficulty with sit-ups, even when muscle strength is normal elsewhere. This loss of abdominal tone is accompanied by increased abdominal fat, with the appearance of a “pot belly.” Another prominent feature of partial and general lipodystrophy is acanthosis nigricans, or speckled areas of increased pigment, which may be found in the skin folds of the neck, the axilla, behind the knee, or between the buttocks and appears like “dirty skin.” An additional phenotypic feature of this illness is coarsening of hair texture, or, if there is chronic inflammation of the scalp, loss of hair in the affected area ( Figure 42.5B ). If the JDM is still active before puberty, then there may be loss of fertility, with masculinization, reflected by increased facial hair and change in body habitus. The clinical features of lipodystrophy are not rare and occur in about 20% of our JDM population: more than 30% had some evidence of fat atrophy, 22% had increased insulin levels, elevated levels of triglycerides were present in 10%, and three tenths of those tested for glucose tolerance had evidence of insulin resistance.


Dystrophic Calcifications


Calcification in soft tissue is a consequence of the duration and severity of the active symptoms of JDM. Although the number of children with JDM with calcium deposits in the soft tissues has decreased when this disease is diagnosed more promptly and treated aggressively to 14%, the national average is still about 30% and, in South Africa, 70% of children with JDM develop calcinosis. The pathologic calcifications often follow delayed diagnosis or insufficient therapy. In children with the JDM rash only who were not given initial aggressive therapy, myositis often develops at a later date and may be complicated by pathologic calcifications. The calcifications may occur at pressure points, often the elbows or buttocks, but also can be found in fascia surrounding muscle ( Figure 42.1E and C ), as well as in palmar surface of the digits in children with overlap syndromes, such as Pm/Scl. The calcifications may change in appearance from massive accumulations to become thinner, more pancake-like collections, which then may resolve spontaneously, draining as a white cheesy or serosanguineous exudate containing macrophages and leaving dry, pitted scars ( Figure 42.1E ). In persistent, active myositis, the calcifications may progress to become a sheath, impairing flexion and function, breaking the barrier of the skin to form a portal for infection. Sepsis is not uncommon, frequently from staphylococci, and is a major contributor to the morbidity and mortality related to this disease. Children with JDM who have calcium deposits have three times higher urinary excretion of γ-carboxyglutamic acid (GLA), a component of the vitamin K-dependent coagulation pathway. An intriguing finding is that children with JDM without pathologic calcifications have GLA excretion that is twice normal, suggesting a prominent role of this protein in the pathophysiology of the disease. Further characterization of the calcifications show that they are not bone-like when studied with Fourier transform infrared spectroscopy, although they do have selective deposition of bone matrix proteins.


Calcification—Differential Diagnosis


It is important to differentiate the etiology of dystrophic calcinosis in JIIM from that in other syndromes such as in heterotopic calcinosis or that following trauma. Heterotopic calcinosis occurs in children with progressive osseous heteroplasia. In these individuals, there may be evidence of both intramembranous and endochondral ossification. In the cases of progressive osseous heterotopia described in the literature, the children presented at birth or within the first 6 months of age with only the cutaneous calcifications. This entity can be differentiated from fibrodysplasia ossificans progressiva, in which congenital malformations of the big toes are present at birth.


Disease Chronicity


Flexion contractures, hyperlipidemia, and cardiovascular disease are a few of the additional consequences of chronic inflammation. The position of comfort in the active phase of acute myositis is the flexed position which, if maintained for too long a period, becomes fixed with loss of both range of motion and of function; calcifications in the flexor tendons compound the problem. ILD is a leading cause of mortality in children with IIM. Short stature, present at diagnosis, is maintained for both boys and girls with JDM once they reach adulthood, when they are at increased risk for atherosclerotic heart disease and the metabolic syndrome, which may share features of the accelerated cardiovascular disease seen in adults with SLE as well as children.


Other Autoimmune Diseases


Many conditions must be considered in the differential diagnosis of inflammatory myopathy in children ( Table 42.5 ). Because myositis is often a component of other autoimmune rheumatic diseases, it is essential to exclude such conditions as SLE, mixed connective tissue disease, chronic arthritis in children (especially systemic onset juvenile rheumatoid arthritis [JRA] ), the spondyloarthropathies, Sjögren’s syndrome, and Kawasaki syndrome. They also may have a myositis specific antibody (MSA) that is often associated with a nonremitting disease course, such as Jo-1, the most common of the antibodies to the t-RNA synthetases ( Table 42.6 ). Furthermore, the inflammation can be concentrated in one specific muscle area (see “Other Myopathies” following), as in cases of orbital myositis ; it can be a nodular or proliferative process, or it can involve an eosinophilic infiltration of the fascia. Children with muscular dystrophy or a dysferlin deficiency associated with inflammation may present with muscle weakness (most often distal) as well as elevated levels of serum derived. Other conditions must also be excluded before the diagnosis of JDM is established. The endocrinopathies must also be included in the differential diagnosis. We have observed proximal and distal muscle weakness and elevated muscle enzymes in children with hypothyroidism, hyperthyroidism, and hyperparathyroidism.



Table 42.5

Differential Diagnosis of JDM

































































































1. Drug- and toxin-induced myopathies Corticosteroids
Ethanol
Lipid-lowering drugs
D-Penicillamine
Colchicine
Ipecac
Chloroquine
Zidovudine (AZT)
L-Tryptophan (eosinophilia myalgia syndrome)
2. Endocrine diseases Hypothyroidism
Hyperthyroidism
Acromegaly
Diabetes mellitus
3. Neurologic disorders Amyotrophic lateral sclerosis
Myasthenia gravis
Multiple sclerosis
Guillain-Barré syndrome
Motor neuron disease
4. Other connective tissue diseases Polymyalgia rheumatica
Rheumatoid arthritis
Systemic sclerosis
Tendinitis and overuse syndromes
5. Metabolic abnormalities Hypokalemia
Hypercalcemia
6. Inherited metabolic defects Acid maltase deficiency
Phosphorylase deficiency (McArdle’s disease)
Lipid metabolic defects (carnitine or carnitine palmitoyltransferase deficiency)
Phosphofructokinase deficiency
7. Muscular dystrophies Duchenne’s/Becker’s
Facioscapulohumeral
Limb-girdle
Myotonic
Dysferlin deficiency
Merosin deficiency
8. Infectious myopathies Bacterial (pyomyositis)
Parasitic
Viral, including human immunodeficiency virus and HTLF-I and -II
9. Mitochondrial myopathies


Table 42.6

Myositis Specific and Associated Antibodies in JIIM




























































































Serologic Group Frequency (%) Clinical Subgroup Association in JIIM Comments References
MSA
Anti-p155/140 (TIF-1-γ) 23–30 JDM and JDM with overlap myositis Associated with extensive photosensitive skin rashes, including the characteristic rashes of JDM, malar rash, V-sign and shawl-sign rashes, and linear extensor erythema, as well as periungual capillary changes, skin ulceration, and generalized lipodystrophy; patients frequently have a chronic illness course; not associated with cancer-associated myositis, which differs from patients with adult IIM with this autoantibody
Anti-MJ (NXP-2) 20–25 Primarily JDM Frequent muscle cramps, dysphonia, joint contractures, and a monocyclic illness course; this autoantibody group seems to have more severe illness, with some reports of increased calcinosis, frequent muscle atrophy, more frequent hospitalizations and gastrointestinal ulceration
Anti-aminoacyl-tRNA synthetases (Jo-1 & non-Jo-1 synthetases) 2–4 JDM, JPM, and overlap myositis More common in JPM (9%) and patients with juvenile overlap myositis (8–13%); frequent ILD, arthritis, Raynaud phenomenon, fevers, and mechanic’s hands, similar to adults with these autoantibodies; among the MSA phenotypes, this group has the highest mortality caused by ILD
Anti-SRP 1 JPM More common in JPM (18%); seen primarily in African American girls with JPM who have severe to profound proximal and distal muscle weakness, frequent falling episodes, Raynaud phenomenon, very high CK levels, wheelchair use, and a chronic illness course; cardiac disease is also likely associated, and these patients are refractory to several therapies, similarly to patients with adult IIM with anti-SRP autoantibodies
Anti-Mi-2 (NuRD) 2–13 JDM and overlap myositis JDM with classic cutaneous findings of Gottron’s papules, heliotrope rash, and malar rash; predominantly seen in Hispanic patients; mild disease, but children have higher CK levels than adults with this autoantibody
Anti-CADM-140 (MDA-5) Unknown JDM Associated with DM and clinically amyopathic DM in adults who frequently have rapidly progressive ILD, cutaneous ulceration, and palmar papules, with a high fatality rate; 7 patients with JDM or amyopathic JDM and ILD have been reported with this autoantibody; to date, most reports are from Japan, and mortality has been high
Anti-SAE 0.2 JDM Associated with DM cutaneous manifestations, which predate development of muscle symptoms, an absence of constitutional manifestations, and low frequency of ILD
MAA
Anti-U1-RNP 5–10 JDM, JPM, and overlap myositis More common in JPM (12%) and overlap myositis (20–27%); associated with arthritis, Raynaud phenomenon, and sclerodactyly
Anti-Ro 2–6 JDM, JPM, and overlap myositis More common in overlap myositis (8–15%); little known about the associated clinical features; associated with impaired lung function in 1 report; in adults, patients respond well to prednisone alone; may be seen in association with anti-Jo-1 autoantibodies
Anti-PM-Scl 1–4 JDM, JPM, and overlap myositis More common in JPM (6%) and overlap myositis (10–25%); based on adult cohorts, this autoantibody is associated with Raynaud phenomenon, arthritis, ILD, and esophageal dysmotility; most frequent in Caucasian patients with an HLA DRB1*0301 immunogenetic association
Anti-Ku 0.2 JDM, JPM, and overlap myositis More common in overlap myositis (2%); based on adult cohorts, frequently associated manifestations include arthralgia, Raynaud phenomenon, myalgias, dysphagia, and less commonly, ILD; patients respond well to corticosteroids alone, except those with lung disease
Other MAAs: anti-U2-, U3-, or U5-RNP; anti-La; anti-Sm; anti-Th <1 JDM, JPM, and overlap myositis Each autoantibody is present more frequently in overlap myositis (1–10%); little known, primarily associated with overlap myositis
Myositis autoantibody negative 28–57 JDM, JPM, and overlap myositis Relatively mild disease in this subgroup; this group is likely heterogeneous, with several different unrecognized autoantibody groups contained within

Abbreviations: ILD, interstitial lung disease; JDM, juvenile dermatomyositis; JPM, juvenile polymyositis; MAA, myositis-associated autoantibodies; MDA-5, melanoma differentiation-associated gene 5; MSA, myositis-specific autoantibodies; NuRD, nucleosome remodeling deacetylase complex; SAE, small ubiquitin-like modifier activating enzyme; SRP, signal recognition particle; TIF-1-γ, transcriptional intermediary factor 1γ.

Reproduced with permission from Rider et al. (2013) with minor modifications.


Laboratory Indicators of Juvenile Dermatomyositis


Magnetic Resonance Imaging


Musculoskeletal inflammation at diagnosis and flare can be identified on magnetic resonance imaging (MRI), which guides the muscle biopsy, EMG, and other serologic assessments. The use of MRI to identify the specific area of often patchy inflammation in the muscle can direct the surgeon to the appropriate site for a muscle biopsy ( Figure 42.6 ). This approach minimizes error in sampling uninvolved areas in this focal disease and can serve, under very special circumstances, as an excellent monitor of a child’s response to therapy. The MRI findings may normalize several months later than the muscle enzymes. P31 magnetic spectroscopy has been used for the past few years and gives additional information about the child’s muscle strength and performance and has confirmed evidence of defective oxidative phosphorylation in diseased muscle.




Figure 42.6


( A ) Sagittal view of the MRI of a child positive for U1-RNP. The regions of muscle inflammation (white areas) are asymmetrical; the undirected biopsy of an area subsequently found not to be involved was read as “normal.” The MRI directed biopsy (white area) showed characteristic inflammation and muscle fiber damage. ( B ) Magnetic resonance image of a child with early symptoms of JDM and normal muscle enzymes. This T2 weighted image displays the patchy nature of the muscular involvement: the white areas (arrows) show the symmetrical edema of active inflammation surrounded by thickened fascia.


Muscle Enzymes


The standard muscle enzymes obtained for diagnosis are creatine kinase (CK), the transaminases aspartate aminotransaminase (AST) and alanine aminotransaminase (ALT), lactate dehydrogenase (LDH), and aldolase. The highest concentration of CK is present in excitable tissues (muscle and nervous system) and occurs in three isoforms, MM, MB, and BB. In skeletal muscle, the MM form predominates (90% to 95%), whereas immature muscle may have increased concentrations of the MB form (20% to 30% in children younger than 1 year of age). AST is found primarily in the heart, liver, skeletal muscle, and kidney, and ALT is also found in skeletal muscle but in much lower concentrations. LDH catalyzes the intraconversion of lactate and pyruvate and is found in most of the tissues of the body. Aldolase is also widely distributed in the tissues of the body, and increased circulating levels of aldolase may be seen not only in myopathies but also in disorders of the liver, the hematologic tissue, and other diseases. The child who has moderately active JDM on muscle biopsy can have an elevated level of aldolase or any one of the other commonly tested muscle derived enzymes, while the others are in the normal range. Furthermore, these muscle enzymes may normalize by 4.5 months after the first JDM symptom, despite continuing disease activity, losing their value as a guide to therapy ( Table 42.7 ).



Table 42.7

Time-related Normalization of Muscle Derived Enzymes at Diagnosis


































Enzyme Normal (%) Non-normal Cutpoint of Untreated JDM in months p
CK 33 (26%) 93 4.65 < 0.01
LDH 16 (13%) 110 2.53 < 0.01
Aldolase 22 (17%) 104 4.65 < 0.01
SGPT 21 (17%) 105 3.68 < 0.01

From Pachman et al. (2006).


Electromyogram


Evidence of inflammatory myopathy on EMG is not specific to JDM. Inasmuch as 20% of EMGs not guided by MRI are “negative” or normal, it is prudent to use MRI or ultrasound to identify the specific area of inflammation in the muscle. Once the location of the electrodes has been chosen (not the site of a future biopsy), insertional irritability followed by spontaneous electrical activity at rest is often observed. This pattern can also be seen in the muscular dystrophies and in early acute myositis. Abnormal, early, full recruitment of muscle fibers with moderate effort occurs in about 45% of patients with JDM, and bizarre, high-frequency discharges occur in 15% to 20% of patients tested. Reduced motor unit activity is seen in Duchenne’s muscular dystrophy (DMD) as well as in JIIM. A comprehensive comparison of DMD and JPM showed that the JPM patients more frequently had complex repetitive discharges on electromyography and a complete response to treatment with prednisone or other immunosuppressive agents than dystrophy patients (44% versus 0%). Myasthenia gravis can coexist with an inflammatory myopathy, resulting in a greater degree of instability of motor-unit potential than is found in the uncomplicated inflammatory myopathies. Given the child’s variable acceptance of this procedure, we often reserve the EMG testing for the definition of cases that are not straightforward. Study of diagnostic testing of 384 children who were entered into the Children’s Arthritis and Rheumatology Research Alliance (CARRA) registry indicated that EMG results were most likely to show nondiagnostic results with 50% of the children requiring an additional diagnostic study to confirm JDM.


Pathology


Muscle Biopsy


The vasculopathy of JDM may occur in the absence of a prominent inflammatory component. Capillaries and arteriole damage with loss of the capillary network results in structural change in the nailfold capillary bed as well as in muscle, with a subsequent decrease in the capillary-to-fiber ratio. Vascular compromise and capillary dropout, with perifasicular atrophy of both type I and type II fibers, are seen in the muscle pathology in JDM ( Figure 42.7 ). Multiple satellite cells are frequently found adjacent to atrophic fibers; focal repair occurs concomitantly with fiber atrophy. Low-grade ischemia may also be related to the increased expression of class I and class II major histocompatibility complex gene products. Greater than 50% expression of class I by muscle fibers is now considered as one diagnostic criterion for inflammatory myopathy. In contrast, in PM, there appears to be much less primary involvement of vessels; that is, they may be normal in number and structure. Some children with PM have the same histologic features as children with the rash of JDM (perifasicular atrophy, capillary occlusion), which may extend the spectrum of disease pathology for the diagnosis of JDM. Other children with PM and antisynthetase antibodies, such as SRP, may have areas of necrosis, in comparison with the documentation of apoptotic cell death in JDM muscle, which increases proportionally to the time that the inflammation is untreated.




Figure 42.7


Muscle histopathology in JDM with 8 months of untreated disease: Hematoxylin and eosin stain shows extensive connective tissue proliferation and fibrosis while muscle fiber size variation is evident. The hallmarks of the disease, perifasicular atrophy and occlusion of capillaries and arterioles, are seen. The active inflammatory infiltrate is composed of primarily mononuclear cells.


In JDM, even when the serum levels of muscle-derived enzymes are normal, the MRI-directed muscle biopsy can reveal active foci of inflammation. Children with active inflammation and normal muscle enzymes may also have elevated levels of vWF:AG and/or neopterin, which can be used to guide therapy (see later discussion).


Skin Involvement


Study of the skin in JDM can yield valuable information about disease pathophysiology. Adults with DM and cutaneous ulceration may develop malignancy, which is not true for children with JDM. DM patients with “mechanics hands” are at higher risk for ILD. Inflamed DM skin contains an infiltrate composed of C4 and CD8 T cells, plasmacytoid dendritic cells, neutrophils along with basal keratinocyte vacuolar alteration with apoptotic keratinocytes, and mucin deposition. Type 1 interferons are produced by plasmacytoid dendritic cells and genome-wide expression data documents a robust upregulation of IFN-inducible genes in DM skin, as well as evidence of other inflammation pathways, complement activation, and epidermal activation and differentiation. In JDM, mast cells were increased in the skin of the child, compared to their number in the inflamed muscle from the same child obtained by MRI directed biopsy. Unlike in SLE, children with JDM do not display immunoflorescent band-like immunoglobulin deposits at the derma-epidermal junction, which is an important point in differentiating the two similar clinical presentations.


Immunologic Data


Humoral Immunity


The complex immune system, consisting of innate and adaptive components, is essential to the pathophysiology of JDM tissue damage, but the complete sequence from immune activation to target damage has not yet been deciphered. Humoral immunity per se may be abnormal in a minority of patients with IIM: children with hypogammaglobulinemia may develop inflammatory myopathy associated with echovirus infection, and IgA deficiency has been identified in a few children with undefined inflammatory myopathy. In immunocompetent children, early in the disease course, the IgM and or/or IgG may be elevated.


Myositis Specific Antibodies


As presented in Table 42.6 the evolving serological classification of JIIM has provided insight into some of the factors contributing to clinical myositis disease heterogeneity. Several methods are used to detect myositis-specific antibodies (MSA). For example, ELIZA determinations of MSA may be confounded by false positives as opposed to immunodiffusion, radiolabelling, and immunoprecipitation methods, which yield protein bands of different migratory characteristics. The most common MSA antibody in children with inflammatory myositis is directed against a doublet protein, p155/140. Specific skin sensitivity and patterns of erythema are associated with p155/140 and the children frequently follow a chronic course. This antibody is not associated with malignancy in DM/JDM and differs from the p155/140 antibody directed against TIF-1-γ (antitranscription intermediary factor-1-γ/α (TIF-1-γ/α)) found in adults with DM/PM who harbor a malignancy. Anti-MJ occurs in 20–25% of patients, is directed against nuclear matrix protein 2 (NXP-2), and is associated with a monocyclic course, more severe disease manifestations, more frequent hospitalizations, and calcinosis. Only 2–4% of pediatric IIM patients have antibody to the aminoacyl-tRNA synthetases (Jo-1 and non-Jo-1 synthetases) present both in JPM (9%) and in overlap myositis (13%). These patients have frequent arthritis, and should be rigorously monitored for ILD, which results in the highest mortality of all the MSAs. Another marker of myositis refractory to treatment is anti-SPR (signal recognition protein), primarily found in JPM girls (18%) who are African-American. Anti-M2 (NuRD) is directed against the 218/240 kDa helicase family proteins and is associated with the hallmark cutaneous findings in JDM patients who are predominantly Hispanic and have a milder disease course. Anti-CADM-140 (MDA-5) is common in amyopathic DM patients (i.e. skin findings but no evidence of muscle involvement) who frequently have rapidly progressive ILD, cutaneous ulcerations, and palmar pustules, and who, in Japan, have a high mortality rate. Anti-SAE (small ubiquitin-like modifier activating enzyme) has been identified in children with JDM and adults with skin symptoms preceding muscle involvement and a low frequency of ILD.


Myositis Associated Antibodies


Myositis associated antibodies (MAAs) are primarily found in adults and in children with overlap myositis as well as in JPM and JDM and include the anti-U1-RNP, anti-Ro, anti-PM-Scl, anti-Ku specific antibodies as well as other, less frequently identified MAAs ( Table 42.6 ). Of note, newer, more sensitive methods of antibody detection have indicated that sera containing minute amounts of antibodies to Ro, La, RNP, and Sm can functionally activate the IFN-α pathway in children with JDM, suggesting that these antibodies may actively participate in perpetuating disease pathology. Finally, a large group of children are negative for known MSA or MAA antibodies, and may be either ANA positive or ANA negative. In addition, myositis antibody negative sera may contain precipitin bands against as-yet-unidentified proteins.


Type 1 Interferons


Current concepts in this fast-moving field concerning the driving force of the pediatric inflammatory myopathies center on the massively upregulated type1 interferon response, first identified in muscle of untreated children with active symptoms of JDM of all disease durations as similar to a massive antiviral response ; for example MXA (myxovirus A) was upregulated 96-fold ( Figure 42.8A ). Further study showed the disease duration did impact on many other aspects of gene expression, including vascular remodeling ( Figure 42.8B ). This type 1 driven interferon response was confirmed by others, both in children and in muscle from adults with DM and PM, but not in adults with IBM. There are at least three major types of interferons: type 1 (interferon-α and interferon-β), type 2 (IFN-γ), and type 3, including interferon-λ—which may antagonize IFN-α activity. Recent evidence in adults with DM has identified IFN-β as a strong component of their inflammatory response. Peripheral blood from children and adults with DM was tested which displayed specific upregulation of type 1 interferon induced genes and their proteins. However, not all the upregulated genes in the untreated muscle were expressed in mRNA from the same child with JDM’s peripheral blood mononuclear cells (PBMCs) obtained at diagnosis. MxA mRNA expression in JDM PBMC obtained at the initial clinic visit was elevated compared to controls and was positively correlated with Disease Activity Score (DAS) for muscle, but not with DAS for skin, suggesting that damage to skin and muscle in JDM may each have a discrete pathophysiology. Decrease in muscle symptoms was associated with a decrease in PBMC MxA mRNA expression. As type 1 interferons induce many genes including MXA , a phase I trial of a monoclonal antibody to type 1 interferon-α was executed, which documented both improved patient symptoms and diminished expression of type 1 interferon genes. The role of TNFα is presented in Figure 42.9 . A proposed scheme that integrates some of the environmental and genetic factors in the development of JDM is presented in Figure 42.10 .




Figure 42.8


Gene expression (GE) profile heat maps for type 1 interferon induced genes, comparing data from MRI directed muscle biopsies from ( A ) untreated children with definite symptoms JDM of 2 months or less duration, with ( B ) GE profiles from muscle biopsies of untreated children with JDM symptoms>2 months’ duration. B . Gene tree created by hierarchical clustering based on gene expression profiles. Upregulated genes were clustered into clusters C, D, and F, while downregulated genes were clustered into clusters A, B, and E, in girls with long (≥2 mo) vs. short (<2 mo) duration of active disease. The color codes represent the ratio between each of the JDM groups compared with the age- and sex-matched control samples. The complete list of genes in clusters A–F can be found in supplemental table 1 of Chen et al. Genes involved in vasculature and extracellular matrix remodeling, which is part of angiogenesis, were differentially expressed between patients with long and short duration of active disease. Interestingly, most of these genes cluster in cluster F, in which genes were downregulated more in patients with short duration of the disease (<2 mo). This finding suggests preferential misregulation of the expression of these specific genes in the early phase of the inflammatory disease process.

Reproduced with permission from Chen et al. (2008).



Figure 42.9


The association of a long duration of untreated disease and the TNFα-308A allele with a chronic disease course in white children with JDM. ( A ) Increased frequency of the TNFα-308A allele in white children with JDM compared with controls. ( B ) Both a long time period of untreated disease (>9 months before therapy given) and the TNFα-308A allele contribute to a subsequent chronic disease course in JDM. The DQA1*0501 allele, while increased in children with JDM, appears to be associated with disease susceptibility, not disease chronicity. ( C ) A chronic disease course is associated with increased production of TNFα by peripheral blood mononuclear cells. ( D ) The muscle fiber expression of TNFα on histochemistry in the diagnostic muscle biopsies from untreated JDM is increased in children with the TNFα-308 A allele. Surprisingly, more muscle fibers than macrophages are positive for TNFα, suggesting that this may be an important mechanism of localization of tissue damage.

Reproduced with permission from Pachman et al. (2001).



Figure 42.10


A proposed scheme for the pathophysiology of JDM. The Major Histocompatibility Locus, DR3 *0301 may serve as a disease susceptibility locus for microbial or other antigenic interactions, while the TNFα-308A allele is associated with increased production of TSP-1 which may play a role in vessel occlusion, as well as a chronic disease course which includes increased pathological calcifications.


T Cell Immunity


Children with active untreated JDM may be lymphopenic, and the duration of untreated disease may influence the lymphocyte phenotype. Studies of the cellular composition in the muscle of children with JDM identified CD4, CD8 positive cells and macrophages as well as mast cells—which were more prominent in matched skin samples. Perifasicular atrophy, one of the hallmarks of juvenile dermatomyositis, is characterized by infiltration of CD4+ T cells (dendritic cells) as well as the destruction of micro capillaries which contribute to muscle cell death. The native CD4+ T cells as well as plasmacytoid dendritic cells, which are also CD4 positive, are active producers of IFN-α (see the previous discussion). A possible local source of IFN-β may be immature muscle cells, which appear to be the target of activated DCs expressing IL-1 and IL-23, implicated in T cell polarization. In turn, the local production of IFN-β after TLR-3 activation in the presence of the Th1 cytokine IFN-γ may explain HLA class I overexpression by muscle fibers themselves—a hallmark of inflammatory myopathy.


Complement


A few studies that included children demonstrated complement activation: the membrane attack complex was localized to the intramuscular microvasculature in 10 of 12 patients and was correlated with the duration of the clinical disease. Complement components and immune complexes appear to participate in the pathophysiology of JDM with evidence of complement activation (C3d) accompanied by increased levels of fibrinopeptide A and vWF:Ag, which are synthesized by and released from damaged endothelial cells (see the following). In children with JDM, decreased levels of C4 protein are directly associated with fewer than normal 4A gene copy number, apparently in linkage disequilibrium with the “autoimmune locus” identified by the GWAS studies.


Cytokines and Chemokines


Investigation of this fast-changing field documents the complex interrelationships of the cytokine network. Many of the type 1 interferon inducible cytokine genes proceed through the NFkB pathway and are pro-inflammatory in nature. A longitudinal investigation of mRNA from adults with DM for analysis of the cytokine-induced gene expression for the signaling pathways of type 1 interferon, TNFα, IL-1β, granulocyte-monocyte colony-stimulating factor, IL-10, and IL-13 demonstrated only a type 1 interferon signature score in 21/24 patients. Similarly, the analysis of children with JDM (MSA specificity not given), showed upregulation of type 1 interferon induced cytokines ; analysis of the cytokine pattern was presented as a potential biomarker for disease activity. There is an increased frequency of the A allele in the TNFα-308 promoter region compared with age-matched controls ( Figure 42.9A ), which is associated with increased production of TNFα protein by peripheral blood mononuclear cells as well as the muscle fibers themselves in untreated children with JDM.


Neopterin


Neopterin is an indicator of macrophage activation and is increased in the serum of untreated children with JDM. Neopterin, a member of the pteridine family, is derived from guanosine triphosphate via guanosine triphosphate cyclohydrolase and is released from macrophages as a consequence of T-cell-dependent interactions involving interferon-γ (IFN-γ). Neopterin levels correlated with the clinical disease activity score in more than 65% of cases. The neopterin levels were not associated with peripheral blood mononuclear cell production of IFN-γ in vitro but with the number of macrophages in the muscle biopsy of the child with JDM; they both decrease in response to appropriate therapy. Measurement of urinary neopterin (first voided specimen) showed a good correlation with disease activity.


Acute Phase Reactants


In uncomplicated JM, the usual indicators of an acute-phase reaction, ESR and C-reactive protein, are often within normal limits, even in those children with ILD. However, ESR and C-reactive protein may be elevated in pediatric patients with severe active disease or infected sites of calcinosis, as well as in children with overlap syndromes, where these acute phase reactants are associated arthritis. Of note, if the child is given intravenous IgG therapy, an elevated ESR may be observed. The lymphopenia is not commonly accompanied by an abnormal platelet count, although mild microcytic anemia may be present.


Von Willebrand Factor Antigen


A sensitive indicator of vasculopathy, an elevated von Willebrand factor antigen (vWF:Ag) may precede a disease flare (when muscle enzyme data are normal), or it may remain elevated after the enzymes have normalized. Of note, the normal range for this antigen is blood group dependent: people with blood group “O” have lower normal ranges (36–157%) than those associated with other blood group antigens, such as blood group B, for which the normal range is 57% to 241%. In about 65% of children with JDM, levels of vWF:Ag are highly associated with a clinical score of disease activity (DAS). These clinical indicators of disease activity—the MRI findings, neopterin, and vWF:Ag levels—are easily available, but are imperfect guides to therapy. It is anticipated that accurate measurements of pro-inflammatory immune markers will help characterize the severity of the immunologically mediated inflammatory process, and become more accessible in the future.


Bone Health


Vitamin D Levels


The few studies reporting data from children with JDM at diagnosis comment that the levels of 25-hydroxy vitamin D are often below normal ranges. These low values are obtained before the children are strongly urged to limit sun exposure, for UVB will exacerbate not only the JDM rash on the exposed skin, but may trigger a full-blown flare in the inflammatory myopathy. It is important to obtain a vitamin D blood level at diagnosis so that appropriate supplementation with 2- or 3-hydroxy-vitamin D can begin concomitantly with the start of corticosteroids, along with supplemental doses of calcium.


Bone Mineral Density


Decreased bone density, frequent in untreated JDM, is measured by DXA, using a “Z score” normalized for age-matched healthy children, and is associated with a longer duration of untreated disease, depressed serum osteocalcin, and 25-hydroxy vitamin D insufficiency. This constellation of events places the child at risk for fracture prior to the initiation of glucocorticoid therapy, and this is escalated by the use of corticosteroids. Of note, untreated children with JDM have an elevated RANKL:OPG ratio at the time of diagnosis, resulting in expansion of the number of osteoclasts and activation of the bone resorptive function. This may lead to suppression of normal bone mineral accretion and a subsequent reduction in the lumbar spine bone mineral density (BMD) Z score. Therefore, children with a longer duration of untreated JDM may have lower than normal lumbar spine BMD Z scores. This risk is further augmented by steroid treatment, which impairs calcium absorption, and the child’s relative decrease in mobility because of weakness, which promotes calcium mobilization from bone.




Course and Therapy


Disease Course


The outcome of JDM has greatly improved since the 1960s when one third of the children died, one third were crippled, and one third recovered. Several types of disease courses have been described—monocyclic, recurrent, and continuous —but it may be that the categorization is, in fact, more dichotomous: a single episode of symptoms vs. disease chronicity. The frequency of calcinosis has decreased from <60% of cases to about 20–30% at diagnosis ( Box 42.1 ). Estimates of the average disease duration of JDM vary from 1.5 years to persistent disease over the child’s lifespan, and this remains a critical therapeutic problem, when the child appears to have recovered from the initial acute onset of the disease. Follow-up studies of children with myositis cannot yet reflect our current aggressive therapy at diagnosis. For example, one study (mean follow-up, 7.2 years) of 65 children documented calcinosis in 34% (unrelated to gender, therapy, or disease course); 35% were still taking medication 3 or more years after diagnosis; 23% had weakness; 40% still had rash. The concept that a persistent rash predisposes the child to pathologic calcifications was confirmed by other investigators. Late disease recurrence after years of apparent remission has been reported, suggesting the need for periodic monitoring with more sensitive indicators of disease activity. It is difficult to predict outcome at the onset of illness, although the magnitude of the initial serum levels of CK appeared to be a direct correlate of disease severity at the time of observation, and the MSA characteristics may provide some indication of course. Several investigators noted that prognosis is directly related to the degree of vascular involvement. Comparison of Korean adults with DM and children with JDM showed that the children had more involvement of neck musculature, had calcifications, and had a better outcome. It is not uncommon for children with JDM to develop another autoimmune disease, such as psoriasis.


Evaluation Tools for Disease Activity and Chronicity


Until recently, there has been little discussion of the development of methods that could distinguish the features of an active inflammatory process from its residue. Although it is clinically held that the active inflammatory process completely resolves in about 33% of the children with JPM and JDM, there are few data, either clinical or histologic, to support this conjecture. One of the operational problems is the lack of standardized tools to assess the child with myopathy before and after therapy, but a funded study is underway to develop an international consensus among experts across the disciplines to evaluate, establish, and validate the criteria for improvement or regression of disease. Furthermore, the impact of the age of the child at disease onset on both the pathophysiology of the disease as well as the degree of potential reversibility has not been defined. In order to address these methodological gaps, several groups have developed criteria for evaluation of the efficacy of therapeutic intervention. A measure of well-being and function, the Child’s Health Assessment Questionnaire (CHAQ), initially developed for children with JRA, was tested in children with JDM and identified the extent of disease-associated disability. The CHAQ was found to be reliable in identifying moderate to severe disability in children with JDM. A second larger investigation (115 patients) confirmed these findings and documented the “floor effect” of the CHAQ, that is, that the CHAQ does not accurately determine JDM disease activity (skin involvement, vasculitis) in children who had relatively good muscle function. Most children with inflammatory myopathy have an initial gratifying response to corticosteroids, but these children may continue to have symptoms not related to muscle strength, such as persistence of rash and evidence of vasculopathy. In another study, global assessment, comparing a Likert scale and a visual analogue scale, was used by physician, parent, and patient to determine disease activity and damage indices in 117 children with inflammatory myopathies. These measures appear to be more sensitive to change earlier in the child’s illness because there is more rapid change in physical findings at that time, but they are less useful later in the disease course. An evaluation of muscle function, the CMAS, was recently developed and also includes tests of muscle strength as well as tests of muscle endurance for children 4 years of age and older. The total score from this test was positively associated also with assessment of the impact of the disease, the manual muscle test, and the serum CK level. Of note, the CMAS has evolved with minor modifications, and is a useful and fairly reproducible tool. Inasmuch as 25% of children with JDM are 4 years of age or younger at diagnosis, it was important to establish that even healthy children age 4 cannot perform three of the effort dependent maneuvers which are needed to document that the younger child with JDM has fully recovered. A validated method for the rapid assessment of disease activity (the disease activity score, DAS) was developed combining the physician evaluation of the extent and severity of involvement of skin, muscle strength, and function with an estimate of small vessel inflammation as reflected in the palate and nailfold capillaries, and can be used in a child with JDM of any age. This global DAS has been adopted as one of the methods on an international basis to assess disease activity in a child with JDM before and after therapy in addition to the CMAS and manual muscle test. A battery of validated tools to evaluate disease chronicity has been developed and is now commonly employed. There is agreement that chronic damage occurs in some children who have been diagnosed with JDM, for example, persistent painless joint contractures associated with fibrosis; the spectrum of partial lipoatrophy, lipodystrophy, and eventual insulin-resistant diabetes; hypertension; and cardiac disease. Less clear is the status of pathologic calcifications, which can resolve on a rare occasion without therapy or after suppression of chronic inflammation. It is anticipated that with the use of targeted assessment tools, more specific information will be obtained to evaluate the efficacy of not only our current therapies but also new interventions that will be more specifically directed at the pathophysiology of the disease.


Therapy


Development of Consensus Driven Guidelines for Classification and Therapy


Specific investigation of the efficacy of the type, duration, and route of medication administered to children with JDM and the other less common inflammatory myopathies is a continuous process. Several consensus meetings developed guidelines for treatment: one for moderately severe JDM, another for moderate JDM. A concerted assessment of the use of corticosteroids in IIM, as there is in asthma, has not yet been organized. Several factors may confound the existing data. Active vasculopathy, which is often not evaluated, may inhibit oral absorption of the drug compared with the intravenous route. The age of the child may alter drug metabolism, once absorption has occurred, so that younger children may require a higher dose per square meter than their adult counterparts—which has been proven for methotrexate in children, and may apply to other forms of medication. Specific recommendations for therapy have to be considered in this context but are limited because of the lack of uniform assessment of age-dependent patient response during therapy. In addition, although there are some clues as to immunological specificity, these indicators of immunologic activity have not been widely tested to confirm that they have normalized at some point after therapy has been initiated.


Oral Corticosteroids


In general, the treatment of mild uncomplicated JDM consisted of oral prednisone (1 to 2 mg/kg) in conjunction with gastric protection ( Table 42.8 ). The child’s linear growth is arrested temporarily if the oral dosage of steroids exceeds 4 mg/m 2 . Stunted growth combined with increased weight gain and IIM-associated decreased mobility proves a challenge for the body-image conscious child. For the adolescent, the increased weight gain and the development of steroid-induced acne when taking oral prednisone frequently pose problems with compliance, which can be circumvented to some extent by the intravenous administration of methylprednisolone. In children with microvascular damage which also occurs in the gastrointestinal tract, absorption of oral prednisone is impaired, providing the rationale for the use of high-dose intermittent methylprednisolone, as shown in Figure 42.11 , in which the absorption of 50 mg of prednisolone IV vs. PO was compared in children with JDM and decreased nailfold capillary end row loops.


Jun 25, 2019 | Posted by in NEUROLOGY | Comments Off on Juvenile Dermatomyositis and Other Inflammatory Myopathies in Children

Full access? Get Clinical Tree

Get Clinical Tree app for offline access