• Non ci sono risultati.

6 Dermatomyositis Jeffrey P. Callen

N/A
N/A
Protected

Academic year: 2022

Condividi "6 Dermatomyositis Jeffrey P. Callen"

Copied!
16
0
0

Testo completo

(1)

Jeffrey P. Callen

Introduction

Dermatomyositis (DM) is one of the idiopathic inflammatory myopathies (IIM) (Callen 2000; Plotz et al. 1995; Targoff 1991). In 1975, Bohan and Peter pub- lished a classical article that first suggested a set of criteria to aid in the diag- nosis and classification of DM and polymyositis (PM). Four of the five criteria related to the muscle disease: 1) progressive proximal symmetrical weak- ness 2) elevated muscles enzymes, 3) an abnormal electromyogram and 4) an abnormal muscle biopsy, while the fifth was the presence of compatible cutaneous disease. It was felt that DM differed from PM only by the presence of cutaneous disease. Recent studies of the pathogenesis of the myopathy have been controversial, some suggesting that the myopathies in DM and PM are pathogenetically different with DM being due to a vascular inflamma- tion (Kuru et al. 2000) while other studies of cytokines suggest that the pro- cesses are similar (Wanchu et al. 1999; Shimizu et al. 2000; Sugiura et al. 2000;

Nyberg et al. 2000). There has been a renewed interest in the pathogenetic mechanisms involved in the myopathy with recent studies revealing abnor- mal levels of nitric oxide, elevation of circulating tumor necrosis factor recep- tors, elevated soluble CD40 expression, and increased expression of interleukin 1-alpha within the muscle. The pathogenesis of the cutaneous disease is poorly understood.

Classification and Clinical Appearance

Classification

Bohan and Peter (1975) suggested five subsets of myositis – DM, PM, myositis

with cancer, childhood DM/PM, and myositis overlapping with another col-

lagen-vascular disorder. In a subsequent publication, Bohan et al. (1977) noted

(2)

that cutaneous disease may precede the development of the myopathy, how- ever it was only recently recognized that another subset of patients with dis- ease that only affects the skin (amyopathic dermatomyositis) [ADM] or DM-sine myositis may occur (Euwer and Sontheimer 1991). A seventh subset known as inclusion body myositis (IBM) has more recently been recognized (Sayers et al. 1992). Perhaps there is an eighth group in which characteristic cutaneous disease is drug-induced (Dourmishev and Dourmishev 1999). Finally, Son- theimer has proposed another subset that classifies patients as hypomyopa- thic dermatomyositis when skin disease is present with subtle muscle disease evident with studies other than enzymatic analysis and a post-myopathic DM for those patients in whom the myositic component of the disease resolves while the skin disease remains active (Sontheimer 2002).

Cutaneous Manifestations

The characteristic and possibly pathognomonic cutaneous features of DM are the heliotrope rash and Gottron’s papules. The heliotrope rash consists of a violaceous to dusky erythematous rash with or without edema in a symmetri- cal distribution involving periorbital skin (Fig. 1). Sometimes this sign is quite subtle and may involve only a mild discoloration along the eyelid margin. A heliotrope rash is rarely observed in lupus erythematosus (LE) and sclero- derma. Occasionally patients may be felt to have angioedema or dermatitis.

Gottron’s papules are found over bony prominences, particularly the meta- carpophalangeal joints, the proximal interphalangeal joints, and/or the distal interphalangeal joints (Fig. 2). They may also be found overlying the elbows, knees and/or feet. The lesions consist of slightly elevated, violaceous papules and plaques. There may be a slight scale and on some occasions there is a

Fig. 1.Facial erythema with periorbital edema are present in this woman with dermatomyositis.

The periorbital changes represent the heliotrope eruption

(3)

thick psoriasiform scale. Within the lesions, there is often telangiectasia. These lesions may be clinically confused with lesions of LE, or, at times, with papu- losquamous disorders, such as psoriasis or lichen planus. Routine histopa- thological evaluation will aid in the differentiation from psoriasis of lichen planus, but cannot reliably distinguish the cutaneous lesions of DM from those of LE.

Several other cutaneous features are characteristic of the disease despite not being pathognomonic. They include malar erythema, poikiloderma in a photosensitive distribution, violaceous erythema on the extensor surfaces, and periungual and cuticular changes. Nailfold changes consist of periungual telangiectasia and/or a characteristic cuticular change with hypertrophy of the cuticle and small, hemorrhagic infarcts with this hypertrophic area. Peri- ungual telangiectasia may be clinically apparent or may be appreciated only by capillary microscopy. Poikiloderma (the combination of atrophy, dyspig- mentation and telangiectasia) may occur on exposed skin such as the extensor surfaces of the arm, the ‘V’ of the neck (Fig. 3) or the upper back (Shawl sign).

Patients rarely complain of photosensitivity, despite the prominent photodis-

Fig. 2.Gottron’s papules. Erythematous scaly plaques are present on the dorsal hands, particularly over the bonfy prominences (MCP, PIP and DIP joints). There is also marked

periungual telangiectasia and cuticular overgrowth

(4)

tribution of the rash. This photosensitive poikilodermatous eruption may be difficult to differentiate from LE. Facial erythema may also occur in DM. This change must be differentiated from LE, rosacea, seborrheic dermatitis or atopic dermatitis. Scalp involvement in dermatomyositis is relatively common and is manifest by an erythematous to violaceous, psoriasiform dermatitis (Kasteler and Callen 1994). Clinical distinction from seborrheic dermatitis or psoriasis is occasionally difficult, but histopathologic evaluation is helpful. Nonscar- ring alopecia may occur in some patients and often follows a flare of the sys- temic disease. Lastly, recent reports have detailed the finding of gingival telangiectasia (Ghali et al. 1999) and angiokeratomas (Shannon and Ford 1999) in children with DM.

DM-sine myositis, also known as amyopathic DM (ADM), is diagnosed in patients with typical cutaneous disease in whom there is no evidence of mus- cle weakness and who repeatedly have normal serum muscle enzyme levels (Bohan et al. 1977; Rockerbie et al. 1989; Stonecipher 1993; Cosnes et al. 1995).

Some patients with ‘ADM’ when studied will have an abnormal ultrasound, magnetic resonance imaging or muscle biopsy. These patients have muscle involvement, and may be better classified as hypomyopathic DM (Sonthei- mer 2002). Since many of the ADM patients are not evaluated beyond clini- cal and enzymatic studies, many feel that the ADM patient represents a systemic process requiring systemic therapies. There are also a fair number of patients whose myositis resolves following therapy, but whose skin disease

Fig. 3.Poikiloderma of the upper chest in a patient with dermatomyositis

(5)

remains as an active, important feature of the disease. These patients are called Post-myopathic DM, and the skin is their major and often only mani- festation of the disease. There is also a small subset of patients who never develop myositis, despite having prominent cutaneous changes, and it is these patients who can be classified as amyopathic DM assuming that they have not received systemic corticosteroids or immunosuppressive agents.

Patients with DM are at times difficult to distinguish from patients with subacute cutaneous lupus erythematosus (SCLE). The lesions of DM differ slightly in their distribution, occurring more over bony prominences, and they are frequently accompanied by severe pruritus; whereas LE lesions tend to occur between the knuckles and are usually asymptomatic. Routine skin bi- opsy is not helpful in the distinction between LE and DM. Immunofluores- cence microscopy (IF) should be negative in DM and positive in LE, however, about 50% of SCLE patients have a negative IF, and IF may be falsely posi- tive on sun-exposed skin. Serologic testing is also imperfect, since only 25%

of DM patients are Mi2 positive, and on a single testing only 60–70% of SCLE patients are Ro (SS-A) antibody positive.

The skin lesions of DM are probably photoaggravated despite the lack of symptoms suggestive of photosensitivity reported by patients (Cheong et al.

1994). Clinical observations suggest that not only is the skin disease exacer- bated by light, but muscle disease may be worsened after sun exposure (Woo et al. 1985; Callen 1993; Zuber et al. 1996; Callen 1999). However, phototesting has not been able to reliably reproduce the skin lesions, thus, the wave- length of light that is responsible for the clinical manifestations (action spec- trum) is not known.

Rare cutaneous manifestations include vesiculobullous lesions (McCollough and Cockerell 1998), an eruption that simulates pityriasis rubra pilaris (Requena et al. 1997), a flagellate erythema (Nousari et al. 1999), vasculitis, erosive le- sions, as well as an exfoliative erythroderma. In small case series it has been suggested that some of these cutaneous manifestations may be more com- mon in patients with an associated malignancy.

Skin lesions of DM may precede the development of myopathy and may persist well after the control and quiescence of the myositis. Patients skin le- sions may flare with sun exposure, but only some of these patients will have a flare of their muscle involvement. Thus, in many instances the course of the skin lesions does not parallel that of the muscle disease.

A variety of other cutaneous lesions have been described in patients with

DM or PM that do not reflect the interface changes observed histopathologi-

cally with the pathognomonic or characteristic lesions. These include pannicu-

litis, plaque-like mucinosis (Fig. 4) (Abernethy et al. 1996; Kaufman et al. 1998),

a flagellate eruption (Nousari et al. 1999), urticaria, as well as changes of hyper-

keratosis of the palms known as mechanics hands. Lastly, children with DM

may develop calcinosis, but in addition insulin-resistance and lipodystrophy

has recently been reported as a relatively common complication despite ade-

quate therapy (Heumer et al 2001).

(6)

Muscle Disease

Clinical and laboratory abnormalities suggestive of muscle disease are char- acteristic features of DM (Wortmann 2000). The myopathy primarily affects the proximal muscles, is usually symmetrical and is slowly progressive over a period of weeks to months. Initial complaints including myalgias, fatigue, or weakness manifest as an inability to climb stairs, to raise the arms for actions like hair grooming or shaving, to rise from a squatting or sitting position, or a combination of these features. Tenderness upon palpation of the muscles is variable. An inability to swallow and symptoms of aspiration may reflect the involvement of striated muscle of the pharynx or upper esophagus. Dysphagia or dysphonia generally signifies a rapidly progressive course and may be as- sociated with poor prognosis.

Systemic Features

Dermatomyositis is a multisystem disorder (Spiera and Kagen 1998). Arthral- gias and/or arthritis may be present in up to one fourth of patients with in- flammatory myopathy. The usual picture is one of generalized arthralgias

Fig. 4.Papular mucinosis-like lesions in a patient with dermatomyositis. Prominent papular lesions are present within the poikilodermatous eruption. Histopathology revealed deposition

of massive amounts of mucin in the dermis

(7)

accompanied by morning stiffness. The small joints of the hands, wrists, and ankles may be involved with a symmetrical nondeforming arthritis.

Esophageal disease as manifested by dysphagia is estimated to be pres- ent in 15% to 50% of patients with inflammatory myopathy. The dysphagia can be of two types: proximal dysphagia or distal dysphagia. Proximal dyspha- gia is caused by involvement of striated muscle in the pharynx or proximal esophagus. This involvement correlates well with the severity of the muscle disease and is steroid-responsive. Distal dysphagia is related to involvement of nonstriated muscle and appears to be more frequent in patients who have an overlap with scleroderma or another collagen vascular disorder. Dyspha- gia is associated with a poor prognosis and correlates with the presence of pulmonary involvement.

Pulmonary disease occurs in dermatomyositis and polymyositis in ap- proximately 15% to 30% of patients (Marie et al. 1998). Interstitial pneumo- nitis is a primary process observed in DM/PM. It is more frequent in patients with esophageal involvement. Lung disease may also occur as a direct com- plication of the muscle disease, such as hypoventilation or aspiration in pa- tients with dysphagia, or may be a result of treatment, as with opportunistic infections or drug-induced hypersensitivity pneumonitis. In a retrospective review of 70 patients with myositis associated interstitial lung disease seen at Mayo Clinic between 1990 and 1998, most of the patients presented with ei- ther symptoms of lung disease or symptoms of myositis alone, with only 15 in whom the involvement occurred simultaneously (Douglas et al 2001). In gen- eral, the lung disease was at first felt to be a pneumonitis that was antibiotic resistant. Biopsy of the lung revealed non-specific interstitial pneumonitis or diffuse alveolar damage in a majority of those who were biopsied. Only two patients had bronchiolitis obliterans organizing pneumonia (BOOP). It is un- clear how many of the patients had dermatomyositis, but perhaps between 8 and 12. Therapy included corticosteroids with or without an immunosup- pressive agent and the prognosis is poorer for these patients than unselected patients with myositis as demonstrated by a 5-year survival of 60.4%. Pa- tients with Jo-1 antibodies (19 of 50 who were tested) had roughly the same features and prognosis as those who did not have this antibody.

Clinically symptomatic cardiac involvement in patients with DM or PM is uncommon, but when present it is associated with a poor prognosis (Gonzalez- Lopez et al. 1996). Various abnormalities have been described which include conduction defects, and rhythm disturbances primarily. Although congestive heart failure, pericarditis, and valvular disease may occur, they are much less frequent. Depending on the report, cardiac manifestations may occur in up to 50% of patients, but only a small proportion of these patients manifest symptoms. It is not known whether the identification of asymptomatic abnor- malities has an effect on long-term outcome, or even if the findings are more prevalent in DM/PM than in an age-matched control group.

Calcinosis of the skin or muscle is unusual in adults but may occur in up to

40% of children or adolescents with DM. Calcinosis cutis is manifested by firm,

(8)

yellow or flesh-colored nodules, often over bony prominences. Occasionally, these nodules can extrude through the surface of the skin, in which case sec- ondary infection may occur. Calcification of the muscles is often asymptom- atic and may be seen only on radiological examination. In severe forms, the calcinosis can cause loss of function, and, rarely bone formation is possible.

Myositis and Malignancy

The relationship of DM-polymyositis to malignancy has been recently clari- fied (Hill et al. 2001). The reported frequency of malignancy in dermatomyo- sitis has varied from 6% to 60% with most large population-based cohort studies revealing a frequency of about 20–25%.

Several Scandinavian studies have documented the increased frequency of malignancy in DM over the general population (Siguregeirsson et al. 1992;

Chow et al. 1995; Ario et al. 1995; Hill et al. 2001). While polymyositis pa- tients had a slight increase in cancer frequency; it was not highly significant and could be explained by a more aggressive cancer search (known as diag- nostic suspicion bias). These studies have not dealt with the amyopathic DM subset, but data from Mayo Clinic suggests that these patients may also have an associated malignancy (el-Azhary and Pakzad 2002). Malignancies may occur prior to the onset of myositis, concurrently with myositis, or after the onset of DM. In addition, the myositis may follow the course of the malignancy (a paraneoplastic course) or may follow its own course independent of the treatment of the malignancy. Studies demonstrating benefits of cancer sur- gery on myositis as well as those showing no relationship of the myositis to the malignancy have been reported.

A wide variety of malignancies have been reported in patients with DM.

Gynecologic malignancy, in particular ovarian carcinoma may be overrepre- sented in DM (Whitmore et al. 1994; Hill et al. 2001). Asians with DM are of- ten found to have nasopharyngeal cancer (Peng et al. 1995). In the recent analysis of combined data from Scandinavia, Hill et al. (2001) again noted the increased association of ovarian cancer, but also noted increases in lung, pancreatic, stomach, colorectal cancer and non-Hodgkin’s lymphoma. Ma- lignancy is more common in older patients (> 50 years) (Marie et al. 1998;

Pautas et al. 2000), but reports of young adults and rarely even children with

DM have appeared, suggesting that age alone should not dissuade the phy-

sician from a careful evaluation (see below). The site of malignancy can be

predicted by the patient’s age (e.g., malignancy in a young man is more of-

ten testicular cancer, whereas, in an elderly male, colon or prostate cancer

would be more common). In the past, there was concern about whether the

use of immunosuppressive therapies would predispose the patient to an ex-

cess cancer risk. This has not proven to be the case with most cancers being

reported within the first three years following diagnosis.

(9)

Juvenile (Childhood) Dermatomyositis (JDMS)

DM is much more common than polymyositis in children and adolescents (Pachman 1995). A fulminant course may be present, but most often the on- set is indolent and children are first thought to have viral infections or der- matitis. Delayed diagnosis is more common in the non-white population. Often JDMS is characterized as a vasculitis, but the major difference of JDMS from adult DM is the greater potential for calcinosis. A recent report detailed the chronic nature of this disease in children, with many patients requiring ther- apy to suppress their disease activity more than three years after diagnosis (Huber et al. 2000). In addition, it was noted that the development of calcinosis was not related to initial therapy, but was associated with a lower score on an assessment instrument of physical function. Pachman et al. (2000) linked the presence of calcinosis and a prolonged course of disease with TNF-308A al- lele in their patients with juvenile DM.

Drug-Induced Dermatomyositis

The etiology of most cases of DM is unknown, however, in a small number of patients, the cutaneous manifestations are due to, or are exacerbated by drugs.

This has been best documented for hydroxyurea in which de-challenges and re-challenges have been performed (Daoud et al. 1997; Marie et al. 2000, Dacey and Callen 2003). However, quinidine, non-steroidal anti-inflammatory drugs, D-penicillamine, and HMG-CoA-reductase inhibitors have also been linked on occasion to DM (Dourmishev and Dourmishev 1999).

Diagnosis and Evaluation of the Patient with Dermatomyositis

The diagnosis of DM is suspected in patients with clinically compatible cuta- neous findings. Exclusion of other possible cutaneous conditions is aided by skin biopsy and the recognition of muscle involvement. In the absence of iden- tifiable myopathy, the differentiation from cutaneous LE may be difficult.

Muscle weakness may be caused by many other disorders including toxins, infections, metabolic abnormalities, and neurologic disorders (Wortmann 2000).

However, the presence of characteristic skin lesions allows the diagnosis to be more firmly established.

Muscle involvement is suspected clinically. Enzymatic testing will reveal

elevations of creatine kinase, aldolase, lactic dehydrogenase or ALT. The CK

seems most specific and most widely available and is a useful test for follow-

ing response to therapy. Additional testing including electromyography (EMG),

(10)

muscle biopsy, ultrasound or MR imaging may be ordered in patients in whom other tests are inconclusive (Garcia 2000).

Serologic tests are often ordered, but their clinical application is at best controversial. Antinuclear antibody testing is frequently positive in DM pa- tients. Several myositis-specific antibodies (MSAs) have been recognized and correlate with certain subsets (Love et al. 1991). Most MSAs are described in PM patients and will not be further discussed here. Anti-Jo-1 antibody is predictive of pulmonary involvement, but rarely occurs in DM patients.

Anti-Mi-2 occurs in roughly 25% of DM patients and although specific for DM, it is not sensitive. Recently Targoff et al. (2000) described a new anti- body to a 155 kd antigen or Se antigen (90–95 kd) that appears to be a marker of amyopathic DM (16 of 18 patients studied). The anti-155 kd antibody may also be associated with juvenile DM and might predict a chronic course.

Anti-Ro (SS-A) antibody may occur rarely. Perhaps as further studies are performed, serologic testing will become clinically useful. Until then, these tests are primarily reserved for investigation.

Once the diagnosis is confirmed, the patient should have a thorough eval- uation. Evaluation has several purposes: assessment of severity, prediction of prognosis and identification of associated disorders. The severity of the myositis often correlates with enzyme levels and degree of weakness. Pa- tients should be assessed for esophageal, pulmonary, and cardiac involvement with tests such as a barium swallow and/or esophageal motility studies, chest x-ray, pulmonary function studies including diffusion studies, and an elec- trocardiogram.

An evaluation of malignancy should be considered in all adult patients with DM (Callen 1982, Callen 2002). The type of evaluation is selected based upon the patient’s age and sex. The likelihood of malignancy increases with age and the sites vary depending on the patient’s age. Malignancy evaluation is repeated with new symptoms or annually for the first three-years following diagnosis. The over-representation of cancer in these patients seemingly ap- proaches normal levels after three-years (Hill et al. 2001), thus, age-specific malignancy screening, along with evaluation of any abnormal symptoms or findings, are recommended for following patients more than three years af- ter the initial diagnosis.

Course and Therapy

Several general measures are helpful in treating patients with DM. Bedrest

is often valuable in the individual with progressive weakness; however, this

must be combined with a range-of-motion exercise program to prevent con-

tractures. Patients who have evidence of dysphagia should have the head of

their bed elevated and should avoid eating meals immediately before retiring.

(11)

Corticosteroids and Immunosuppressive Agents

The mainstay of therapy for DM is the use of systemic corticosteroids. Tradi- tionally, prednisone is given at a dose of 0.5 to 1 mg/kg daily as initial ther- apy. The treatment should continue for at least one month after the myositis has become clinically and enzymatically inactive. At this point, the dose is slowly tapered, generally over a period lasting one and a half to two times as long as the period of active treatment. Approximately 25% of patients with dermatomyositis will not respond to systemic corticosteroids, another 25–50%

will develop significant steroid-related side effects. Therefore, early inter- vention with steroid-sparing agents primarily immunosuppressive agents, such as methotrexate, azathioprine, cyclophosphamide, mycophenolate mofetil, chlo- rambucil, or cyclosporin may be an effective means of inducing or maintain- ing a remission (Sinoway and Callen 1993; Villalba and Adams 1996; Gelber et al. 2000; Vencovsky et al. 2000). Roughly one-half to three fourths of pa- tients treated with an immunosuppressive agent will respond with an in- crease in strength, a decrease in enzyme levels, or a reduction in corticosteroid dosage. However, there few double-blind, placebo-controlled studies that demonstrate the effectiveness of any of these agents.

Additional Therapeutic Options

Patients who fail to respond to these immunosuppressives may respond to pulse methylprednisolone therapy (Callen et al. 1994; Sawhney et al. 2000;

Al-Mayouf et al. 2000a) combination immunosuppressive therapy (Villalba et al. 1998), etanercept (Saadeh 2000), infliximab (Hengstman et al. 2000) or total body irradiation. Early enthusiasm for plasmapheresis was followed by a placebo-controlled study that failed to demonstrate effectiveness. (Miller et al. 1992). However, in a double-blind, placebo-controlled study, Dalakas and co-workers (1993) demonstrated the benefits of high dose intravenous immune globulin for recalcitrant DM. Further open-label studies have dem- onstrated similar results (Cherin et al 2002).

Therapy of cutaneous disease in patients with DM is often difficult be-

cause even though the myositis may respond to treatment with corticosteroids

and/or immunosuppressive drugs, the cutaneous lesions often persist. Al-

though cutaneous disease may be of minor importance in patients with seri-

ous fulminant myositis, in many patients, cutaneous disease becomes the

most important aspect of their disorder. Most patients with cutaneous lesions

are photosensitive; thus, the daily use of a broad-spectrum sunscreen with a

high sun protective factor is recommended. Topical therapy with an appro-

priately selected corticosteroid or a non-steroidal immunomodulators such

as tacrolimus or pimecrolimus may be useful adjunctive therapy (Hollar and

Jorizzo 2004). Hydroxychloroquine HCL in dosages of 200 mg to 400 mg per

day is effective in approximately 80% of patients treated as a steroid-sparing

(12)

agent (Woo et al. 1984). Patients who do not respond well to hydroxychloro- quine can be switched to chloroquine phosphate 250 to 500 mg per day or can have quinacrine HCL 100 mg twice daily added to the regimen. Patients on continuous antimalarial therapy should have periodic ophthalmologic ex- aminations and blood counts. It appears that patients with DM have a greater frequency of drug eruptions from antimalarials, thus patients should be warned about this possibility (Pelle and Callen 2002).

Methotrexate in doses of 15–35 mg per week has been reported to be use- ful for skin lesions of DM (Zieglschmid et al. 1995; Kasteler and Callen 1997).

These studies, however, are uncontrolled, open-label observations. The need for routine liver biopsy in the DM patient treated with methotrexate is con- troversial, but patients who are obese, diabetic, or have abnormal liver func- tion tests should probably have periodic liver biopsies.

Other immunosuppressive agents have not been systematically studied as treatment of cutaneous lesions of DM, but some anecdotes suggest that mycophenolate mofetil might be beneficial. Lastly, intravenous immune glob- ulin administered monthly can result in clearing of patients with cutaneous lesions.

Calcinosis is a complication of disease in children and adolescents. This process may be prevented by aggressive early treatment. Preliminary analy- sis of the use of intravenous methylprednisolone suggested that this therapy might lessen the frequency and severity of this process (Callen et al. 1994).

Others have suggested that immunosuppressives may similarly reduce the chance of calcinosis (Al-Mayouf et al. 2000b). Once established, calcinosis is difficult to treat. Although possible, spontaneous regression is unusual. Indi- vidual patients have been treated with low-dose warfarin or oral aluminum hydroxide, however, no studies have documented the usefulness in larger groups of patients. Recent reports of long-term administration of diltiazem are promising (Oliveri et al. 1996).

The prognosis of dermatomyositis varies greatly, depending on the series of patients studied. Factors that affect prognosis include the patient’s age, the severity of myositis, the presence of dysphagia, the presence of cardio- pulmonary involvement, the presence of an associated malignancy, and the response to corticosteroid therapy (Marie et al. 1999). It seems to be well established by retrospective reports that the use of corticosteroids and/or immu- nosuppressive therapies improves the prognosis.

Summary

DM is a condition primarily of the skin and muscles, but other systemic fea-

tures may occur. DM appears to be the predominant myopathy in children,

whereas in adults many patients without skin disease occur (polymyositis or

inclusion body myositis). The pathogenesis of the muscle disease is becoming

(13)

better understood, but the cutaneous disease mechanisms remain enigmatic.

DM in adults is associated with malignancy and thus a careful evaluation of each patient should be part of their initial and followup assessments. Patients should also be evaluated for the presence of esophageal, pulmonary and car- diac disease. Calcinosis is more frequent in children with DM, and early ag- gressive therapy may limit the chance of this complication. Corticosteroids, immunosuppressives, biologic agents and/or immune globulin are effective therapies for the myopathy of DM, whereas the skin disease is best mana- ged with sunprotection, topical corticosteroids, antimalarials, methotrexate and/or immune globulin. The prognosis is good except for patients with ma- lignancy, those with severe weakness and those with cardiopulmonary dys- function.

References

Abernethy ML, Arterberry JF, Callen JP (1996) Low-dose methotrexate as an adjunctive ther- apy with surgery for ectropion complicating dermatomyositis. Dermatology 192(2): 153–155 Al-Mayouf SM, Laxer RM, Schneider R, Silverman ED, Feldman BM (2000a) Intravenous im- munoglobulin therapy for juvenile dermatomyositis: efficacy and safety. J Rheumatol 27:

2498–2503

Al-Mayouf S, Al-Mazyed A, Bahabri S (2000b) Efficacy of early treatment of severe juvenile dermatomyositis with intravenous methylprednisolone and methotrexate. Clin Rheu- matol 19: 138–141

Airio A, Pukkala E, Isomäki (1995) Elevated cancer incidence in patients with dermatomyosi- tis: A population based study. J Rheumatol 22: 1300–1303

Bohan A, Peter JB (1975) Polymyositis and dermatomyositis. N Engl J Med 292: 344–347 and 403–407 (two part article)

Bohan A, Peter JB, Bowman RL, Pearson CM (1977) A computer-assisted analysis of 153 pa- tients with polymyositis and dermatomyositis. Medicine 56: 255–286

Callen AM, Pachman LM, Hayford J, Chung A, Ramsey-Goldman R (1994) Intermittent high dose intravenous methylprednisolone (IV pulse) prevents calcinosis and shortens disease course in juvenile dermatomyositis (JDMS). Arth Rheum 37: R10A

Callen JP (1982) The value of malignancy evaluation in patients with dermatomyositis. J Am Acad Dermatol 6: 253–259

Callen JP (1993) Photodermatitis in a 6-year-old child. Arhthritis Rheum 36: 1483–1485 Callen JP (1999) Photosensitivity in collagen vascular diseases. Semin Cutan Med Surg 18(4):

293–296

Callen JP (2000) Dermatomyositis. Lancet 355: 53–57

Callen JP (2000) When and how should the patient with dermatomyositis or amyopathic der- matomyositis be assessed for possible cancer? Arch Dermatol 138: 969-971

Cheong W-K, Hughes GRV, Norris PG, Hawk JLM (1994) Cutaneous photosensitivity in dermatomyositis. Brit J Dermatol 131: 205–208

Cherin P, Pelletier S, Teixeira A, Laforet P, Genereau T, Simon A, Maisonobe T, Eymard B, Herson S (2002): Results and long-term followup of intravenous immunoglobulin infu- sions in chronic, refractory polymyositis. Arthritis Rheum 46: 467-74.

Chow WH, Gridley G, Mellemkjaer L, McLaughlin JK, Olsen JH, Fraumeni JF Jr (1995) Can- cer risk following polymyositis and dermatomyositis: A nationwide cohort study in Den- mark. Cancer Causes and Control 5: 9–13

Cosnes A, Amaudric F, Gherardi R, Verroust J, Wechsler J, Revuz J, Roujeau JC (1995) Dermatomyositis without muscle weakness. Long-term follow-up of 12 patients without systemic corticosteroids. Arch Dermatol 131: 1381–1385

(14)

Dacey MJ, Callen JP (2003) Hydroxyurea-induced dermatomyositis-like eruption. J Am Acad Dermatol 48: 439-441.

Dalakas MC, Illa I, Dambrosia JM, Soueidan SA, Stein DP, Otero C, Dinsmore ST, McCrosky S (1993) A controlled trial of high-dose intravenous immune globulin infusions as treat- ment for dermatomyositis. N Engl J Med 329: 1993–2000

Dalakas MC (1998) Molecular immunology and genetics of inflammatory muscle diseases.

Arch Neurol 54: 1509–1512

Daoud MS, Gibson LE, Pittelkow MR (1997) Hydroxyurea dermopathy. A unique lichenoid eruption complicating long-term therapy with hydroxyurea. J Am Acad Dermatol 36:

178–182

Douglas WW, Tazezlaer HD, Hartman TE, Hartman RP, Decker PA, Schroeder DR, Ryu JH (2001) Polymyositis-dermatomyositis-associated interstitial lung disease. Am J Respir Crit Care Med. 164: 1182-5.

Dourmishev AL, Dourshimev LA (1999) Dermatomyositis and drugs. Adv Exp Med Biol 445:

187–191

El-Azhary RA, Pakzad SY (2002) Amyopathic dermatomyositis: retrospective review of 37 cases. J Am Acad Dermatol 46: 560-565

Euwer RL, Sontheimer RD (1991) Amyopathic DM (DM siné myositis). J Am Acad Dermatol 24: 959–966

Garcia J (2000) MRI in inflammatory myopathies. Skeletal Radiol 29: 425–438

Gelber AC, Nousari HC, Wigley FM (2000) Mycophenolate mofetil in the treatment of severe skin manifestations of dermatomyositis: a series of 4 cases. J Rheumatol 27: 1542–1545 Ghali FE, Stein LD, Fine J-D, Burkes EJ, McCauliffe DP (1999) Gingival telangiectases. An

underappreciated physical sign of juvenile dermatomyositis. Arch Dermatol 135:

1370–1374

Gonzalez-Lopez L, Gamez-Nava JI, Sanchez L, Rosas E, Suarez-Almazor M, Cardona-Munoz C, Ramos-Remus C (1996) Cardiac manifestations in dermato-polymyositis. Clin Exp Rheumatol 14: 373–379

Hengstman G, van den Hoogen F, van Engelen B, Barrera P, Netea M, van den Putte L (2000) Anti-TNF-blockade with infliximab (remicade) in polymyositis and dermatomyositis. Ar- thritis Rheum 43: S193A

Hill CL, Zhang Y, Sigurgeirsson B, Pukkala E, Mellemkjaer L, Airio A, Evan SR, Felson DT (2001) Frequency of specific cancer types in dermatomyositis and polymyositis: A popula- tion-based study. Lancet 357: 96–100

Huemer C, Kitson H, Malleson PN, Sanderson S, Huemer M, Cabral DA, Chanoine J-P, Petty RE (2001) Lipodystrophy in patients with juvenile dermatomyositis – evaluation of clinical and metabolic abnormalities. J Rheumatol 28: 610-615.

Hollar CB, Jorizzo JL (2004) Topical tacrolimus 0.1% ointment for refractory skin disease in dermatomyositis: a pilot study.

J Dermatolog Treat 15: 35-9.

Huber AM, Lang B, LeBlanc CM, Birdi N, Bolaria RK, Malleson P (2000) Medium- and long-tern functional outcomes in a multicenter cohort of children with juvenile dermato- myositis. Arthritis Rheum 43: 541–549

Kasteler JS, Callen JP (1994) Scalp involvement in dermatomyositis. Often overlooked or misdiagnosed. JAMA 272: 1939–1941

Kasteler JS, Callen JP (1997) Low-dose methotrexate administered weekly in an effective corticosteroid-sparing agent for the treatment of the cutaneous manifestations of derma- tomyositis. J Am Acad Dermatol 36: 67–71

Kaufmann R, Greiner D, Schmidt P, Wolter M (1998) Dermatomyositis presenting as pla- que-like mucinosis. Brit J Dermatol 138: 889–892

Kuru S, Inukai A, Liang Y, Doyu M, Takano A, Sobue G (2000) Tumor necrosis factor-a ex- pression in muscles of polymyositis and dermatomyositis. Acta Neuropathol 99: 585–588 Love LA, Leff RL, Fraser DD, Targoff IN, Dalakas M, Plotz PH, Miller FW (1991) A new ap- proach to the classification of idiopathic inflammatory myopathy: Myositis-specific auto- antibodies define useful homogeneous patient groups. Medicine 70(6): 360–374 Lundberg IE, Nyberg P (1998) New developments in the role of cytokines and chemokines in

inflammatory myopathies.Curr Opinion in Rheumatol 10: 521–529

(15)

Marie I, Hatron P-Y, Hachulla E, Wallaert B, Michon-Pasturel U, Devulder B (1998) Pulmo- nary involvement in polymyositis and in dermatomyositis. J Rheumatol 25: 1336–1343 Marie I, Hatron PY, Levesque H, Hachulla E, Hellot MF, Michon-Pasturel U, Courtois H,

Devulder B (1999) Influence of age on characteristics of polymyositis and dermatomyositis in adults. Medicine 78: 139–147

Marie I, Joly P, Levesque H, Heron F, Courville P, Cailleux N, Courtois H (2000) Pseudo-der- matomyositis as a complication of hydroxyurea therapy. Clin Exp Rheumatol 18: 536–537 McCollough ML, Cockerell CJ (1998) Vesiculo-Bullous Dermatomyositis. Am J Dermato-

pathol 20: 170–174

Miller FW, Leitman SF, Cronin ME, Hicks JE, Leff RL, Wesley R, Fraser DD, Dalakas M, Plotz PH (1992) Controlled trial of plasma exchange and leukapheresis in polymyositis and dermatomyositis. N Engl J Med 326: 1380–1384

Nousari HC, Ha VT, Laman SD, Provost TT, Tausk FA (1999) ‘Centripetal Flagellate Ery- thema’: A Cutaneous Manifestation Associated with Dermatomyositis. J Rheumatol 26:

692–695

Nyberg P, Wikman A-L, Nennesmo I, Lundberg I (2000) Increased expression of Interleukin 1a and MHC class I in muscle tissue of patients with chronic, inactive polymyositis and dermatomyositis. J Rheumatol 27: 940–948

Oliveri MB, Palermo R, Mautalen C, Hübscher O (1996) Regression of calcinosis during Diltiazem treatment of juvenile dermatomyositis. J Rheumatol 23: 2152–2155

Pachman LM (1995) An update on juvenile dermatomyositis. Curr Opin Rheumatol 7:

437–441

Pachman LM, Liotta-Davis MR, Hong DK, Kinsella TR, Mendez EP, Kinder JM, Chen EH (2000) TNFa-308A allele in juvenile dermatomyositis. Arthritis Rheum 43: 2368–2377 Pautas E, Cherin P, Piette J-C, Pelletier S, Wechsler B, Cabane J, Herson S (2000) Features of

polymyositis and dermatomyositis in the elderly: A case-control study. Clin Exp Rheu- matol 18: 241–244

Pelle MT, Callen JP (2002) Adverse cutaneous reactions to hydroxychloroquine are more common in patients with dermatomyositis than in patients with cutaneous lupus erythe- matosus. Arch Dermatol 138: 1231-1233

Peng J-C, Sheem T-S, Hsu M-M (1995) Nasopharyngeal carcinoma with dermatomyositis.

Analysis of 12 cases. Arch Otolaryngol Head Neck Surg 121: 1298–1301

Plotz PH, Rider LG, Targoff IN, Raben N, O’Hanlon TP, Miller FW (1995) Myositis: Immuno- logic Contributions to Understanding Cause, Pathogenesis, and Therapy. Ann Intern Med 122: 715–724

Requena L, Grilli R, Soriano L, Escalonilla P, Farina C, Martin L (1997) Dermatomyositis with a pityriasis rubra pilaris-like eruption: a little-known distinctive cutaneous manifestation of dermatomyositis. Brit J Dermatol 136: 768–771

Rockerbie NR, Woo TY, Callen JP, Giustina T (1989) Cutaneous changes of dermatomyositis precede muscle weakness. J Am Acad Dermatol 20: 629–632

Saadeh CK (2000) Etanercept is effective in the treatment of polymyositis/dermatomyositis which is refractory to conventional therapy including steroids and other disease modify- ing agents. Arhtritis Rheum 43: S193A

Sawhney S, Sidoti G, Woo P, Murray KJ (2000) Clinical characteristics and outcome of idio- pathic inflammatory myositis (IIM) in childhood: A 2 year follow up. Arthritis Rheum 43: A Sayers ME, Chou SM, Calabrese LH (1992) Inclusion body myositis: Analysis of 32 cases. J

Rheumatol 19: 1385–1389

Shannon PL, Ford MJ (1999) Angiokeratomas in juvenile dermatomyositis. Pediatr Dermatol 16: 448–451

Shimizu T, Tomita Y, Son K, Nishinarita S, Sawada S, Horie T (2000) Elevation of serum solu- ble tumor necrosis factor receptors in patients with polymyositis and dermatomyositis.

Clin Rheumatol 19: 352–359

Sontheimer RD (2002) Would a new name hasten the acceptance of amyopathic dermatomyo- sitis (dermatomyositis sine myositis) as a distinctive subset within the idiopathic inflam- matory dermatomyopathies spectrum of clinical illness? J Am Acad Dermatol 46: 626-636.

Spiera R, Kagen L (1998) Extramuscular manifestations in idiopathic inflammatory myopa- thies. Curr Opinion in Rheumatol 10: 556–561

(16)

Siguregeirsson B, Lindelöf B, Edhag O, Allander E (1992) Risk of cancer in patients with dermatomyositis or polymyositis. N Engl J Med 325: 363–367

Sinoway PA, Callen JP (1993) Chlorambucil: An Effective Corticosteroid-Sparing Agent for Patients with Recalcitrant Dermatomyositis. Arthritis Rheum 36: 319–324

Sugugiura T, Kawaguchi Y, Harigai M, Takagi K, Ohta S, Fukasawa C, Hara M, Kamatani N (2000) Increased CD40 expression on muscle cells of polymyositis and dermatomyositis:

Role of CD40-CD40 ligand interaction in IL-6, IL-8, IL-5, and monocyte chemoattractant protein-1 production. J Immunol 164: 6593–6000

Stonecipher MR, Jorizzo JL, White WL, Walker FO, Prichard E (1993) Cutaneous changes of dermatomyositis in patients with normal muscle enzymes: DM siné myositis? J Am Acad Dermatol 28: 951–956

Targoff IN (1991) Dermatomyositis and polymyositis. Curr Prob Dermatol 3: 131–180 Targoff IN, Trieu EP, Sontheimer RD (2000) Autoantibodies to 155 kd and Se antigens in pa-

tients with clinically-amyopathic dermatomyositis. Arthritis Rheum 43: S194A

Vencovsky J, Jarosova K, Machacek S, Studynkova J, Kafvova J, Bartunkova J, Nemcova D, Charvat F (2000) Cyclosporine A versus methotrexate in the treatment of polymyositis and dermatomyositis. Scand J Rheumatol 29: 95–102

Villalba L, Adams EM (1996) Update on therapy for refractory dermatomyositis and polymyo- sitis. Curr Opinion Rheumatol 8: 544–551

Villalba L, Hicks JE, Adams EM, Sherman JB, Gourley MF, Leff RL, Thornton BC, Burgess SH, Plotz PH, Miller FW (1998) Treatment of refractory myositis. A randomized crossover study of two new cytotoxic regimens. Arthritis Rheum 41(3): 392–399

Wanchu A, Khullar M, Sud A, Bambery P (1999) Nitric oxide production is increased in pa- tient with inflammatory myositis. Nitric Oxide 3: 454–458

Whitmore SE, Rosenshein NB, Provost TT (1994) Ovarian cancer in patients with dermato- myositis. Medicine 73: 153–160

Woo TY, Callen JP, Voorhees JJ, Bickers DR, Hanno R, Hawkins C (1984) Cutaneous lesions of dermatomyositis are improved by hydroxychloroquine. J Am Acad Dermatol 10:

592–600

Woo TR, Rasmussen J, Callen JP (1985) Recurrent photosensitive dermatitis preceding juve- nile dermatomyositis. Pediatr Dermatol 2: 207–212

Wortmann RL (2001) Idiopathic Inflammatory diseases of muscle. In Treatment of the Rheu- matic Diseases. Weisman ML, Weinblatt ME, Louie JS (eds) W. B. Saunders Co. pp 390–402

Zieglschmid-Adams ME, Pandya AG, Cohen SB, Sontheimer RD (1995) Treatment of derma- tomyositis with methotrexate. J Am Acad Dermatol 32: 754–757

Zuber M, John S, Pfreundschuh M, Gause A (1996) A young woman with a photosensitive pruritic rash on her face and upper trunk. Arthritis Rheum 39: 1419–1422

Riferimenti

Documenti correlati

CECs, circulating endothelial cells; EPCs, endothelial pro- genitor cells; BS, Behçet syndrome; HC, healthy controls; AAV, anti-neutrophil cytoplasmic antibody-associated vas-

[r]

In these conditions, it was obviously necessary to perform a first orthograde approach, in order to remove the endodontic material used previously and disinfect the root canal,

IL LIBERO CONVINCIMENTO DEL GIUDICE NEL PROCESSO PENALE ED IN PARTICOLAR MODO NELLA VALUTAZIONE DELLA PROVA SCIENTIFICA.. Dalle prove legali

Questo “filone di pensiero” si incarna in grandi nomi – alcuni dei quali scomparsi, altri ancora attivi che si può avere il privilegio di incontrare per strada o, persino, chiamare

esperienze di collaborazione scientifica con alcune università e centri di ricerca in cina | multiple experiences in scientific collaboration with universities and research

In order, to represent the temporal vagueness in its definition, not only the topo- logical primitives representing the precedence relation among phases have been redefined using

Storch, Induced neural stem cells (iNSCs) in neurodegenerative diseases. Han, D.W., et al., Direct reprogramming of fibroblasts into neural stem cells by defined