<<

4.2 Progressive Systemic

Nicolas Hunzelmann and Thomas Krieg

Introduction

Systemic sclerosis (SSc) belongs to the group of “diffuse inflammatory con- nective tissue diseases” or “ vascular diseases” comprising a variety of severe, sometimes life-threatening systemic diseases which often have a chronic, debilitating course. SSc is characterized by the involvement of the and various internal organs (e.g. , , ). The inflammatory and fibrotic process destroys the normal architecture of the affected organs leading to dysfunction and failure. The disease activity is highly variable and often unpredictable. The severity of the disease process in SSc leads to a re- duced lifespan, impaired mobility and loss of autonomy. SSc mainly evolves along pathological changes of the vascular system, the and of the extracellular matrix including its major cell type, the . The resulting leading to and failure of the af- fected organs largely determines the outcome of the disease process. How- ever, despite intense research efforts the relationship and interaction between the pathophysiological processes affecting the vascular system, the immune system and the extracellular matrix are only incompletely understood.

Immune Dysregulation

One of the hallmarks of SSc is a perturbed immunoregulation (resulting in the presence of ), which appears to be influenced by additional factors such as genetic and exogenous factors. Autoimmunity in SSc is char- acterized by HLA gene restricted responses against nuclear and nucleolar antigens. The mechanisms inducing the antibody production are unknown but clinical associations with autoantibody specificities suggest that these antigen-restricted responses are involved in disease specific . These autoimmune phenomena are in a not well understood way related to the inflammatory process with lymphocytic perivascular infiltrates in the skin 166 Nicolas Hunzelmann and Thomas Krieg and lung evident early on in the disease process and preceding the develop- ment of fibrosis. The similarity of the condition with some aspects of graft versus host disease has frequently been noted. Recently it was suggested that , i.e. the persistence of foetal cells in the maternal bone marrow and other organs like the skin might be a risk factor, also explaining the female excess (Artlett et al. 1998; Evans et al. 1999). However, subsequent studies found similar frequencies of microchimerisms compared to normal controls but nevertheless an increased number of microchimeric fetal cells in patients (Burastero et al 2003).

Vascular Pathology

The relationship between autoimmune responses and the vascular pathol- ogy is unclear, as Raynaud’s syndrome and vascular abnormalities may be evident many years prior to the onset of disease (Blockmans et al. 1996). The morphological changes that can be observed on a ultrastructural level, i.e. basement membrane thickening, intimal hyperplasia and inflammatory cell infiltration have been interpreted as a sign indicating microvascular injury as a primary event in this disease (Prescott et al. 1992). Depending on the study population and statistical methodology, between 5 and 20% of all individuals presenting with Raynaud’s phenomenon are re- ported to subsequently develop SSc. A constellation of additional signs and symptoms indicative of microvascular damage separates SSc patients from others presenting with Raynaud’s phenomenon. These include nailfold capil- laroscopic changes (Maricq et al. 1980), hand/foot edema, digital ulcers, cal- cifications and teleangiectasia. The combination of a fibrotic microvascular and hyperreactive vasoconstrictor status is thought to represent the primary lesion responsible for the vasospastic episodes. Tissue hypoxia normally in- duces new blood vessel growth by induction of a variety of angiogenic fac- tors. In SSc, loss of capillaries is a typical and early disease manifestation which has been related to an increase in angiostatic factors and programmed endo- thelial cell death (apoptosis) where a number of possible mechanisms have been proposed (Sgonc et al. 1996; Kahaleh and Fan 1997; Hebbar et al, 2000). A recent study suggests that latent infection contributes to the known phenomenon of endothelial cell cytotoxicity of scleroderma se- rum by identifying IgG autoantibodies that bind a cytomegalovirus and induce apoptosis in human endothelial cells (Lunardi et al. 2000).

Dysregulation of Extracellular Matrix Synthesis

The dysregulation of extracellular matrix synthesis is the third major patho- physiologic change, with the extent and progression of the fibrotic process Progressive 167 being important prognostic factors in the disease process. It has been well established by in situ hybridization and by fibroblast cultures obtained from involved tissue (e.g. skin or lung) that scleroderma display an ac- tivated phenotype producing increased amounts of various and expressing adhesion molecules such as ICAM-1 (LeRoy et al. 1974; Uitto et al. 1979; Scharffetter et al. 1988; Majewski et al. 1995). The newly synthe- sized extracellular matrix is deposited particularly around skin appendages and at the border of the to the subcutaneous tissue, partially replac- ing the latter (Perlish et al. 1985). The collagen bundles running parallel with the skin surface show swelling and variation in thickness. Although the bio- synthesis of collagens has been investigated in detail, its metabolism and turnover in vivo is not yet fully understood. In physiological situations in- volving increased collagen synthesis, as e.g. in wound healing, the amount of collagen in the tissue is obviously tightly controlled by its similarly increased degradation. Similarly, in a fibrotic disease, the net gain of collagens must thus involve a disturbed balance between the synthetic and degradative pro- cesses. The most commonly used approach to study collagen degradation is the study of collagen degrading enzymes (Herrmann et al. 1991; Mauch et al. 1998). However, the results are difficult to interprete in terms of the in vivo situation, as a combination of several enzymes including the corresponding inhibitors are likely to be involved in the degradation of a single collagen fi- ber. A different approach to this question is to study the degradation prod- ucts as they appear in vivo. Interestingly, we and others could demonstrate that increased levels of ICTP, a degradation product of cross-linked type I collagen, are common in patients with SSc (Heickendorff et al. 1995; Hunzel- mann et al. 1998b). They correlate well with the skin score, a commonly used indicator of the severity of the disease (Subcommittee of the ARA 1980; Kaha- leh et al. 1986). This indicates that the concentrations of circulating ICTP re- flect the type I collagen load in this disease. We found the highest values in patients with very active and extensive disease. Recently, a study on the uri- nary excretion of two mature cross-links of collagen, hydroxylysyl and lysyl pyridinoline, also suggested that in SSc more fibrillar collagens are degraded than in the normal state (Stone et al. 1995). Furthermore these crosslinks can usually only be detected in bone, suggesting that occurence of these crosslinks in the skin is related to the sclerotic process. Therefore these studies indicate that the increased deposition of type I collagen is accompanied by an increased turnover and altered crosslink formation of this molecule, indicating an even more complex derangement of synthetic and degradative processes in this disease than previously acknowledged. The factors which finally lead to the activated phenotype of scleroderma fibroblasts are not entirely clear. Several studies suggest the contribution of transforming growth factor-b (TGF-b) (Kulozik et al. 1990), a potent inducer of collagen synthesis, to the progression of skin sclerosis. This notion is fur- ther supported by the detection of connective tissue growth factor (CTGF) gene expression in skin biopsies of SSc patients (Igarashi et al. 1995) and SSc 168 Nicolas Hunzelmann and Thomas Krieg fibroblasts (Shi-Wen et al. 2000) as TGF-b is also known to induce CTGF. Recent studies indicate that a deficiency in SMAD-7 an inhibitory protein in the TGF-b signalling pathway is characteristic of scleroderma fibroblasts (Dong et al. 2002). The increased biosynthesis of collagen is accompanied by an el- evation of the steady-state mRNA levels in vitro and in vivo of about 1.5–2x which appears to be due to both increased transcriptional activation as well as increased mRNA stability (Eckes et al. 1996; Jimenez and Saitta, 2000).

Clinical Appearence/Classification

The incidence of SSc is reported to be 2–20/million population and the prev- alence 4–290/million population. Based on distinct clinical aspects and courses of the disease an internationally accepted classification was established with two forms: limited cutaneous SSc (lSSc) and diffuse cutaneous SSc (dSSc). The disease is much commoner in females than males for reasons that are not entirely clear with a female-to-male ratio of 3–9:1. There are some popu- lations at high risk as e.g. the Choctaw Indians from Oklahoma suggesting that genetic factors are critical. However, twin studies are inconclusive and familial aggregation is rare. Both forms, however, lead to life threatening involvement of internal organs and large areas of the skin and are associated with marked excess mortality. The quality of life is severely reduced and the patients require continous med- ical support. Institution of regular physical as early as possible to pre- vent loss of function is mandatory. Patient support groups play an important role in helping these patients to cope with their difficult psychosocial situa- tion. The two major clinical variants are distinguished primarily on the degree and extent of skin involvement. The term is used when fea- tures commonly encountered in other connective tissue diseases are present such as or systemic erythematosus. The hallmark of SSc is fibrosis of the skin resulting early on in oedema, of- ten the first indication of SSc skin involvement, which is followed by fibrotic induration and finally atrophy. Several skin scoring methodologies have been developed with the modified Rodnan skin score having the broadest distri- bution (Kahaleh et al, 1986; Furst et al. 1998). It is assessed by palpation of skin using a 0–3 scale (normal, mild, moderate or severe thickening) at sev- enteen areas. Following the skin score and the distribution of cutaneous induration is of major clinical importance, as patients with diffuse cutaneous scleroderma are much more likely to have significant heart and/or renal dis- ease than those with the limited form of SSc, furthermore early disability and premature mortality are observed in this group (Clements et al. 1990). The period when skin thickening is most rapidly is also a time in which decline in visceral function is most likely to occur (Seibold 1994) (Fig. 1). Progressive Systemic Scleroderma 169

Fig. 1. Longitudinal development of skin score in patients with diffuse systemic sclerosis (—) and limited systemic sclerosis (- - - -)

Diffuse Cutaneous SSc (dSSc) dSSc is characterized by distal and proximal extremity and truncal skin thick- ening. Usually the elbows and knees are considered the dividing line. Lead- ing symptoms of dSSC are: 1) onset of Raynaud’s syndrome within 1 year of onset of skin changes (puffy or hidebound); 2) presence of tendon friction rubs; 3) early and significant incidence of interstitial lung disease, oliguric renal failure, diffuse gastrointestinal disease, and myocardial involvement; 4) ab- sence of anti CENP-B antibodies; 5) nailfold capillary dilatation and capillary destruction; 6) anti-DNA-topoisomerase I antibodies (30%).

Limited Cutaneous SSc (lSSc)

Leading symptoms of lSSc are the following: 1) Raynaud’s syndrome for years (occasionally decades); 2) skin involvement limited to hands, face, and fore- arms (acral) or absent; 3) a significant late incidence of , with or without interstitial lung disease; 4) a high incidence of anti centromer (CENP-B) antibodies (70–80%); 5) dilated nailfold capillary loops, usually with- out capillary dropout; 6) skin calcifications and teleangiectasia particularly affecting the face and hands; 7) occasional late development of small bowel mal absorption. A recent study suggests that systemic sclerosis sine scleroderma is a sub- set of SSc which should be included into the spectrum of SSc with limited cu- taneous involvement and should not be considered as a distinct disorder (Poormoghim et al. 2000). Except for the absence of skin thickening, the group of patients with systemic sclerosis sine scleroderma had no significant 170 Nicolas Hunzelmann and Thomas Krieg differences in individual internal involvements, laboratory features, se- rum autoantibody type (e.g. anti-centromer) or survival rate compared with patients with lSSc. There was a tendency but no significant difference toward more pronounced pulmonary arterial hypertension and reduced carbon mon- oxide (< 70% of predicted).

Scleroderma Overlap Syndromes

The most common overlap syndromes are Mixed Connective Tissues Disease (MCTD), (PM-Scl-associated) and the Synthetase Syndrome (Jo 1-associated). MCTD and the Synthetase Syndrome are more extensively dealt with in chapter 7. Scleromyositis is a scleroderma/polymyositis or scleroderma/dermatomyo- sitis overlap disorder associated with antibodies directed to the nucleolar PM-Scl complex and associated with HLA-DR3 (Genth et al. 1990). In a re- cent study of 108 cases, 83% of patients had characteristic manifestations (Jablonska and Blasczyk 1999). These findings include Raynaud’s syndrome, scleroderma-like and -like cutaneous changes of the face and hands including hyperkeratotic changes on the fingers, and ar- thritis. Pulmonary involvement occurs in about 30 to 60% of the patients (Mar- guerie et al. 1992). This syndrome is also a rather common subtype in children as about one third of the reported cases in the study of Jablonska (1999) are children with a mean age of onset at nine years. The course of this overlap syndrome is rather benign and usually responds to small or moderate doses of corticosteroids.

Environmentally Related Scleroderma-Like Syndromes

A broad variety of environmental factors have been reported to induce scleroderma (reviewed in Straniero et al. 1989). However, with few excep- tions (contaminated tryptophan, contaminated rape seed oil, vinyl chloride, trichlorethylene) these cases are likely to represent random occurences. These scleroderma-like disorders often lack several features of SSc, in particular the autoimmune phenomena (autoantibody synthesis). In this respect, there has been much concern and publicity on the role of silicone (e.g. in the form of surgical implants) as a possible environmental factor for connective tissue diseases such as SSc, but despite several epidemiological studies, no link has been established (Janowsky et al. 2000). In contrast, several studies indicate that silica dust-associated scleroderma can not be distinguished from SSc in terms of antibody profile and pheno- type (Haustein and Anderegg 1998). This is supported by experimental data suggesting that silica dust induces pathophysiological events similar to SSc (Haustein and Anderegg 1998). Progressive Systemic Scleroderma 171

Diagnosis

As in many other chronic diseases, diagnosis is in general readily performed once the illness has fully developed. Due to the often smoldering disease on- set and the uncharacteristic changes ocurring early on in SSc, including Raynaud’s phenomenon, pain or swelling clinical diagnosis of SSc and the differentiation from other diffuse inflammatory connective tissue diseases (rheumatoid , systemic lupus erythemtosus, polymyositis) or disor- ders characterised by abnormal extracellular deposition (e.g. amyloidosis) may be difficult if not impossible. Although each of the diffuse inflammatory connective tissue diseases are clinically distinct entities, they share some general analogies and often display a high level of clinical variability result- ing not uncommonly in overlap syndromes which pose a particular diagnos- tic and therapeutic challenge. Investigations related to the autoimmune phenomena and the vascular changes (e.g. capillaroscopy) are therefore necessary to perform. In some cases, follow up of the patient over time will indicate whether the patient finally de- velops inflammatory connective tissue disease, overlap syndrome etc. or may experience remission of an early flare up of autoimmune phenomena reflect- ing undifferentiated connective tissue disease (Williams et al. 1999).

Autoantibody Profile

The identification of autoantibodies is helpful in establishing the correct di- agnosis, indicating the prognosis and providing a guide to treatment and fol- low up (Fritzler 1993). In several studies, more than 95% of patients show

Fig. 2. Antibody profile and clinical classification of progressive systemic sclerosis and overlap syndromes 172 Nicolas Hunzelmann and Thomas Krieg

Table 1. Clinical Characteristics of Autoantibodies Associated with Progressive Systemic Sclerosis

Antibody Antigen Comments Frequency

Scl-70 DNA-topoisomerase I increased risk for up to 70% of dSSc; tumors 10 to 20% of SSc

U1 RNP U1 small nuclear overlap syndrome ribonucleoprotein to SLE

Fibrillarin U3 RNP poor prognosis 10–20% of dSSc

RNA Polymerase I, III sub units of RNA 20% of dSSc polymerase centromere kinetochores, CENP- limited disease 60–80% of lSSc; A, B, C, E 15% of SSc

Th/To Rnase P limited disease 2%

PM-Scl nuclear protein- characteristic skin 15% of overlap complex changes syndromes

Ku Nucleolar overlap syndrome < 10% of overlap heterodimer syndromes

Jo1 histidyl-tRNA SSc/polymyositis 10% of overlap synthetase overlap syndromes

antinuclear antibodies (ANA) (Bunn et al. 1998), thus making the diagnosis of SSc in a patient without ANA quite unlikely. Although there is no anti- body which can be related to disease activity like the presence of anti-ds DNA antibodies in systemic , patient classification ac- cording to serologic subsets can be meaningful. Nearly 85% of patients can be associated to one of seven SSc related antibodies (Table 1) and each of these antibodies describes a subset which to a different degree has charac- teristic clinical manifestations (Fig. 2, 3).

Clinical Presentation

At clinical presentation, sclerodactily is present in about 95% of patients and Raynaud’s syndrome is present in about 90%. If none of these features (in- cluding ANA) is present, the patient is likely to have a disorder other than SSc. Differential diagnosis includes in particular the generalized form of localized scleroderma, , scleromyxoedema, scleroderma adultorum Buschke, amyloidosis, porphyria cutanea tarda and acrodermatitis chronica Progressive Systemic Scleroderma 173

Fig. 3. Clinical phenotype of progressive systemic sclerosis and overlap syndromes. A. Diffuse systemic scleroderma: diffuse sclerosis with hyperpigmentation of the trunk. B. with contractures and atrophy of the fingers. C. Cutaneous calcinosis. D. PM-Scl overlap syndrome: mechanic hands with of the fingers atrophicans in the inflammatory phase (Table 2) and rarely sclerodermiform genodermatoses. Furthermore, one has to be aware of the vast differential diagnoses of Raynaud’s phenomenon in the patients presenting in the initial phase of the disease.

Table 2. Scleroderma-Like Disorders

I. Sclerotic disorders Scleroderma adultorum Buschke Scleroderma diabeticorum Scleroderma amyloidosum Scleromyxedema Environmentally related scleroderma-like syndromes Graft versus host disease Porphyria cutanea tarda Acrodermatitis chronica atrophicans

II. Sclerodermiform genodermatoses Werner Syndrome Progeria Acrogeria/Metageria 174 Nicolas Hunzelmann and Thomas Krieg

Associated Diseases

Association of SSc with other autoimmune diseases is relatively common in- cluding primary biliary , Sjogren-syndrome (including the detection of anti-Ro/La antibodies) and the subset of overlap syndromes, dermatomyo- sitis and polymyositis.

Therapy

The treatment of SSc is challenging due to the complex disease process and the difficulty to specifically treat distinct subgroups of this relatively rare dis- ease. Thus in 1995, an American College of Comittee published guidelines for the conduct of clinical trials in SSc (White et al. 1995). To date, there is no proven effective disease modifying treatment of SSc. Neverthe- less, there have been significant breakthroughs in the treatment of several individual end-organ manifestations leading to improvements in patients longevity and quality of life. SSc poses a particular problem to the medical system due to the relative rarity of the disease requiring specialised care by the general practicioner. Therefore, diagnosis and care should be at least in part in the hands of specialists who have daily exposure to this disease and have access to a laboratory trained in autoimmune serology, to dermatohis- topathology, and modern diagnostic radiologic procedures (e.g. CT, MRT, angiography). Cooperation with different is often necessary to provide optimal care due to the nature of the disease affecting other organ systems than the skin (e.g. rheumatology, pulmonary , , ). Specialized care should be provided in a setting where the out- patient facilities have also access to hospital beds to ensure timely and ap- propriate treatment for patients presenting with exacerbation of their disease. which has access to treatment facilities to prevent loss of function is another prerequisite for these specialised facilities. Although there are single medical centers often linked to research centers which can provide care along the guidelines cited above they are usually not embed- ded into the medical system in a way that ensures access for patients not liv- ing in regions where these centers exist. Patient support groups which to date in part make up for these shortcomings should play an important role in communicating the special needs of these patients to society. Internet based resources will play an increasing role for the information of patients and recruitement for ongoing studies in the future (e.g. Http://www.sclero.org, www.Sklerodermie.info). Therefore an important future aim will be to develop competence and communication based networks which ensure participation of all levels involved in the care for these patients. Progressive Systemic Scleroderma 175

Immunosuppressive Agents

D-penicillamine seems to affect both collagen and the immune system thus making it an ideal candidate to treat SSc. Interestingly, it took until the mid 90s to start a double blind randomized trial to investigate the effect of D-peni- cillamine in SSc. Unfortunately, no difference was found between a dose of 62.5 mg and 750 mg D-penicillamine daily indicating lack of efficacy (Clements et al. 1999). Also photopheresis (extracorporeal photochemotherapy) that has shown promise in several uncontrolled studies (Rook et al. 1992), failed in a recent crossover study to demonstrate a favourable effect (Enomoto et al. 1999). Several smaller trials investigated the use of cyclosporin A, which unfortu- nately was associated with considerable toxicity, especially nephropathy (Denton et al. 1994). , although its toxicity profile in patients with SSc was better than exspected, produced inconsistent results in two controlled studies (van den Hoogen et al. 1996; Pope et al. 1998).

Antifibrotic Agents

Interferon-g is the most potent known to inhibit collagen synthesis. Several uncontrolled studies applying the cytokine over up to one year have been performed to investigate the potential role of interferon-g in the treat- ment of SSc showing no major effect on the disease course (Hunzelmann et al. 1997). Interferon-a has also been shown to inhibit collagen synthesis (Dun- can et al. 1995). However, a placebo controlled study of early diffuse SSc found no benefit for skin sclerosis and pulmonary function, but a greater mortality in the active treatment arm (Black et al. 1998). Relaxin is a pregnancy-related hormone that has tissue remodeling and antifibrotic effects. Relaxin has been tested in a phase 2 trial where in the low dose group a significant improve- ment of the modified Rodnan skin score was found (p = 0.049) wheras in the high dose group no such effect was seen (Seibold et al. 2000).

Organ-Specific

Skin Involvement

General measures include skin protection from cold and trauma, skin care with moistening creams, lymph drainage and active physiotherapy. Calcium channel blockers or angiotensin II receptor type 1 antagonists can be given to decrease symptoms of Raynaud’s syndrome (Dziaio et al. 1999). In severe cases of finger tip ulcerations and impending digit amputation, intravenous prostacycline analogues may be of value (Zachariae et al. 1996; Pope et al. 2000). New agents as antagonists or phosphodiesterase-inhibitors are still under investigation. Ectopic calcifications or calcinosis when compromising 176 Nicolas Hunzelmann and Thomas Krieg blood circulation or causing symptoms may be removed surgically or by the use of CO2-laser (Bottomly et al. 1996). Laser (i.e. argon or flashlamp pumped dye laser) therapy is the treatment of choice to remove teleangiectasias, which may also involve the mucosa. UV radiation (UVA1 or bath-PUVA) with small patients numbers in un- controlled studies has been reported to be beneficial. In localized scleroderma, evidence for the efficacy of UVA1 or bath-PUVA is increasing although no double blind prospective study is available (Kerscher et al. 1996). Recent stud- ies have shown that UVA irradiation alone, and more so in conjunction with photosensitizing agents, increases the expression, synthesis and activation of metalloproteinases. In addition, a variety of and soluble factors in vitro and in vivo are modulated by UVA and can affect connective tissue re- modeling (Scharffetter et al. 1991; Herrmann et al. 1993). Clinical and ultra- sound evaluation revealed that the sclerotic lesions disappeared or markedly improved during PUVA bath photochemotherapy in 13 of the 17 enrolled pa- tients within less than three months. We have additional experience in 14 pa- tients suffering from localized scleroderma who improved substantially from bath PUVA therapy as monitored by skin score, cutaneous elastometry and evaluation of skin thickness by ultrasound analysis (Hunzelmann et al. 1998b). In a recent publication, the therapeutic potential of UVA1 therapy has been evaluated in localized scleroderma (Stege et al. 1998). This study corroborates and extends previous observations that in vivo UVA1 irradiation exposure of healthy human skin is associated with the induction of interstitial collagen- ase RNA expression in situ which may play a role in the remodelling of the fibrotic connective tissue.

Musculosceletal Involvement

Musculosceletal involvement, and musculosceletal pain being the most frequent complaints, is common in scleroderma and may lead to sec- ondary fibromyalgia. Muscle weakness and some increase in serum creatine kinase levels are quite common. Inflammatory arthritis can occur but raises the suspicion of the presence of an overlap syndrome and only rarely results in mutilating arthritis. Corticosteroids should be avoided due to their long term side effects and association with nephropathy in higher doses (Steen et al. 1998). Non steroidal anti-inflammatory agents should also be prudently cho- sen due to their potential side effects on renal function, blood pressure and gastrointestinal function. The superiority of the use of cyclo-oxygenase 2 in- hibitors remains to be proven.

Renal Involvement

Acute renal crisis is a serious and potentially fatal SSc complication associ- ated with an acute reduction on cortical blood flow, hyperreninemia, hyper- Progressive Systemic Scleroderma 177 tension which occurs most likely in diffuse cutaneous scleroderma of less than four years duration. Thus regular control of blood pressure (at least twice a week) is recommended to detect acute renal involvement early on. Some pa- tients will progress to renal failure and dialysis or renal transplantation. Chronic renal involvement is asociated with a slowly progressive obliterative vasculo- pathy. Before the advent of ACE inhibitor therapy and other improvements in the management of advanced renal disease, survival for longer than 3–6 months was almost unknown. Particularly in acute renal crisis, ACE inhibi- tors are the mainstay of treatment significantly prolonging patient survival (Steen et al. 1990). Additional administration of intravenous may be considered. Nevertheless, prognosis of established renal crisis is still relatively poor with about one third of patients progressing to renal replace- ment therapy. Here, a five year kidney graft survival rate of 47% was re- ported comparable to that of patients with SLE (Chang and Spiera 1999).

Pulmonary Involvement

Pulmonary fibrosis in SSc affects to different degrees the parenchymal and the vascular system. In early disease, inflammatory alveolitis may precede and/or accompany interstitial fibrosis leading to loss of pulmonary function as evidenced by decreased diffusing capacity and vital capacity. Bronchoalveolar lavage (in experienced hands) and high resolution chest computertomogra- phy will help to determine the degree of . Several studies indi- cate that alveolitis can be treated with (White et al. 2000). Pulmonary hypertension may most prominently develop in patients with limited cutaneous scleroderma of long duration with relatively little intersti- tial disease determining the prognosis of these patients. Here infusion or in- halation of prostacycline analogues or endothelin antagonists may improve the outcome, which usually results in death within five years unless heart- lung transplantation is performed (Pigula et al. 1997; Badesch et al. 2000; Hoeper et al. 2000; Rubin et al, 2002).

Gastrointestinal Involvement

The is frequently involved with a frequency for the oe- sophagus in about 80%, the , and in about 40–70% (Sjogren 1996). The pathology is characterized both by atro- phy of the smooth muscles that line the gastrointestinal tract and involvement of the myenteric nerve plexus. Main symptoms associated are heartburn, eso- phageal dysfunction in the upper gastrointestinal tract and diarrhoea due to bacterial overgrowth, fetal incontinence in the distal tract. Prokinetics (e.g. octreotide) are of limited use in severe and recently, the prokine- tic cisapride has been withdrawn from the market due to associated cardiac arrythmias leading to death. Proton pump inhibitors and to a lesser extent 178 Nicolas Hunzelmann and Thomas Krieg

H2-blockers are effective in controlling reflux apart from typical conservative measures (no late meals etc.). Bacterial overgrowth and fungal infections (e.g. candida esophagitis) can be dealt with by intermittent anti- microbial therapy and antimycotics. Rarely, teleangiectasias may also be pres- ent on the mucosa representing a potential source of occult intestinal bleeding.

Cardiac Involvement

The nature and severity of cardiac disease depends on the extent of myocar- dial fibrosis, a primary component of this disorder, and on the extent to which concurrent fibrosis of the lung and thickening and fibrosis of the small pulmonary place an additional burden on the circulation. Large per- fusion abnormalities on thallium scans are predictive of shortened survival and an increased number of cardiac events (Steen et al. 1996). Also, intermit- tent vascular ischemia is observed which probably reflects similar pathophy- siological changes as observed in the peripheral vasculature (Raynaud’s syndrome). Arrhythmias are quite common in SSc but seldom meet the defi- nition of severe arrhythmia.

Novel Therapeutic Perspectives

A number of studies are currently performed or planned to investigate the effect of new therapeutic concepts. Due to the relative rarity of these dis- eases and distinct subgroups, however, national and international coopera- tive studies will be required to prove the efficacy of these new approaches. The role of collagen as an autoantigen in SSc is not entirely proven. How- ever, in a recent study on the use of oral administration of bovine collagen a reduction of reactivity to human collagen was found accompanied by significantly improved skin thickness score and carbon monoxide diffusing capacity (McKown et al. 2000). Recombinant technology gives now rise to the development of recombi- nant cytokines and anti-cytokines that may have therapeutic potential, the principle of anti-TNF-alpha therapy in being the most prominent example. TGF-beta as a potent stimulus of collagen synthesis is thought to drive the fibrotic process. Recent studies in a mouse model (McCor- mick et al. 1999) and in human glaucoma in the human indicate a potential for the use of TGF-beta antibodies. Studies have been recently initiated to evaluate the beneficial effect of autologous haemopoetic stem cell transplantation in systemic lupus erythe- matosus and SSc. Previous studies showed that patients with who undergo bone marrow transplant for haemopoietic or other ma- lignancy are frequently noted to experience a remission of their autoimmune disease (Clements et al. 1997; Tyndall et al. 1997). However, to date only un- controlled studies have been reported with divergent effects. Progressive Systemic Scleroderma 179

Summary

Despite intense research efforts and major advances in the understanding of particular aspects of the disease process, the etiology of SSc is still unknown and the pathogenesis only partly understood. Thus the concept that SSc evolves along pathological changes of the vascular system, the immune sys- tem and of the extracellular matrix has not significantly changed over the last twenty years. Nevertheless, survival has markedly improved over the past two decades, with a 5-year survival rate over 80%, although to date there is no proven effective disease-modifying treatment of SSc. This is due to significant breakthroughs in the treatment of several individual end-organ manifestations. Current therapies of SSc rely mainly on drugs directed to the major skin and visceral complications such as Raynaud’s phenomenon, digital ulceration, nephropathy, gastrointestinal and pulmonary involvement. Although the introduction of drugs that treat these complications has changed mark- edly, the mortality and life quality of subgroups of SSc, the associated side effects and lack of efficacy in certain subgroups of organ involvement indicate that the available treatment options are often still unsatisfactory (e.g. digital ulceration, pulmonary fibrosis, ). Therefore, future treatments should specifically modulate distinct pathogenetic events in SSc.

References

Artlett CM, Smith CB, Jimenez SA (1998) Identification of fetal DNA and cells in skin lesions from women with systemic sclerosis. N Engl J Med 338: 1186–1191 Badesch D, Tapson VF, McGoon MD (2000) Continuous intravenous epoprostenol for pulmo- nary hypertension due to the scleroderma spectrum of disease. Ann Int Med 132: 425–434 Black CM, Silman SJ, Herrick AI (1998) Interferon-alpha does not improve outcome at one year in patients with diffuse cutaneous scleroderma: results of a randomized, double blind, placebo-controlled trial. Arthritis Rheum 42: 299–305 Blockmans D, Beyens G, Verhaege R (1996) Predictive value of nailfold capillaroscopy in the diagnosis of connective tissue diseases. Clin Rheumatol 15: 148–153 Bottomley WW, Goodfield MJ, Sheehan-Dare RA (1996) Digital calcification in systemic scle- rosis: effective treatment with good tissue preservation using the carbon dioxide laser. Br J Dermatol 135: 302–302 Bunn CC, Denton CP, Shi-Wen X, Knight C, Black CM (1998) Anti-RNA Polymerases and other autoantibody specificities in systemic sclerosis. Br J Rheumatol 37: 15–20 Burastero SE, Galbiati S, Vassallo A, Sabbadini MG, Bellone M, Marchionni L et al. (2003) Cel- lular microchimerism as a lifelong physiologic status in parous women: an immunologic basis for its amplification in patients with systemic sclerosis. Arthiritis Rheum 48: 1109–1116 Chang YJ, Spiera H (1999) Renal transplantation in scleroderma. Medicine 78: 382–385 Clements PJ, Lachenbruch PA, Ng SC (1990) Skin score. A semiquantitative measure of cuta- neous involvement that improves prediction of prognosis in systemic sclerosis. Arthritis Rheum 33: 1256–1263 Clements PJ, Furst DE (1997) Choosing appropriate patients with systemic sclerosis for treat- ment by autologous stem cell transplantation. J Rheumatol 24: 85–88 Clements PJ, Furst DE, Wong WK (1999) High dose versus low dose D-penicillamine in early diffuse systemic sclerosis: analysis of a two year, double blind, randomized, controlled clinical trial. Arthritis Rheum 42: 1194–203 180 Nicolas Hunzelmann and Thomas Krieg

Denton CP, Sweny P Abdulla A, Black CM (1994) Acute renal failure occurring in sclero- derma treated with cyclosporin A: a report of three cases. Br J Rheumatol 33: 90–92 Dong C, Zhu S, Wang T, Yoon W, Li Z, Alvarez RJ et al. (2002) Deficient Smad 7 expression: aputative molecular defect in scleroderma. PNAS 99: 3908–13 Duncan MR, Hasan A, Berman B (1995) Pentoxifylline, pentifylline and interferons decrease type I and III procollagen mRNA levels in dermal fibroblasts: evidence for mediation by nuclear factor 1 downregulation. J Invest Dermatol 104: 282–286 Dziadzio M, Denton CP Smith R (1999) Losartan therapy for raynauds phenomenon and scleroderma: clinical and biochemical findings in a fifteen-week, randomized, parallel group, controlled trial. Arthritis Rheum 42: 2646–2455 Eckes B, Mauch C, Huppe G, Krieg T (1996) Differential regulation of and tran- script stability of pro a1(I) collagen and fibronectin in activated fibroblasts derived from patients with systemic sclerosis. Biochem J 315: 549–553 Enomoto, Mekkes JR, Bossuyt PM (1999) Treatment of patients with systemic sclerosis with extracorporeal photochemotherapy (photopheresis). J Am Acad Dermatol 41: 915–922 Evans PC, Lambert N, Maloney S (1999) Long term fetal microchimerism in peripheral blood mononuclear scell subsets in healthy women and women with scleroderma. Blood 93: 2033–2037 Fritzler MJ (1993) Autoantibodies in scleroderma. J Dermatol 20: 257–268 Furst DE, Clements PJ, Steen VD (1998) The modified rodnan skin score is an accurate reflec- tion of skin biopsy thickness in systemic sclerosis. J Rheumatol 25: 84–88 Genth E, Mierau R, Genetzky P (1990) Immunogenetic associations of scleroderma-related antinuclear antibodies. Arthritis Rheum 33: 657–665 Jablonska S, Blaszyk M (1999) Scleroderma Overlap Syndromes. Adv Exp Med Biol 455: 85–92 Janowsky EC, Kupper LL, Hulka BS (2000) Meta-analyses of the relation between silicone breast implants and the risk of connective tissue. N Engl J Med 342: 781–790 Haustein UF, Anderegg U (1998) Silica induced scleroderma – clinical and experimental as- pects. J Rheumatol 25: 1917–1926 Hebbar M, Peyrat JP, Hornez L, Hatron PY, Hachulla E, Devulder B. (2000) Increased concen- trations of the circulating angiogenesis inhibitor endostatin in patients with systemic scle- rosis. Arthritis Rheum 43: 889–93 Heickendorff L, Zachariae H, Bjerring P, Halkier-Sorensen L, Sondergaard K (1995) The use of serologic markers for collagen synthesis and degradation in systemic sclerosis. J Am Acad Dermatol 32: 584–588 Herrmann K, Heckmann M, Kulozik M, Haustein UF, Krieg T (1991) Steady state mRNA lev- els of collagens I,III, fibronectin and collagenase in skin biopsies of systemic sclerosis pa- tients. J Invest Dermatol 97: 219–225 Herrmann G, Wlaschek M, Lange TS, Prenzel K, Goerz G, Scharffetter-Kochanek K (1993) UVA irradiation stimulates the synthesis of various matrix-metalloproteinases (MMPs) in cultured human fibroblasts. Exp Dermatol 2: 92–97 Hoeper MM et al. (2000) Long term treatment of primary pulmonary hypertension with aerolized , a prostacyclin analogue. N Engl J Med 342: 1866–70 Hunzelmann N, Anders S, Fierlbeck G, Hein R, Herrmann K, Albrecht M, Bell S, Thur J, Muche R, Adelmann-Grill B,Wehner-Caroli J, Gaus W, Krieg T (1997) Systemic sclero- derma: multicenter trial of one year treatment with recombinant interferon-gamma. Arch Dermatol 133: 609–613 Hunzelmann N, Risteli J, Risteli L, Sacher C, Black CM, Krieg T (1998a) Increased circulating concentrations of the type I collagen specific degradation product ICTP in systemic sclero- sis reflect the extent of skin involvement: A new serum marker for clinical severity in SSc patients. Br J Dermatol 139: 1020–1025 Hunzelmann N, Scharfetter-Kochanek K, Hager C, Krieg T (1998b) Management of localized scleroderma. Sem Cutan Med Surg 17: 34–41 Igarashi A, Nashiro K, Kikuchi K (1995) Significant correlation between connective tissue growth factor gene expression and skin sclerosis in tissue sections from patients with sys- temic sclerosis. J Invest Dermatol 105: 280–284 Progressive Systemic Scleroderma 181

Jimenez SA and Saitta B (2000) Alterations in the regulation of expression of the a1(I) colla- gen gene (COL1A1) in systemic sclerosis (scleroderma). Springer Semin Immunopathol 21: 397–414 Kahaleh MB, Sultany GL, Smith EA, Huffstetter JE, Loadholt CB, LeRoy EC (1986) A modi- fied scleroderma skin scoring method. Clin Exp Rheumatol 4: 367–369 Kahaleh M, Fan PS (1997) Mechanism of serum mediated endothelial injury in scleroderma: identification of a granular enzyme in scleroderma skin and sera. Clin Immunol Immu- nopathol 83: 32–40 Kerscher M, Meurer M, Sander C, Volkenandt M, Lehmann P, Plewig G, Röcken M (1996) PUVA bath photochemotherapy for localized scleroderma. Arch Dermatol 132: 1280–1282 Kulozik M, Hogg A, Lankat-Buttgereit B, Krieg T (1990) Co-localization of transforming growth factor b2 with a1(1) procollagen mRNA in tissue sections of patients with systemic sclerosis. J Clin Invest 86: 917–921 LeRoy EC (1974) Increased collagen synthesis by scleroderma fibroblasts in vitro. J Clin In- vest 54: 880–889 Lunardi C (2000) Systemic sclerosis autoantibodies bind the human cytomegalovirus late protein UL94 and induce apoptosis in human endothelial cells. Na- ture Med 6: 1183–1186 Majewski S, Hunzelmann N, Johnson JP, Jung C, Mauch C, Ziegler-Heitbrock H, Riethmüller G, Krieg T (1991) Expression of intercellular adhesion molecule-1 (ICAM-1) on fibroblasts in the skin of patients with systemic sclerosis. J Invest Dermatol 97: 667–671 Marguerie C (1992) The clinical and immunogenetic features of patients with autoantibodies to the nucleolar anitgen PM-Scl. Medicine 71: 327–336 Maricq H, LeRoy E, Dangelo W (1980) Diagnostic potential of in vivo capillary microscopy in scleroderma and related disorders. Arthritis Rheum 23: 183–189 Mauch C (1998) Regulation of connective tissue turnover by cell-matrix interactions. Arch Dermatol Res 290: S30–36 McCormick LL, Zhang Y, Tootell E, Gilliam AC (1999) Anti-TGF-beta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: a model for human scleroderma. J Immunol 163: 5693–5699 McKown KM, Carbone LD, Bustillo J (2000) Induction of immune tolerance to human type I collagen in patients with systemic sclerosis by oral administration of bovine type I colla- gen. Arthritis Rheum 43: 1054–1061 Perlish JS, Lemlich G, Fleischmajer R (1988) Identification of collagen fibrils in scleroderma skin. J Invest Dermatol 90: 48–54 Pigula FA, Griffith BP, Zenati MA (1997) Lung transplantation for respiratory failure resulting from systemic disease. Ann Thorac Surg 64: 1630–1634 Poormoghim H, Lucas M, Fertig N, Medsger TA (2000) Systemic sclerosis sine scleroderma: demographic, clinical, and serologic features and survival in forty eight patients. Arthritis Rheum 43: 444–451 Pope J, Bellamy N, Seibold J (1998) A controlled trial of methotrexate versus placebo in early diffuse scleroderma – preliminary analysis. Arthritis Rheum 41: S420 Pope J, Fenlon D, Thompson A (2000) Iloprost and cisaprost for Raynaud’s phenomenon in progressive systemic sclerosis. Cochrane Database Syst Rev 2: CD000953 Prescott RJ, Freemont A, Jones C (1992) Sequential dermal microvascular and perivascular changes in the development of scleroderma. J Pathol 166: 255–263 Rook AH, Freundlich B, Jegasothy BV (1992) Treatment of systemic sclerosis with extracor- poreal photochemotherapy. Results of a multicenter trial. Arch Dermatol 128: 337–346 Rubin LJ, Badesch DB, Barst RJ, Galie N, Black CM et al. (2002) therapy for pulmo- nary hypertension. NEJM 346: 896–903 Scharffetter K, Lankat-Buttgereit B, Krieg T (1988) Localization of collagen mRNA in normal and scleroderma skin by in situ hybridization. Eur J Clin Invest 18: 9–17 Scharffetter K, Wlaschek M, Hogg A, Bolsen K, Schothorst A, Goerz G, Krieg T, Plewig G (1991) UVA irradiation induces collagenase in human dermal fibroblasts in vitro and in vivo. Arch Dermatol Res 283: 506–511 Seibold J (1994) Systemic sclerosis. In: Klipped J, Dieppe P, Mosby St. Louis (Eds) Rheumato- logy, 8.1–8.14 182 Nicolas Hunzelmann and Thomas Krieg

Seibold J, Korn J, Simms R (2000) Recombinant human relaxin in the treatment of sclero- derma. A randomized, double blind, placebo-controlled study. Ann Int Med 132: 871–879 Sgonc R, Gruschwitz M, Dietrich H (1996) Endothelial cell apoptosis is a primary pathogene- tic event underlying skin lesions in avian and human scleroderma. J Clin Invest 98: 785–792 Shi-Wen X, Pennington D, Holmes A (2000) Autocrine overexpression of CTGF maintains fi- brosis: RDA analysis of fibrosis genes in systemic sclerosis. Exp Cell Res 25: 213–224 Sjogren RW (1996) Gastrointestinal features of scleroderma. Curr Opin Rheumatol 8: 569–575 Steen VD, Constantino JP, Shapiro AP, Medsger TA (1990) Outcome of renal crisis in sys- temic sclerosis: relation to availability of angiotensin converting enzyme (ACE) inhibitors. Ann Int Med 113: 352–357 Steen VD, Follansbee WP, Conte CG, Medsger TA (1996) Thallium perfusion defects predict subsequent cardiac dysfunction in patients with systemic sclerosis. Arthritis Rheum 39: 677–681 Steen VD, Medsger TA (1998) Case-control study of corticosteroids and other drugs that ei- ther precipitate or protect from the development of scleroderma renal crisis. Arthritis Rheum 41: 1613–1619 Stege H, Berneburg M, Humke S (1997) High-dose UVA1 for localized scleroderma. J Am Acad Dermatol 36: 938–944 Stone PJ, Korn JH, North H, Lally EV, Miller LC, Tucker LB, Strongwater S, Snider GL, Franzblau C (1995): Cross-linked elastin and collagen degradation products in the urine of patients with scleroderma. Arthritis Rheum 38: 517–524 Straniero NR, Furst DE (1989) Environmentally induced systemic sclerosis like illness. Baillie- res Clin Rheumatol 3: 63–79 Subcommittee for Scleroderma Criteria of the American Rheumatism Association Diagnosis and Therapeutic Criteria Committee (1980) Preliminary criteria for the classification of systemic sclerosis (scleroderma). Arthritis Rheum 23: 581–590 Tyndall A, Black C, Finke J, Peter H, Gratwohl A (1997) Treatment of systemic sclerosis with autologous hemopoetic stem cell transplantation. The Lancet 349: 254–356 Uitto J, Bauer EA, Eisen AZ (1979) Scleroderma. Increased biosynthesis of triple helical type I and type III procollagen associated with unaltered expression of collagenase by skin fibroblasts in culture. J Clin Invest 64: 921–930 van den Hoogen FH, Boerbooms AM, Swaak AJ (1996) Comparison of methotrexate with pla- cebo in the treatment of systemic sclerosis: a 24 week randomized double blind trial, fol- lowed by a 24 week observation trial. Br J Rheumatol 35: 364–372 White B, Bauer EA, Goldsmith LA (1995) Guidelines for clinical trials in systemic sclerosis (scleroderma). I. disease modifying interventions. The American College of Rheumato- logy Committee on Design and Outcomes in Clinical Trials in Systemic sclerosis. Arthritis Rheum 38: 351–360 White B, Moore WC, Wigley FM (2000) Cyclophosphamide is associated with pulmonary function and survival benefit in patients with scleroderma and alveolitis. Ann Int Med 132: 947–954 Williams HJ, Alarcon GS, Joks R (1999) Early undifferentiated connective tissue disease. VI an inception cohort after 10 years: disease remissions and changes in diagnoses in well es- tablished and undifferentiated CTD. J Rheumatol 26: 816–825 Zachariae H, Halkier-Sorensen L, Bjerring P, Heickendorff L (1996) Treatment of ischemic digital ulcers and prevention of gangrene with intravenous iloprost in systemic sclerosis. Acta Derm Venereol 76: 236–238