265

Immunosuppression for Connective Tissue Disease–Related Pulmonary Disease Toby M. Maher, MB, MSc, PhD, FRCP1,2,3

Street, London, United Kingdom 2 Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom 3 Centre for Inflammation and Tissue Repair, University College London, Rayne Institute, London, United Kingdom

Address for correspondence Toby M. Maher, MB, MSc, PhD, FRCP, NIHR Biological Research Unit, Royal Brompton Hospital, Sydney Street, London, SW3 6NP, United Kingdom (e-mail: [email protected]).

Semin Respir Crit Care Med 2014;35:265–273.

Abstract

Keywords

► interstitial lung disease ► rituximab ► cyclophosphamide ► systemic sclerosis ► therapy ► polymyositis ► pulmonary vasculitis

With improvements in the systemic treatment of the various connective tissue diseases (CTDs) the pulmonary complications of these conditions are now, for many patients, the major cause of morbidity and impaired quality of life. Furthermore, at least in scleroderma, pulmonary disease has become the leading cause of death for this patient group. Although, the pathogenesis of CTD-related pulmonary disease is poorly understood there is an assumption that it arises as a sequelae of immune-mediated injury to the lung. As a result, immunosuppressant agents form the mainstay of treatment for pulmonary disease occurring in the context of CTD. There is, however, a paucity of clinical trial data available to inform treatment decisions across the spectrum of CTDrelated pulmonary disease. The best available evidence has been generated in scleroderma-associated interstitial lung disease. Therefore, treatment decisions are informed by registry data, case series, and individual case reports. With this in mind, corticosteroids together with azathioprine and mycophenolate mofetil are widely used for the management of mild disease or as a maintenance therapy, while cyclophosphamide and rituximab have emerged as treatments for refractory or rapidly progressive disease. This article examines the data underpinning the use of different immunosuppressants in CTD-associated pulmonary disease while highlighting limitations in the existing knowledge base and identifying questions for future clinical study.

As has been described elsewhere, a range of pulmonary complications are encountered across the different connective tissue diseases (CTDs). The nature of these pulmonary manifestations frequently differs according to the underlying CTD but they are, in general, driven by the same immunemediated processes, which occur in other disease-affected organs. As such, with a few exceptions, immunosuppressant drugs form the mainstay of treatment for pulmonary disease arising in individuals with CTD. This review will explore the current knowledge base concerning immunosuppressant therapy for pulmonary-related CTD manifestations including;

Issue Theme Pulmonary Complications of Connective Tissue Disease; Guest Editors, Danielle Antin-Ozerkis, MD, and Jeffrey Swigris, DO, MS

specific choice of immunosuppressant, timing of treatment, and the anticipated benefits of therapy.

Treatment Principles and Evidence Base Despite pulmonary complications being one of the most important causes of morbidity and mortality in individuals with CTD there have been a very few clinical trials conducted in this area.1–4 To date, only two appropriately powered, randomized controlled trials of treatment have been conducted in this patient group; with both of these assessing the

Copyright © 2014 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662.

DOI http://dx.doi.org/ 10.1055/s-0034-1371531. ISSN 1069-3424.

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

1 NIHR Biological Research Unit, Royal Brompton Hospital, Sydney

CTD-ILD Treatment

Maher

benefit of cyclophosphamide compared with placebo for the treatment of scleroderma-related interstitial lung disease (ILD).5,6 Thus, there is much that is unknown regarding optimal treatment of CTD-related pulmonary disease with decisions resting on the results of open label studies, registry data, and expert opinion. Important lacunae in the knowledge base includes the following: (1) the optimal time for initiating treatment and whether this should be at the time of discovery of pulmonary complications or only in those with extensive or progressive disease, (2) whether early and aggressive treatment alters disease outcomes in a manner akin to that seen in rheumatoid arthritis (RA) joint disease, (3) the relative beneficial effects of individual immunosuppressants and whether combination therapy is better than single agent treatment, (4) whether in some circumstances, for example, RA-associated ILD, immunosuppressant drugs may contribute to the development and progression of pulmonary disease, (5) whether treatments targeted at one organ system, for example, joint disease are effective in the treatment of other organ disease manifestations, and (6) the optimal duration of treatment or which measures should be used to guide treatment withdrawal. Answering these questions, although far from easy (due to the relative rarity of the diseases involved, the varied pulmonary presentations of disease and a lack of validated trial endpoints) will require carefully designed, multicenter studies of sufficient duration.7 The studies should be a priority for both the respiratory and rheumatology clinical communities and the relevant patient advocacy groups and charities. While individual CTDs give rise to a reasonably predictable range of pulmonary complications, autoimmune disease in general can affect any compartment of the lung giving rise to a variety of disease presentations. These include: airways disease (e.g., obliterative bronchiolitis in RA), ILD (with the observed histological lesions covering the full spectrum of those encountered in the idiopathic interstitial pneumonias, ranging from usual interstitial pneumonia (UIP) through to desquamative interstitial pneumonitis and even diffuse alveolar damage), pulmonary vascular disease (both pulmonary hypertension and vasculitis), and pleural disease (with

involvement extending from acute serositis through to chronic progressive pleural thickening). While the details of the patterns of pulmonary involvement in individual CTDs are discussed in other articles in this issue, ►Table 1 provides a summary of the characteristic pulmonary complications seen in the different CTDs with an indication of their relative frequency. The majority of evidence supporting the use of immunosuppressants in the treatment of CTD-related pulmonary disease relates to the management of ILD and, to a lesser extent, vasculitis. In certain CTDs where pulmonary hypertension appears to arise as a consequence of vasculitic inflammation, for example, systemic lupus erythematosus (SLE) and mixed connective tissue disease (MCTD), treatment with immunosuppressants can be effective. However, in scleroderma, a disease in which pulmonary hypertension occurs in up to 20% of cases, immunosuppression has no effect on either pulmonary pressures or subsequent disease progression and is therefore avoided unless indicated for other disease manifestations.8–10 Airways disease, especially obliterative bronchiolitis (OB), is another pulmonary complication that appears to respond poorly to immunosuppressant therapy and for which aggressive immunosuppression may even be deleterious.11 There is some evidence to suggest that repeated airway infection may be a cofactor for the development of OB, in particular in the lung transplant literature, where OB is the most common manifestation of chronic graft rejection.12,13 In lung transplant recipients protection against airway injury (through antibiotic prophylaxis14 and the prevention of microaspiration15) reduces the incidence and severity of OB. Anecdotally, the same may well be true in RArelated OB. The role of immunosuppression in the management of pleural disease complications is unclear and may vary between diseases being seemingly more effective in the acute plural manifestations of SLE than in the chronic pleural thickening occasionally seen in rheumatoid disease. In the context of CTD-ILD there are very little high-quality clinical trial data to guide the decision to initiate treatment. There is reasonable evidence across the CTDs to suggest that both the histological pattern of interstitial involvement and

Table 1 Connective tissue diseases and common pulmonary manifestations ILD UIP

NSIP

Airways

Pleural

Vascular

DAH

OP

Systemic sclerosis



þþþ







þþþ



Rheumatoid disease

þþ

þ

þ

þþ

þþ

þ



Primary Sjogren syndrome



þ



þþ

þ

þ



Mixed CTD

þ

þþ

þ

þ

þ

þþ



Idiopathic inflammatory myositis



þþ

þþþ





þ



Systemic lupus erythematosus



þ

þ

þ

þþþ

þ

þþ

Abbreviations: CTD, connective tissue disease; DAH, diffuse alveolar hemorrhage; ILD, interstitial lung disease; NSIP, nonspecific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumonia. Note: Symbols represent relative frequency of each complication (, not seen; , very low prevalence; þ, low prevalence; þþ, medium prevalence; and þþþ, high prevalence). Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

266

Maher

severity of lung function impairment are both prognostic determinants (with UIP conferring a worse prognosis than nonspecific interstitial pneumonia [NSIP]3,16). Nonetheless, there is insufficient evidence to use these factors as a guide for therapy. Given an absence of evidence supporting diseasemodifying effects of either early and/or aggressive treatment of CTD-ILD combined with knowledge of the potential toxic effects of immunosuppressant treatment, the majority of clinicians tend to base their therapeutic decisions on disease severity and the level of functional impairment experienced by the affected patient. For individuals with CTD-ILD, the underlying histological lesion, as well as predicting prognosis, does, to a certain extent, predicts response to treatment. As demonstrated in ►Fig. 1, both organizing pneumonia and cellular NSIP can be expected to regress with therapy resulting in functional and lung function improvement. UIP, by contrast, is characterized by fixed fibrosis and architectural destruction (see ►Fig. 2) and the best that can be hoped for with treatment is stabilization of disease and prevention of further decline. With all the caveats concerning the current evidence base in mind, the remainder of this article will focus on individual immunosuppressants and the evidence supporting their use across the different CTDs and for the variety of pulmonary complications observed in these diseases.

Corticosteroids Although, corticosteroids are very widely used in the management of CTD-associated pulmonary disease, there are surprisingly little data to guide their use. The dose and route of corticosteroid administration is driven predominantly by local practice, expert opinion, and by reference to data generated from nonpulmonary disease. For patients with

Fig. 1 High-resolution computed tomography (CT) scans from a 52year-old man with scleroderma-related interstitial lung disease before (A) and 6 months after (B) treatment with six intravenous doses of cyclophosphamide (600 mg/m 2 body surface area) given at monthly intervals together with 10 mg oral prednisolone daily. The initial CT shows diffuse ground glass attenuation with some reticular change and associated traction bronchiectasis. Surgical lung biopsy confirmed mixed cellular and fibrotic nonspecific interstitial pneumonitis. Following therapy there has been a marked improvement in the CT appearances and this corresponded with a 30% improvement in lung function indices.

Fig. 2 Low-power photomicrograph (A) and high-resolution computed tomography (CT) scan (B) from a 54-year-old man with endstage rheumatoid-associated interstitial lung disease. The histological image demonstrates the characteristic appearance of usual interstitial pneumonia, with marked architectural destruction of the lung with loss of alveolar spaces, dense extracellular matrix deposition, fibroblastic foci, and bronchiolized honeycomb cysts. The CT scan shows the corresponding radiological changes of honeycomb destruction of the lung. The architectural destruction, characterized by the honeycomb change, exemplifies why this form of interstitial lung disease is poorly treatment responsive and conveys a grave prognosis.

scleroderma prednisolone doses are usually limited to 20 mg on alternate days or 10 mg daily so as to minimize the risk of steroid-induced renal crises.17 A recent singlecenter retrospective review of scleroderma cases suggested that, over 12 months, single agent corticosteroid treatment is associated with improved lung function (increase in forced vital capacity [FVC] of 158 mL) compared with no therapy (which resulted in a loss of 61 mL of FVC).18 Organizing pneumonia in the context of the idiopathic inflammatory myopathies is the pulmonary complication of CTD most sensitive to corticosteroid treatment. For this reason high-dose corticosteroids (either pulsed intravenous methyl prednisolone or an initial dose of 0.75–1 mg/kg/d of prednisolone) are typically used as first-line therapy in this disorder. This approach to management is supported by data from case reports and outcome observed in cryptogenic organizing pneumonia but has not been subject to assessment in clinical trials.19–22 Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

267

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

CTD-ILD Treatment

CTD-ILD Treatment

Maher

Cyclophosphamide The alkylating agent cyclophosphamide is the best studied of all the immunosuppressants used for CTD-related pulmonary disease. It has been assessed as a treatment for scleroderma ILD in two multicenter randomized, placebo controlled trials. The first of these studies, by Tashkin et al compared oral cyclophosphamide (at a dose of 2 mg/kg body weight per day) to matching placebo for 1 year in 158 patients with scleroderma-associated ILD.6 The primary study endpoint was change in FVC at 1 year. Cyclophosphamide treatment was associated with a small (2.53%) but statistically significant (p ¼ 0.03) improvement in FVC when compared with placebo. There were no differences in serious adverse events between placebo and cyclophosphamide although there were more episodes of leucopoenia and hematuria in the active treatment arm. In the second study by Hoyles et al, 45 patients were randomized to receive a combination of lowdose prednisolone, six infusions of cyclophosphamide (given at monthly intervals at a dose of 600 mg/m2 body surface area), followed by azathioprine (2.5 mg/kg/d to a maximum of 200 mg daily) or triple placebo.5 Primary outcome was change in percent predicted FVC and single-breath diffusing capacity for carbon monoxide (DLco) at 1 year. There was a trend (p ¼ 0.08) toward improved FVC in the active treatment arm. As has been argued cogently by Wells et al, both of these studies were hampered in their recruitment of patients because they were conducted at a time when cyclophosphamide was already judged by most clinicians to be the standard of care for patients with progressive scleroderma-associated ILD.23 As such there was a reluctance by investigators to enroll individuals with progressive disease. Instead such individuals were, in the most part, treated outside the study with open label therapy. This view is borne out by the lack of disease progression observed in the placebo arms in both studies. Given that these studies were therefore conducted in individuals with relatively stable fibrotic disease it is perhaps unsurprising that relatively little treatment effect was observed. Several open-label retrospective studies of cyclophosphamide given for scleroderma-associated ILD have been reported in the literature.24–31 Overall these favor cyclophosphamide and tend to suggest a significant improvement in FVC. In most cases, pulsed intravenous cyclophosphamide and daily oral cyclophosphamide appear to have similar efficacy albeit that intravenous dosing appears to be better tolerated. Longitudinal follow-up of the study by Tashkin et al suggests that the beneficial effect of cyclophosphamide treatment wanes over time.6,32,33 Optimal duration of treatment therefore remains to be defined. As in the study by Hoyles et al many centers follow pulsed cyclophosphamide with either azathioprine or mycophenolate mofetil (MMF).5 The efficacy of this approach and appropriate length of treatment regimen remain to be defined.27,34 Cyclophosphamide has not been subjected to clinical trial assessment in any other CTDassociated pulmonary complication. Retrospective case series suggest that it has a similar efficacy to that seen in scleroderma ILD when used as a treatment for ILD associated with Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

polymyositis, dermatomyositis or MCTD.35–37 It is unknown whether early treatment of CTD-ILD with cyclophosphamide has a disease-modifying effect and for this reason most centers only treat individuals with either extensive or progressive disease. In scleroderma the classification of ILD into limited or extensive, as described by Goh et al,38 provides a useful method for identifying patients requiring early aggressive treatment of their ILD. Ultimately, these questions require carefully designed and appropriately powered studies to provide the necessary answers for clinicians dealing with the practical issue of how best to treat scleroderma and other CTD-ILDs. The other disease area for which cyclophosphamide is considered first- or second-line treatment is in the management of CTD-related vasculitis and acute alveolar hemorrhage.39–41 In such cases the choice of treatment is analogous to that used in the management of other systemic vasculitidies.40,42–44

Azathioprine The purine analogue azathioprine exerts an immunosuppressant effect through inhibition of T- and B-lymphocyte proliferation. Although, widely used in combination with oral corticosteroids in the treatment of CTD-ILD, azathioprine has not been studied alone in any prospective randomized trials. In the study by Hoyles et al of intravenous cyclophosphamide, azathioprine (at a dose of 2.5 mg/kg/d) was used as maintenance therapy for 6 months following initial treatment with intravenous cyclophosphamide.5 As noted earlier, the active treatment arm was associated with a trend toward improved FVC at 12 months. In a prospective open-label study of 13 patients, Paone et al reported that 100 mg daily of azathioprine for 12 months maintained improvements achieved after 1 year’s treatment with intravenous cyclophosphamide in patients with scleroderma-associated ILD.45 In an observational cohort study, Bérezné et al reported similar stability in 23 patients treated with azathioprine following cyclophosphamide for scleroderma ILD.27 A retrospective review of 11 scleroderma patients treated with combined oral corticosteroids and azathioprine, suggested that for patients tolerating treatment, therapy was associated with stabilization or improvement in FVC compared with pretreatment baseline values.27 In idiopathic inflammatory myopathy (IIM), a retrospective analysis of a Taiwanese disease registry reported that use of azathioprine was associated with improved survival.46 The effect of azathioprine in rheumatoid-associated ILD, SLE, or Sjogren-related ILD has not been reported. For patients with pulmonary hypertension in the context of SLE, MCTD, and RA azathioprine is the most frequently used immunosuppressive therapy albeit with limited evidence to support its use or define efficacy.47

Methotrexate The antifolate drug methotrexate is thought to exert its immunosuppressant effect through inhibition of purine metabolism and consequent suppression of T-cell activation and intercellular adhesion molecule expression.48 Despite

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

268

CTD-ILD Treatment

Mycophenolate Mofetil MMF is an inosine monophosphate dehydrogenase inhibitor, which reduces T- and B-cell proliferation through reduction in purine synthesis. MMF has been shown to be well tolerated in systemic sclerosis with retrospective studies suggesting that the drug has favorable effects on systemic manifestations of disease.55–59 In another retrospective study, Fischer et al looked at MMF use in 125 individuals with CTD-ILD, the largest proportion (44) of whom had scleroderma, and were able to demonstrate that MMF was associated with reduced steroid requirement and sustained improvement in FVC.60 An analysis of 69 patients across seven published case series suggests that MMF may stabilize disease in sclerodermaassociated ILD.61 In a consensus statement by the scleroderma clinical trials consortium and the Canadian Scleroderma Research Group, MMF was recommended by three-quarters of the 117 experts surveyed as maintenance therapy for scleroderma ILD.8 The scleroderma lung study (SLS) II is currently evaluating the effect of MMF as first-line therapy compared with cyclophosphamide in scleroderma ILD in a 2year randomized controlled trial (NCT00883129).

Calcineurin Inhibitors The calcineurin inhibitors ciclosporin and tacrolimus inhibit T-cell activation and signal transduction and are typically used to prevent rejection following solid organ transplant. Both drugs have been reported in case studies and small case series to be effective in the treatment of ILD associated with either polymyositis or dermatomyosistis.62–69 The largest of these studies reports 15 cases (identified from a registry of 179 patients with myositis) of IIM-associated ILD treated with calcineurin inhibitors for a median of 24 months. The majority (11 cases) were being treated for refractory disease.62 Overall, 13 of the 15 treated individuals had an improvement in lung function following initiation of a calcineurin inhibitor with an overall mean increase in FVC of 10% occurring over the course of active treatment. The available evidence suggests that calcineurin inhibitors should be considered for use as a therapy in individuals with treatment-resistant IIM-ILD or for those patients who relapse following standard first- and second-line therapy.

269

Rituximab Rituximab, a chimeric (human/mouse) monoclonal antibody with a high affinity for the CD20 surface antigen expressed on pre-B and B-lymphocytes, results in rapid depletion of B cells from the peripheral circulation for 6 to 9 months.70 Evidence for the effectiveness of B-cell depletion exists in several immune-mediated conditions, including RA,71–73 antineutrophil cytoplasmic antibody-associated vasculitis44,74 and immune thrombocytopenic purpura.75 In a small open label, randomized proof of principle study of rituximab in scleroderma ILD (given at a dose of 375 mg/m2 weekly for 4 weeks at baseline and again at 24 weeks), Daoussis et al reported a significant improvement in FVC, compared with baseline, at 1 year in the rituximab group. In the active treatment group FVC increased from 68.1  19.7% predicted to 75.6  19.7% predicted (p ¼ 0.0018).76 In the cohort receiving best standard care there was an overall reduction in FVC over the 12 months of the study. Rituximab has been reported as an effective rescue therapy for patients with scleroderma ILD unresponsive to treatment with corticosteroid.77 In individuals with IIM-ILD for whom cyclophosphamide is contraindicated or else fails to induce a therapeutic response, rituximab may be an effective alternative. In a retrospective cohort study, Keir et al reported outcomes in nine individuals (six of whom had either polymyositis or dermatomyositis) with CTD-ILD. Overall rituximab, even though used as rescue therapy, was associated with clinically important improvements in lung function, gas exchange, and radiological disease extent.78 A larger follow-up cohort of 50 patients with fibrotic ILD (including 10 with IIM, 8 with scleroderma, 2 with RA, and 2 with MCTD) treated with rituximab reports similar results, with an average 8.9% improvement in FVC after 6 to 12 months in patients with CTD-ILD.79 An ongoing clinical trial, the RECITAL study (NCT01862926), is assessing, in a double dummy, randomized controlled trial, the efficacy of rituximab compared with intravenous cyclophosphamide when given as first-line therapy in progressive CTD-ILD (including scleroderma, IIM, and MCTD). Like cyclophosphamide, rituximab has been reported to have efficacy in the treatment of pulmonary vasculitis and diffuse alveolar hemorrhage occurring in the context of preexisting connective tissue disease.79–81

Imatanib The tyrosine kinase inhibitor, imatinib, prevents protein phosphorylation through inhibition of the specific tyrosine kinase, BCR-Abl. The drug was originally developed as a therapy for chronic myeloid leukemia. In vitro studies in human lung fibroblasts and in vivo studies utilizing the murine bleomycin model, albeit with the use of prophylactic dosing (i.e., at the time of bleomycin administration) of imatinib, suggest that imatinib may have antifibrotic actions.82 Sabnani et al reported the use of imatinib (200 mg daily) in combination with intravenous cyclophosphamide (500 mg every 3 weeks) in five patients with sclerodermaassociated ILD.83 The combination was well tolerated but Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

methotrexate being the established first-line therapy for rheumatoid-related joint disease49 and having proven efficacy in preventing relapse of the vasculitidies,43 its role in the management of CTD-ILD is unclear. Mainly this is because of methotrexate’s perceived capacity to induce pulmonary complications.50,51 While methotrexate is undoubtedly capable of causing ILD, this appears to occur almost exclusively in individuals with RA and the incidence of this complication has probably been historically overestimated.52 Methotrexate is occasionally used for scleroderma ILD in individuals intolerant of azathioprine or MMF.8 There are reports of methotrexate being an effective second-line agent for ILD associated with MCTD and the idiopathic inflammatory myopathies.53,54

Maher

CTD-ILD Treatment

Maher

efficacy was not determined. In a phase IIa open-label, single arm study, Spiera et al reported the use of imatinib (400 mg daily) for 1 year in 24 patients with diffuse cutaneous systemic sclerosis. In this group FVC improved, on average, by 6.4% compared with baseline over 12 months (p ¼ 0.008).84 More recently Khanna et al have reported a study of imatinib 600 mg/daily in 20 patients with scleroderma-associated ILD.85 At this dose imatinib was poorly tolerated with common adverse effects including fatigue, edema, diarrhea, nausea, and vomiting, generalized rash and new onset proteinuria. Only 60% of subjects completed the study. No beneficial effect of imatinib on FVC was observed. The use of imatinib for other CTD-ILDs is limited to a single case report in which 400 mg daily of imatinib for 20 weeks was apparently effective in improving symptoms due to MCTD-associated ILD.86 These mixed results preclude the routine use of imatinib in scleroderma ILD but are sufficient to merit further randomized, placebo-controlled studies.

of the CTDs respiratory disease is the leading cause of both morbidity and mortality. Despite an increasing repertoire of available therapies for the treatment of immune-mediated disease, there have been few significant advances in the management of the pulmonary complications of CTD. Corticosteroids together with azathioprine or MMF form the mainstay of treatment in milder cases while cyclophosphamide and, increasingly, rituximab represent the major treatment options for refractory or rapidly progressive disease. There is however, a desperate need for high-quality clinical trials in patients with CTD-related pulmonary disease so as to define both the best therapies and also the optimal management approach for this debilitating group of disorders.

Note T.M. is in receipt of unrestricted academic industry grants from GSK and Novartis. In the last 3 years T.M. or his institution have received advisory board or consultancy fees from; Actelion, Boehringer Ingelheim, GSK, InterMune, Novartis, Lanthio, Takeda, Sanofi-Aventis, and UCB. T.M. has received speaker’s fees from UCB, Boehringer Ingelheim, InterMune, and AstraZeneca. T.M. has participated as an investigator in industry sponsored clinical trials run by Boehringer Ingelheim, GSK, InterMune, Novartis, Roche, and Celgene.

Other Immunomodulatory Treatments A range of monoclonal antibody therapies have emerged in the fields of rheumatology and oncology in the last decade. While many of these drugs have an established role in the management of the systemic manifestations of CTD, their efficacy as potential treatments for CTD-ILD is largely unknown. The proinflammatory cytokine interleukin (IL)-6 has been implicated in the pathogenesis of pulmonary fibrosis and has been shown to be elevated in the serum of individuals with scleroderma ILD.87–90 Tocilizumab, an anti-IL-6 monoclonal antibody, has been reported to improve lung function in an individual with RA-ILD and in a 15-year-old girl with biopsy proven desquamative interstitial pneumonitis on a background of undefined autoimmune disease.91,92 Other monoclonal approaches that may have a potential future role in the management of CTD-ILD include, the anticomplement 5 antibody eculizumab19 and the anti CD-52 antibody alemtuzumab.93 By contrast the antitumor necrosis factor (TNF)-α antibodies, particularly infliximab and etanarcept, have been associated with an increased risk of developing ILD in individuals with rheumatoid disease.94–96 Although, TNF-α inhibition has been reported to be an effective treatment for the ILD associated with dermatomyositis,97 these drugs should be used with caution in individuals with preexisting lung disease in the context of CTD. In addition to the monoclonal antibodies, other immunomodulatory drugs that may be beneficial in the treatment of the pulmonary complications of CTD are the anti-IL-1Rα antibody, anakinra98,99 and hydroxychloroquine.100 Intravenous immunoglobulin therapy has also been reported to arrest the progression of ILD in several case reports and series of individuals with polymyositis or dermatomyositis.19,101,102

References 1 Tyndall AJ, Bannert B, Vonk M, et al. Causes and risk factors for

2 3

4

5

6

7

8

Conclusion As the systemic management of the CTDs has improved the pulmonary complications associated with these disorders have become an increasingly important problem. In many Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

9

death in systemic sclerosis: a study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann Rheum Dis 2010; 69(10):1809–1815 Fischer A, du Bois R. Interstitial lung disease in connective tissue disorders. Lancet 2012;380(9842):689–698 Kim EJ, Collard HR, King TE Jr. Rheumatoid arthritis-associated interstitial lung disease: the relevance of histopathologic and radiographic pattern. Chest 2009;136(5):1397–1405 Fischer A, Swigris JJ, du Bois RM, et al. Anti-synthetase syndrome in ANA and anti-Jo-1 negative patients presenting with idiopathic interstitial pneumonia. Respir Med 2009;103(11):1719–1724 Hoyles RK, Ellis RW, Wellsbury J, et al. A multicenter, prospective, randomized, double-blind, placebo-controlled trial of corticosteroids and intravenous cyclophosphamide followed by oral azathioprine for the treatment of pulmonary fibrosis in scleroderma. Arthritis Rheum 2006;54(12):3962–3970 Tashkin DP, Elashoff R, Clements PJ, et al; Scleroderma Lung Study Research Group. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med 2006;354(25):2655–2666 Khanna D, Brown KK, Clements PJ, et al. Systemic sclerosisassociated interstitial lung disease-proposed recommendations for future randomized clinical trials. Clin Exp Rheumatol 2010; 28(2, Suppl 58):S55–S62 Walker KM, Pope J; participating members of the Scleroderma Clinical Trials Consortium (SCTC); Canadian Scleroderma Research Group (CSRG). Treatment of systemic sclerosis complications: what to use when first-line treatment fails—a consensus of systemic sclerosis experts. Semin Arthritis Rheum 2012;42(1):42–55 Launay D, Humbert M, Berezne A, et al. Clinical characteristics and survival in systemic sclerosis-related pulmonary hypertension associated with interstitial lung disease. Chest 2011;140(4): 1016–1024

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

270

Maher

10 Opitz C, Klein-Weigel PF, Riemekasten G. Systemic sclerosis - a

27 Bérezné A, Ranque B, Valeyre D, et al. Therapeutic strategy

systematic overview: part 2 - immunosuppression, treatment of SSc-associated vasculopathy, and treatment of pulmonary arterial hypertension. Vasa 2011;40(1):20–30 Devouassoux G, Cottin V, Lioté H, et al; Groupe d’Etudes et de Recherche sur les Maladies “Orphelines” Pulmonaires (GERM”O”P). Characterisation of severe obliterative bronchiolitis in rheumatoid arthritis. Eur Respir J 2009;33(5):1053–1061 Magnusson J, Westin J, Andersson LM, Brittain-Long R, Riise GC. The impact of viral respiratory tract infections on long-term morbidity and mortality following lung transplantation: a retrospective cohort study using a multiplex PCR panel. Transplantation 2013;95(2):383–388 Gregson AL, Wang X, Weigt SS, et al. Interaction between Pseudomonas and CXC chemokines increases risk of bronchiolitis obliterans syndrome and death in lung transplantation. Am J Respir Crit Care Med 2013;187(5):518–526 Vos R, Vanaudenaerde BM, Ottevaere A, et al. Long-term azithromycin therapy for bronchiolitis obliterans syndrome: divide and conquer? J Heart Lung Transplant 2010;29(12):1358–1368 Abbassi-Ghadi N, Kumar S, Cheung B, et al. Anti-reflux surgery for lung transplant recipients in the presence of impedance-detected duodenogastroesophageal reflux and bronchiolitis obliterans syndrome: a study of efficacy and safety. J Heart Lung Transplant 2013;32(6):588–595 Tansey D, Wells AU, Colby TV, et al. Variations in histological patterns of interstitial pneumonia between connective tissue disorders and their relationship to prognosis. Histopathology 2004;44(6):585–596 Kowal-Bielecka O, Landewé R, Avouac J, et al; EUSTAR Co-Authors. EULAR recommendations for the treatment of systemic sclerosis: a report from the EULAR Scleroderma Trials and Research group (EUSTAR). Ann Rheum Dis 2009;68(5):620–628 Ando K, Motojima S, Doi T, et al. Effect of glucocorticoid monotherapy on pulmonary function and survival in Japanese patients with scleroderma-related interstitial lung disease. Respir Investig 2013;51(2):69–75 Gordon PA, Winer JB, Hoogendijk JE, Choy EH. Immunosuppressant and immunomodulatory treatment for dermatomyositis and polymyositis. Cochrane Database Syst Rev 2012;8:CD003643 Kotani T, Makino S, Takeuchi T, et al. Early intervention with corticosteroids and cyclosporin A and 2-hour postdose blood concentration monitoring improves the prognosis of acute/subacute interstitial pneumonia in dermatomyositis. J Rheumatol 2008;35(2):254–259 Connors GR, Christopher-Stine L, Oddis CV, Danoff SK. Interstitial lung disease associated with the idiopathic inflammatory myopathies: what progress has been made in the past 35 years? Chest 2010;138(6):1464–1474 Lazor R, Vandevenne A, Pelletier A, Leclerc P, Court-Fortune I, Cordier JF. Cryptogenic organizing pneumonia. Characteristics of relapses in a series of 48 patients. The Groupe d’Etudes et de Recherche sur les Maladles “Orphelines” Pulmonaires (GERM”O”P). Am J Respir Crit Care Med 2000;162(2 Pt 1):571–577 Wells AU, Latsi P, McCune WJ. Daily cyclophosphamide for scleroderma: are patients with the most to gain underrepresented in this trial? Am J Respir Crit Care Med 2007;176(10):952–953 Domiciano DS, Bonfá E, Borges CT, et al. A long-term prospective randomized controlled study of non-specific interstitial pneumonia (NSIP) treatment in scleroderma. Clin Rheumatol 2011; 30(2):223–229 Broad K, Pope JE. The efficacy of treatment for systemic sclerosis interstitial lung disease: results from a meta-analysis. Med Sci Monit 2010;16(9):RA187–RA190 Wanchu A, Suryanaryana BS, Sharma S, Sharma A, Bambery P. High-dose prednisolone and bolus cyclophosphamide in interstitial lung disease associated with systemic sclerosis: a prospective open study. Int J Rheum Dis 2009;12(3):239–242

combining intravenous cyclophosphamide followed by oral azathioprine to treat worsening interstitial lung disease associated with systemic sclerosis: a retrospective multicenter open-label study. J Rheumatol 2008;35(6):1064–1072 White B, Moore WC, Wigley FM, Xiao HQ, Wise RA. Cyclophosphamide is associated with pulmonary function and survival benefit in patients with scleroderma and alveolitis. Ann Intern Med 2000;132(12):947–954 Poormoghim H, Moradi Lakeh M, Mohammadipour M, Sodagari F, Toofaninjed N. Cyclophosphamide for scleroderma lung disease: a systematic review and meta-analysis. Rheumatol Int 2012;32(8): 2431–2444 Mittoo S, Wigley FM, Wise RA, Woods A, Xiao H, Hummers LK. Long term effects of cyclophosphamide treatment on lung function and survival in scleroderma patients with interstitial lung disease. Open Rheumatol J 2011;5:1–6 Tochimoto A, Kawaguchi Y, Hara M, et al. Efficacy and safety of intravenous cyclophosphamide pulse therapy with oral prednisolone in the treatment of interstitial lung disease with systemic sclerosis: 4-year follow-up. Mod Rheumatol 2011;21(3):296–301 Theodore AC, Tseng CH, Li N, Elashoff RM, Tashkin DP. Correlation of cough with disease activity and treatment with cyclophosphamide in scleroderma interstitial lung disease: findings from the Scleroderma Lung Study. Chest 2012;142(3):614–621 Roth MD, Tseng CH, Clements PJ, et al; Scleroderma Lung Study Research Group. Predicting treatment outcomes and responder subsets in scleroderma-related interstitial lung disease. Arthritis Rheum 2011;63(9):2797–2808 Dheda K, Lalloo UG, Cassim B, Mody GM. Experience with azathioprine in systemic sclerosis associated with interstitial lung disease. Clin Rheumatol 2004;23(4):306–309 Bodolay E, Szekanecz Z, Dévényi K, et al. Evaluation of interstitial lung disease in mixed connective tissue disease (MCTD). Rheumatology (Oxford) 2005;44(5):656–661 Okada M, Suzuki K, Matsumoto M, et al. Intermittent intravenous cyclophosphamide pulse therapy for the treatment of active interstitial lung disease associated with collagen vascular diseases. Mod Rheumatol 2007;17(2):131–136 Corte TJ, Ellis R, Renzoni EA, et al. Use of intravenous cyclophosphamide in known or suspected, advanced non-specific interstitial pneumonia. Sarcoidosis Vasc Diffuse Lung Dis 2009;26(2): 132–138 Goh NS, Desai SR, Veeraraghavan S, et al. Interstitial lung disease in systemic sclerosis: a simple staging system. Am J Respir Crit Care Med 2008;177(11):1248–1254 Jayne D. Treating vasculitis with conventional immunosuppressive agents. Cleve Clin J Med 2012;79(Suppl 3):S46–S49 Casian A, Jayne D. Management of alveolar hemorrhage in lung vasculitides. Semin Respir Crit Care Med 2011;32(3): 335–345 Schwarz MI, Zamora MR, Hodges TN, Chan ED, Bowler RP, Tuder RM. Isolated pulmonary capillaritis and diffuse alveolar hemorrhage in rheumatoid arthritis and mixed connective tissue disease. Chest 1998;113(6):1609–1615 Cordier JF, Cottin V. Alveolar hemorrhage in vasculitis: primary and secondary. Semin Respir Crit Care Med 2011;32(3): 310–321 Lynch JP III, Tazelaar H. Wegener granulomatosis (granulomatosis with polyangiitis): evolving concepts in treatment. Semin Respir Crit Care Med 2011;32(3):274–297 Stone JH, Merkel PA, Spiera R, et al; RAVE-ITN Research Group. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med 2010;363(3):221–232 Paone C, Chiarolanza I, Cuomo G, et al. Twelve-month azathioprine as maintenance therapy in early diffuse systemic sclerosis patients treated for 1-year with low dose cyclophosphamide pulse therapy. Clin Exp Rheumatol 2007;25(4):613–616

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

28

29

30

31

32

33

34

35

36

37

38

39 40

41

42

43

44

45

Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

271

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

CTD-ILD Treatment

CTD-ILD Treatment

Maher

46 Yu KH, Wu YJ, Kuo CF, et al. Survival analysis of patients with

64 Ando M, Miyazaki E, Yamasue M, et al. Successful treatment with

dermatomyositis and polymyositis: analysis of 192 Chinese cases. Clin Rheumatol 2011;30(12):1595–1601 Chung L, Liu J, Parsons L, et al. Characterization of connective tissue disease-associated pulmonary arterial hypertension from REVEAL: identifying systemic sclerosis as a unique phenotype. Chest 2010;138(6):1383–1394 Johnston A, Gudjonsson JE, Sigmundsdottir H, Ludviksson BR, Valdimarsson H. The anti-inflammatory action of methotrexate is not mediated by lymphocyte apoptosis, but by the suppression of activation and adhesion molecules. Clin Immunol 2005;114(2): 154–163 Smolen JS, Landewé R, Breedveld FC, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis 2013 Atzeni F, Boiardi L, Sallì S, Benucci M, Sarzi-Puttini P. Lung involvement and drug-induced lung disease in patients with rheumatoid arthritis. Expert Rev Clin Immunol 2013;9(7): 649–657 Roubille C, Haraoui B. Interstitial lung diseases induced or exacerbated by DMARDS and biologic agents in rheumatoid arthritis: A systematic literature review. Semin Arthritis Rheum 2013 Sathi N, Chikura B, Kaushik VV, Wiswell R, Dawson JK. How common is methotrexate pneumonitis? A large prospective study investigates. Clin Rheumatol 2012;31(1):79–83 Coppo P, Clauvel JP, Bengoufa D, Oksenhendler E, Lacroix C, Lassoued K. Inflammatory myositis associated with anti-U1small nuclear ribonucleoprotein antibodies: a subset of myositis associated with a favourable outcome. Rheumatology (Oxford) 2002;41(9):1040–1046 Kim P, Grossman JM. Treatment of mixed connective tissue disease. Rheum Dis Clin North Am 2005;31(3):549–565, viii Vanthuyne M, Blockmans D, Westhovens R, et al. A pilot study of mycophenolate mofetil combined to intravenous methylprednisolone pulses and oral low-dose glucocorticoids in severe early systemic sclerosis. Clin Exp Rheumatol 2007;25(2):287–292 Simeón-Aznar CP, Fonollosa-Plá V, Tolosa-Vilella C, Selva-O’Callaghan A, Solans-Laqué R, Vilardell-Tarrés M. Effect of mycophenolate sodium in scleroderma-related interstitial lung disease. Clin Rheumatol 2011;30(11):1393–1398 Koutroumpas A, Ziogas A, Alexiou I, Barouta G, Sakkas LI. Mycophenolate mofetil in systemic sclerosis-associated interstitial lung disease. Clin Rheumatol 2010;29(10):1167–1168 Gerbino AJ, Goss CH, Molitor JA. Effect of mycophenolate mofetil on pulmonary function in scleroderma-associated interstitial lung disease. Chest 2008;133(2):455–460 Swigris JJ, Olson AL, Fischer A, et al. Mycophenolate mofetil is safe, well tolerated, and preserves lung function in patients with connective tissue disease-related interstitial lung disease. Chest 2006;130(1):30–36 Fischer A, Brown KK, Du Bois RM, et al. Mycophenolate mofetil improves lung function in connective tissue disease-associated interstitial lung disease. J Rheumatol 2013;40(5):640–646 Tzouvelekis A, Galanopoulos N, Bouros E, et al. Effect and safety of mycophenolate mofetil or sodium in systemic sclerosis-associated interstitial lung disease: a meta-analysis. Pulm Med 2012; 2012:143637 Labirua-Iturburu A, Selva-O’Callaghan A, Martínez-Gómez X, Trallero-Araguás E, Labrador-Horrillo M, Vilardell-Tarrés M. Calcineurin inhibitors in a cohort of patients with antisynthetaseassociated interstitial lung disease. Clin Exp Rheumatol 2013; 31(3):436–439 Matsuki Y, Yamashita H, Takahashi Y, et al. Diffuse alveolar damage in patients with dermatomyositis: a six-case series. Mod Rheumatol 2012;22(2):243–248

tacrolimus of progressive interstitial pneumonia associated with amyopathic dermatomyositis refractory to cyclosporine. Clin Rheumatol 2010;29(4):443–445 Polosa R, Edwards CJ. Tacrolimus for antisynthetase syndrome with interstitial lung disease? Eur Respir J 2008;32(1):244–245, author reply 245–246 Guglielmi S, Merz TM, Gugger M, Suter C, Nicod LP. Acute respiratory distress syndrome secondary to antisynthetase syndrome is reversible with tacrolimus. Eur Respir J 2008;31(1): 213–217 Ochi S, Nanki T, Takada K, et al. Favorable outcomes with tacrolimus in two patients with refractory interstitial lung disease associated with polymyositis/dermatomyositis. Clin Exp Rheumatol 2005;23(5):707–710 Oddis CV, Sciurba FC, Elmagd KA, Starzl TE. Tacrolimus in refractory polymyositis with interstitial lung disease. Lancet 1999;353(9166):1762–1763 Cavagna L, Caporali R, Abdì-Alì L, Dore R, Meloni F, Montecucco C. Cyclosporine in anti-Jo1-positive patients with corticosteroidrefractory interstitial lung disease. J Rheumatol 2013;40(4): 484–492 Leandro MJ, Cambridge G, Ehrenstein MR, Edwards JC. Reconstitution of peripheral blood B cells after depletion with rituximab in patients with rheumatoid arthritis. Arthritis Rheum 2006; 54(2):613–620 Isaacs JD, Cohen SB, Emery P, et al. Effect of baseline rheumatoid factor and anticitrullinated peptide antibody serotype on rituximab clinical response: a meta-analysis. Ann Rheum Dis 2013; 72(3):329–336 Keystone EC, Cohen SB, Emery P, et al. Multiple courses of rituximab produce sustained clinical and radiographic efficacy and safety in patients with rheumatoid arthritis and an inadequate response to 1 or more tumor necrosis factor inhibitors: 5year data from the REFLEX study. J Rheumatol 2012;39(12): 2238–2246 Emery P, Fleischmann R, Filipowicz-Sosnowska A, et al; DANCER Study Group. The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trial. Arthritis Rheum 2006;54(5): 1390–1400 Jones RB, Tervaert JW, Hauser T, et al; European Vasculitis Study Group. Rituximab versus cyclophosphamide in ANCA-associated renal vasculitis. N Engl J Med 2010;363(3):211–220 Arnold DM, Dentali F, Crowther MA, et al. Systematic review: efficacy and safety of rituximab for adults with idiopathic thrombocytopenic purpura. Ann Intern Med 2007;146(1): 25–33 Daoussis D, Liossis SN, Tsamandas AC, et al. Effect of long-term treatment with rituximab on pulmonary function and skin fibrosis in patients with diffuse systemic sclerosis. Clin Exp Rheumatol 2012;30(2, Suppl 71):S17–S22 Daoussis D, Liossis SN, Tsamandas AC, et al. Is there a role for B-cell depletion as therapy for scleroderma? A case report and review of the literature. Semin Arthritis Rheum 2010;40(2):127–136 Keir GJ, Maher TM, Hansell DM, et al. Severe interstitial lung disease in connective tissue disease: rituximab as rescue therapy. Eur Respir J 2012;40(3):641–648 Keir GJ, Maher TM, Ming D, et al. Rituximab in severe, treatmentrefractory interstitial lung disease [published online ahead of print November 29, 2013]. Respirology. doi: 10.1111/resp.12214 Giuggioli D, Manfredi A, Colaci M, Manzini CU, Antonelli A, Ferri C. Systemic sclerosis and cryoglobulinemia: our experience with overlapping syndrome of scleroderma and severe cryoglobulinemic vasculitis and review of the literature. Autoimmun Rev 2013;12(11):1058–1063

47

48

49

50

51

52

53

54 55

56

57

58

59

60

61

62

63

Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

272

Maher

81 Braun-Moscovici Y, Butbul-Aviel Y, Guralnik L, et al. Rituximab:

91 Keidel SM, Hoyles RK, Wilkinson NM. Efficacy of tocilizumab for

rescue therapy in life-threatening complications or refractory autoimmune diseases: a single center experience. Rheumatol Int 2013;33(6):1495–1504 Daniels CE, Wilkes MC, Edens M, et al. Imatinib mesylate inhibits the profibrogenic activity of TGF-beta and prevents bleomycinmediated lung fibrosis. J Clin Invest 2004;114(9):1308–1316 Sabnani I, Zucker MJ, Rosenstein ED, et al. A novel therapeutic approach to the treatment of scleroderma-associated pulmonary complications: safety and efficacy of combination therapy with imatinib and cyclophosphamide. Rheumatology (Oxford) 2009; 48(1):49–52 Spiera RF, Gordon JK, Mersten JN, et al. Imatinib mesylate (Gleevec) in the treatment of diffuse cutaneous systemic sclerosis: results of a 1-year, phase IIa, single-arm, open-label clinical trial. Ann Rheum Dis 2011;70(6):1003–1009 Khanna D, Saggar R, Mayes MD, et al. A one-year, phase I/IIa, open-label pilot trial of imatinib mesylate in the treatment of systemic sclerosis-associated active interstitial lung disease. Arthritis Rheum 2011;63(11):3540–3546 Distler JH, Manger B, Spriewald BM, Schett G, Distler O. Treatment of pulmonary fibrosis for twenty weeks with imatinib mesylate in a patient with mixed connective tissue disease. Arthritis Rheum 2008;58(8):2538–2542 De Lauretis A, Sestini P, Pantelidis P, et al. Serum interleukin 6 is predictive of early functional decline and mortality in interstitial lung disease associated with systemic sclerosis. J Rheumatol 2013;40(4):435–446 Liu X, Das AM, Seideman J, et al. The CC chemokine ligand 2 (CCL2) mediates fibroblast survival through IL-6. Am J Respir Cell Mol Biol 2007;37(1):121–128 Moodley YP, Scaffidi AK, Misso NL, et al. Fibroblasts isolated from normal lungs and those with idiopathic pulmonary fibrosis differ in interleukin-6/gp130-mediated cell signaling and proliferation. Am J Pathol 2003;163(1):345–354 Yoshida M, Sakuma J, Hayashi S, et al. A histologically distinctive interstitial pneumonia induced by overexpression of the interleukin 6, transforming growth factor beta 1, or platelet-derived growth factor B gene. Proc Natl Acad Sci U S A 1995;92(21): 9570–9574

interstitial lung disease in an undifferentiated autoinflammatory disorder partially responsive to anakinra. Rheumatology (Oxford) 2013 Mohr M, Jacobi AM. Interstitial lung disease in rheumatoid arthritis: response to IL-6R blockade. Scand J Rheumatol 2011; 40(5):400–401 Thompson B, Corris P, Miller JA, Cooper RG, Halsey JP, Isaacs JD. Alemtuzumab (Campath-1H) for treatment of refractory polymyositis. J Rheumatol 2008;35(10):2080–2082 Horai Y, Miyamura T, Shimada K, et al. Eternacept for the treatment of patients with rheumatoid arthritis and concurrent interstitial lung disease. J Clin Pharm Ther 2012;37(1):117–121 Perez-Alvarez R, Perez-de-Lis M, Diaz-Lagares C, et al. Interstitial lung disease induced or exacerbated by TNF-targeted therapies: analysis of 122 cases. Semin Arthritis Rheum 2011;41(2):256–264 Ramos-Casals M, Brito-Zerón P, Muñoz S, et al. Autoimmune diseases induced by TNF-targeted therapies: analysis of 233 cases. Medicine (Baltimore) 2007;86(4):242–251 Park JK, Yoo HG, Ahn DS, Jeon HS, Yoo WH. Successful treatment for conventional treatment-resistant dermatomyositis-associated interstitial lung disease with adalimumab. Rheumatol Int 2012;32(11):3587–3590 Zong M, Dorph C, Dastmalchi M, et al. Anakinra treatment in patients with refractory inflammatory myopathies and possible predictive response biomarkers: a mechanistic study with 12 months follow-up. Ann Rheum Dis 2013 Furlan A, Botsios C, Ruffatti A, Todesco S, Punzi L. Antisynthetase syndrome with refractory polyarthritis and fever successfully treated with the IL-1 receptor antagonist, anakinra: A case report. Joint Bone Spine 2008;75(3):366–367 Douglas WW, Tazelaar HD, Hartman TE, et al. Polymyositisdermatomyositis-associated interstitial lung disease. Am J Respir Crit Care Med 2001;164(7):1182–1185 Suzuki Y, Hayakawa H, Miwa S, et al. Intravenous immunoglobulin therapy for refractory interstitial lung disease associated with polymyositis/dermatomyositis. Lung 2009;187(3):201–206 Bakewell CJ, Raghu G. Polymyositis associated with severe interstitial lung disease: remission after three doses of IV immunoglobulin. Chest 2011;139(2):441–443

82

83

84

85

86

87

88

89

90

92

93

94

95

96

97

98

99

100

101

102

Seminars in Respiratory and Critical Care Medicine

Vol. 35

No. 2/2014

273

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

CTD-ILD Treatment

Copyright of Seminars in Respiratory & Critical Care Medicine is the property of Thieme Medical Publishing Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Immunosuppression for connective tissue disease-related pulmonary disease.

With improvements in the systemic treatment of the various connective tissue diseases (CTDs) the pulmonary complications of these conditions are now, ...
257KB Sizes 0 Downloads 3 Views