Skip to main content
Drugs in Context logoLink to Drugs in Context
. 2021 Jan 15;10:2020-8-8. doi: 10.7573/dic.2020-8-8

Efficacy and safety of mycophenolate mofetil in the treatment of rheumatic disease-related interstitial lung disease: a narrative review

Giulia Cassone 1,2,3, Marco Sebastiani 2, Caterina Vacchi 1,2, Gian Luca Erre 4, Carlo Salvarani 2,3, Andreina Manfredi 2,
PMCID: PMC7813435  PMID: 33505480

Abstract

Mycophenolate mofetil (MMF) is an antimetabolite with a potent inhibitory effect on proliferation of T and B lymphocytes used since the early 1990s for the prevention of acute allograft rejection after organ transplant. MMF is also widely used for the treatment of a variety of rheumatic diseases (RDs) and their pulmonary involvement. Interstitial lung disease (ILD) is a heterogeneous group of progressive fibrotic diseases of the lung, which is often secondary to RD and represents a major cause of morbidity and mortality. MMF is considered the main alternative to cyclophosphamide as a first-line agent to treat RD-related ILD or as possible maintenance therapy after cyclophosphamide, with a lower rate of side-effects. However, as for other immunosuppressive agents, the use of MMF in RD-ILD is supported by poor scientific evidence. In this narrative review, we describe the available data and recent advances on the effectiveness and safety of MMF for the treatment of ILD related to RD, including rheumatoid arthritis, systemic sclerosis, primary Sjögren syndrome, systemic lupus erythematosus, idiopathic inflammatory myopathies, undifferentiated connective tissue disease, interstitial pneumonia with autoimmune features and antineutrophil cytoplasmic antibody-associated vasculitis.

Keywords: connective tissue diseases, efficacy, interstitial lung disease, lung fibrosis, mycophenolate mofetil, rheumatic diseases, safety

Introduction

Mycophenolate mofetil (MMF) is a prodrug of mycophenolic acid (MPA), an inhibitor of inosine monophosphate dehydrogenase, which inhibits de novo guanosine nucleotide synthesis and exerts a series of immunosuppressive effects.1 MMF has been used since the early 1990s for the prevention of acute allograft rejection and is also widely used for the treatment of a variety of rheumatic diseases (RDs) (dose usually ranging from 1 to 3 g).2,3

Interstitial lung disease (ILD) is a heterogeneous group of progressive fibrotic diseases of the lung, often secondary to RD, and represents a major cause of morbidity and mortality.47 RD-ILDs represent the second most common diagnosis in tertiary ILD referral centres.8 In particular, ILD can complicate connective tissue diseases (CTDs) and rheumatoid arthritis (RA), yet it also complicates antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) and sarcoidosis, with variable prevalence and different grade of severity of pulmonary involvement and mortality rate according to the specific RD.5,917 Moreover, increasing interest and a deeper knowledge have been emerged regarding a subgroup of patients with ILD and clinical and/or serological findings suggestive but not diagnostic for a definite RD, defined as interstitial pneumonia with autoimmune features (IPAF).1820

RD-ILD can be characterized by all the histological/radiological patterns described for idiopathic interstitial pneumonias.4,21 Some authors speculated about more favourable responses to immunosuppressive therapy for non-specific interstitial pneumonia (NSIP) or organizing pneumonia patterns. However, no strong evidence-based data support this hypothesis.22,23

The pathogenesis of ILD in CTD, AAV, and IPAF shares some similarities and is substantially characterized by long-term and aggressive systemic inflammation and immune activation with consecutive damage to lung tissues and the development of a profibrotic microenvironment.15,2428 The main actors of the fibrotic process are inflammatory cytokines, activated macrophages, fibroblasts, T and B cells, adaptive immunity and antibodies, and reactive oxygen species.15,2428 Usual interstitial pneumonia (UIP) in RD-ILD also shows analogies with idiopathic pulmonary fibrosis (IPF), in particular regarding their natural history and clinical behaviour with a progressive fibrosing phenotype.2931 Moreover, the same genetic predisposition has been described for the IPF and UIP patterns in RA-associated and AAV-associated ILDs.3135 Finally, similarly to IPF, patients with RD-ILD may also experience an acute exacerbation (AE).36

The management of ILD in patients with RD is challenging and could be decisive to improve their quality of life and decrease mortality and the high utilization of healthcare resources. However, due to the paucity of randomized controlled trials (RCTs) and the substantial heterogeneity in disease behaviour, the therapeutic choice for RD-ILD is currently based on an empirical approach dependent on the personal experience and expertise of the medical team. The best available evidence has been generated in systemic sclerosis-associated ILD (SSc-ILD).37,38

Several therapeutic agents have been suggested and current treatment is essentially based on immunosuppression. The scientific background supporting the use of an immunosuppressive drug in RD-ILD comprises a direct anti-inflammatory effect on the primary aetiopathogenetic process of ILD and an indirect effect by decreasing the RD activity, which could influence the ILD progression. Recently, the use of antifibrotic agents has also been proposed.30,37,38

The pharmacological immunosuppressive properties of MMF, as described (see pharmacodynamic effects section) support the scientific rationale for the use of MMF as a treatment for severe RD-ILD. Indeed, MMF is usually considered the main alternative to cyclophosphamide (CYC) as a first-line agent to treat RD-ILD or as possible maintenance therapy after CYC, with a lower rate of side-effects.3,3942

In this review, we describe the available data and recent advances on the effectiveness and safety of MMF for the treatment of ILD related to RD, including RA, SSc, primary Sjögren syndrome (pSS), systemic lupus erythematosus (SLE), idiopathic inflammatory myopathies (IIMs), undifferentiated CTD (UCTD), IPAF, and AAV (Table 1).

Table 1.

Available evidence for the use of mycophenolate mofetil in RD-ILD.

Author, year (ref.) Article type Number of patients
Rheumatoid arthritis
Fischer et al., 2013 (40) Retrospective 18
Oldham et al., 2016 (66) Retrospective 8
Saketkoo et al., 2008 (41) Case series 3
Systemic sclerosis
Tashkin et al., 2016 (42) RCT: SLS II 63
Volkmann et al., 2017 (70) RCT: SLS I–II 69
Naidu et al., 2020 (71) RCT 20
SLS III (NCT03221257) (94) RCT: SLS III NA
Stratton et al., 2001 (73) Prospective 13
Liossis et al., 2006 (85) Prospective 6
Vanthuyne et al., 2007 (86) Prospective 16
Derk et al., 2009 (82) Prospective 15
Simeón-Aznar et al., 2011 (83) Prospective 14
Mendoza et al., 2012 (81) Prospective 25
Panopoulos et al., 2013 (92) Case–control 26
Tzouvelekis et al., 2012 (77) Retrospective + systematic review 10 (total 69)
Nihtyanova et al., 2007 (78) Retrospective 109
Zamora et al., 2008 (88) Retrospective 17
Gerbino et al., 2008 (87) Retrospective 13
Koutroumpas et al., 2010 (74) Retrospective 10
Le et al., 2011 (75) Retrospective 98
Owen et al., 2016 (89) Retrospective 18
Baqir et al., 2017 (91) Retrospective 46
Adler et al., 2018 (79) Retrospective NA
Saketkoo et al., 2009 (68) Case series 4
Yilmaz et al., 2014 (80) Case series 12
Herrick et al., 2010 (84) NA NA
Primary Sjögren syndrome
None
Idiopathic inflammatory myopathies
Morganroth et al., 2010 (106) Retrospective 16
Mira-Avendano et al., 2013 (103) Retrospective 9
Hanaoka et al., 2019 (104) Retrospective 19
Huapaya et al., 2019 (105) Retrospective 44
Cozzani et al., 2013 (110) Case report 1
Girard et al., 2013 (128) Case report 1
Sundaragiri et al., 2014 (116) Case report 1
Tsuchiya et al., 2014 (109) Case report 1
Kulkarni et al., 2015 (117) Case report 1
Gil et al., 2016 (108) Case report 1
Hayashi et al., 2017 (126) Case report 1
Hisanaga et al., 2017 (127) Case report 1
Koyama et al., 2017 (107) Case report 1
Ruegg et al., 2019 (115) Case report 1
Systemic lupus erythematosus
Al Rashidi et al., 2011 (132) Case report 1
UCTD and IPAF
McCoy et al., 2018 (135) Retrospective 28
ANCA-associated vasculitis
None
Sarcoidosis
Brill et al., 2013 (147) Retrospective 10
Hamzeh et al., 2014 (146) Retrospective 37
Papiris et al., 2019 (145) Retrospective 8
Other articles: cumulative data on more diseases or drugs
Zhang et al., 2015 (67) RCT 23 RD-ILD
Swigris et al., 2006 (39) Retrospective 28 RD-ILD
Saketkoo et al., 2009 (68) Retrospective 10 RD-ILD
Fischer et al., 2013 (40) Retrospective 125 RD-ILD

ANCA, antineutrophil cytoplasmic antibody; ILD, interstitial lung disease; IPAF, interstitial pneumonia with autoimmune features; NA, not available; RCT, randomized controlled trials; RD, rheumatoid disease; SLS, Scleroderma Lung Study; UCTD, undifferentiated connective tissue disease.

Methodology and study selection

A systematic literature review was conducted by two authors (GC and CV) using PubMed, Web of Science, Scopus, Embase, Latin American and Caribbean Health Sciences Literature, and Cochrane Central databases. The research strategy was ((mycophenolate mofetil* OR mycophenolate sodium*) AND (interstitial lung disease* OR interstitial pneumonia* OR lung fibrosis*) AND (rheumatoid arthritis* OR connective tissue diseases* OR systemic sclerosis* OR primary Sjögren syndrome* OR systemic lupus erythematosus* OR idiopathic inflammatory myopathies* OR undifferentiated CTD* OR interstitial pneumonia with autoimmune features* OR ANCA-associated vasculitis* OR small vessel vasculitis* OR sarcoidosis*)) in the text, title, and abstract fields. We considered RCTs, systematic reviews, observational studies, case series, and case reports. We did not consider abstract or grey literature. We also used the snowballing technique to search the bibliographies for relevant references from the reference list or moving forward to the citing articles. No language restriction was considered, no years of publication restriction were applied, and only published articles were considered. A narrative review was conducted due to the low quality of available studies, consisting mainly of case reports or case series and retrospective studies.

Pharmacological properties of MMF

Clinical pharmacokinetics

MMF is an ester of MPA and was synthesized to increase the bioavailability of MPA. Following administration, MMF is rapidly and completely absorbed and is entirely metabolized by liver carboxylesterases 1 and 2 to MPA by a pre-systemic de-esterification. MPA is almost completely metabolized by the enzyme glucuronyl transferase to the pharmacologically inactive and stable phenolic glucuronide MPAG, which is excreted in urine and represents almost all of the administered dose. The glucuronide metabolite is then converted to MPA through enterohepatic recirculation. MMF escaping metabolism in the intestine enters the liver via the portal vein and is transformed to pharmacologically active MPA in the liver cells. In addition to MPAG, other major metabolites of MPA are MPA acyl-glucuronide, 7-O-MPA glucoside, and small amounts 6-O-des-methyl-MPA.

Almost all of the whole administered dose is excreted in the urine as MPAG. The average apparent half-life of MMF is 17.9 (±6.5) hours after oral administration and 16.6 (±5.8) hours after intravenous administration. Plasma clearance of MMF is 193 mL/min after an oral dose and 177 (±31) mL/min after an intravenous dose. Effectively, oral MMF is 100% bioavailable as MPA in healthy individuals. These properties of MMF lead to a small intra-individual and inter-individual variability for plasma MPA and to predictable pharmacokinetics changes in pathophysiological situations.43,44

Pharmacodynamics

The immunosuppressive effects of MMF are mainly derived from its cytostatic effect on T and B lymphocytes and, hence, on the inhibition of antibody production. Three other mechanisms may also contribute to the anti-inflammatory activity of MMF.13,45 First, MMF can induce apoptosis of activated T lymphocytes and suppress the T lymphocytic response to allogeneic cells and other antigens. MPA also suppresses dendritic cell maturation decreasing their capacity of antigen presentation to T lymphocytes. Second, MMF inhibits the glycosylation and expression of adhesion molecules as well as the recruitment of lymphocytes and monocytes into sites of inflammation. Third, by depleting tetrahydrobiopterin, MMF decreases the production of NO by inducible NO synthase and the consequent tissue damage mediated by peroxynitrite. Moreover, by decreasing the recruitment of monocyte-macrophage lineage cells, MMF decreases the production of TNFα and IL-1, both of which are cytokines implicated in the recruitment and proliferation of fibroblasts.

Adverse effects and contraindications

MMF is usually safer and better tolerated than CYC. Unlike azathioprine (AZA), the deficiency in thiopurine methyltransferase is not a potential concern when prescribing MMF. On the contrary, MMF should theoretically be avoided in patients with the rare hereditary deficiency of hypoxanthine-guanine phosphoribosyl transferase (e.g. Lesch–Nyhan or Kelley–Seegmiller syndromes).

Adverse effects and monitoring

Gastrointestinal (GI) symptoms and bone marrow suppression are the most frequent adverse effects of MMF. Other common side-effects include hepatitis and increased risk of infections. A higher risk of cancer and progressive multifocal leukoencephalopathy have also been described.46,47

Common and uncommon side-effects of MMF are listed in Table 2. GI side-effects are usually dose related and tend to reduce over time. In patients with GI intolerance to MMF (mainly diarrhoea), the enteric-coated mycophenolate sodium can be an alternative formulation. However, it has not been formally tested in any controlled studies, and some authors demonstrated similar rates of GI side-effects for both drugs.48,49 In some patients, compliance improves by increasing the number of divided daily doses (maintaining the same total daily dose). Other patients may require dose adjustments. In patients with active peptic ulcer or other GI disease, such as inflammatory bowel diseases, MMF should be used with caution.

Table 2.

Side-effects of mycophenolate mofetil. Incidences include concomitant use of corticosteroids and other immunosuppressants.

>10% 1% to 10% Rare
Cardiovascular Hypertension, hypotension, tachycardia, lower extremity oedema Exacerbation of hypertension, peripheral oedema, phlebitis, thrombosis Endocarditis, venous thrombosis
Central nervous system Pain, headache, insomnia, dizziness, depression, chills, confusion, drowsiness, hypertonia, malaise, myasthenia, paraesthesia Anxiety, fatigue Meningitis, progressive multifocal leukoencephalopathy
Dermatologic Skin rash, ecchymoses, cellulitis Acne vulgaris, pruritus Alopecia, hypersensitivity reaction, Kaposi sarcoma
Endocrine and metabolic Hyperglycaemia, hypercholesterolaemia, hypomagnesaemia, hypokalaemia, hypocalcaemia, increased lactate dehydrogenase, hyperkalaemia, acidosis, weight loss, hyperuricaemia, hyperlipidaemia, hypophosphataemia Diabetes mellitus
Gastrointestinal Abdominal pain, nausea, diarrhoea, constipation, vomiting, decreased appetite, dyspepsia, oesophagitis, gastric ulcer, gastritis, gastrointestinal haemorrhage, hernia of abdominal cavity, intestinal obstruction, stomatitis, upper abdominal pain, flatulence Abdominal distension, gastroesophageal reflux disease, gingival hyperplasia, oral candidiasis Mucocutaneous candidiasis, anorexia, colitis, duodenal ulcer, oesophageal ulcer, gastrointestinal perforation, haematemesis, haemorrhagic colitis, haemorrhagic gastritis, melena, pancreatitis, peritonitis
Genitourinary Urinary tract infection, haematuria Urinary retention
Haematologic and oncologic Leukopenia, anaemia, leukocytosis, thrombocytopenia, benign skin neoplasm, disorder of haemostatic components of blood, neoplasm, pancytopenia, skin carcinoma Lymphocele, severe neutropenia, malignant neoplasm, malignant lymphoma, lymphoproliferative disorder Agranulocytosis, bone marrow failure, hypogammaglobulinaemia lymphadenopathy, lymphopenia, pure red cell aplasia
Hepatic Increased liver enzymes, hepatitis, increased serum alkaline phosphatase Abnormal hepatic function tests
Infection Bacterial infection, viral infection, cytomegalovirus disease, fungal infection Influenza, wound infection, herpes simplex infection, herpes zoster infection, sepsis Protozoal infection, atypical mycobacterial infection, BK virus, polyomavirus infection, reactivation of Hepatitis C virus (HCV), reactivation of Hepatitis B virus (HBV), tuberculosis
Neuromuscular and skeletal Asthenia, tremor, back pain, arthralgia Muscle cramps, myalgia, peripheral pain Osteomyelitis
Renal Increased serum creatinine, increased blood urea nitrogen Renal insufficiency, renal tubular necrosis
Respiratory Dyspnoea, cough, pleural effusion Dyspnoea on exertion, nasopharyngitis, pneumonia, sinusitis, upper respiratory tract infection Pharyngitis, respiratory tract infection, bronchiectasis interstitial pulmonary disease, pulmonary oedema, pulmonary fibrosis, wheezing, xerostomia
Miscellaneous Fever

This table is adapted from the Medication Guide of MMF.44

Cytopenia is the major potential concern and requires regular monitoring, which should also include renal and liver function and signs of lymphoma or myeloproliferative disorders.50 The risk for malignancies, mainly lymphoproliferative disorders, is related to the intensity and duration of therapy. Moreover, even if a direct association between lung fibrosis and cancer is still missing, emerging evidence, mainly in IPF, suggest that progressive lung fibrosis represents a risk factor for lung cancer development. In IPF, lung cancer often occurs in the peripheral areas and lower lobes where fibrotic changes are predominant.51 Furthermore, radiologic features of RD-ILD and lung cancer can overlap substantially. Thus, the interpretation of chest high-resolution computer tomography (HRCT) can be difficult and should be done with caution, notably regarding atypical nodes or masses and/or lymph nodes.52

There are no large studies examining the incidence of infections in patients with rheumatic illness treated with MMF. The existing studies53,54 are too small to allow generalizations. On the other hand, data from larger studies regarding solid organ transplants showed conflicting results.5557

MMF has antimicrobial properties and seems to exert a protective effect against Pneumocystis jirovecii.58,59 Therefore, the use of prophylaxis for Pneumocystis pneumonia is controversial. The association between MMF and possible viral infections, such as herpes zoster and cytomegalovirus infection, is also disputable.6063

Nevertheless, patients with RD are not directly comparable to patients with solid organ transplants. In fact, in patients with RD, the increased risk of infections can derive from the exposure to other immunosuppressive agents and to higher cumulative doses of corticosteroids (CSs).64 Furthermore, the aetiopathogenetic alteration of the immune system in RD may lead itself to an increased susceptibility to infections.

In addition, pulmonary infections can be more frequent and more severe in a context of ILD.64 Infections have been suggested to play a role both in the pathogenesis of ILD and as potential triggers of AE, mainly in IPF. Thus, diagnosis and treatment of acute lower respiratory tract infections as early as possible is required to prevent a life-threatening condition like AE. In this context, the use of an immunosuppressive drug such as MMF, especially in combination with high-dose steroids, may increase the risk of severe pulmonary complications and mortality in patients with RD-ILD.65

For all these reasons, vaccinations, excluding live attenuated vaccines, are highly recommended in patients with rheumatic conditions who take MMF.

Contraindications

MMF has been associated with an increased risk of congenital abnormalities and should be avoided during pregnancy. Reliable contraception should be employed by women of childbearing age. MMF is excreted in breast milk and is contraindicated during breastfeeding. A pregnancy test should be performed immediately prior to initiation and 8–10 days later in females of childbearing age, followed by repeat tests during therapy.

Rheumatoid arthritis

ILD is the most common manifestation of lung involvement in RA. Unlike CTD-ILD, the most common histopathologic type is UIP. In addition to the histological/radiological pattern, RA-ILD shares many other analogies with IPF. It shows a similar clinical behaviour, often with a progressive fibrosing phenotype, and a comparable prognosis and survival.2931

Evidence-based use of MMF in RA-ILD is still missing; moreover, it is ineffective for the articular manifestations of the disease. No controlled studies are available to recommend the use of MMF in RA-ILD.40,41,6668 Saketkoo et al.68 described a clinical improvement in physiological lung assessment and radiological stabilization in a small case series of three patients with RA-ILD. In 2016, a retrospective study from the UK observed a better survival of patients with RA-ILD treated with MMF than with AZA.69 The relative risk of death for any cause was increased in patients treated with prednisone, whereas it was unaltered for AZA and decreased for MMF. The authors suggested a better outcome following treatment with MMF rather than with CSs or AZA in patients with RA-ILD. In a series of 125 patients with CTD-ILD, including 18 with RA, MMF was associated with modest improvements in forced vital capacity (FVC) and in diffusing capacity of the lungs for carbon monoxide (DLCO) and a reduction in the prednisone dose.40 Finally, in 2016, Oldham et al.66 compared the use of AZA and MMF in patients with fibrotic CTD-ILD, including 15 patients with RA-ILD. Both groups demonstrated pulmonary function stability over time, with the AZA group demonstrating a marginal improvement but much more side-effects.

Connective tissue diseases

MMF is one of the most common immunosuppressive agents currently used for the treatment of CTD-ILD. However, there have been no prospective studies about the safety or efficacy of this approach. The best available evidence has been generated only in a small number of RCTs for SSc-ILD.37,42,70,71

MMF demonstrated stability of lung function and a low rate of adverse events in a small cohort of patients with mixed CTD-ILD.39,40,67,68 In another longitudinal retrospective study of 125 patients with CTD-ILD, MMF seemed to either stabilize or improve FVC and DLCO over a median of 2.5 years of follow-up, with a low rate of discontinuation.40 In CTDs and vasculitides, MMF is largely used for the treatment of systemic manifestations other than lung involvement as well as for maintenance after induction therapy with other immunosuppressants such as CYC. Unfortunately, it is not effective in treating joint arthritis.

Systemic sclerosis

ILD is a frequent complication of SSc, often progressive and with a poor prognosis. The most common histopathologic subtype is NSIP.72 However, the optimal treatment and the therapy’s timing for SSc-ILD is still area of uncertainty. Since 2001, many retrospective reviews and small prospective case series68,7388 have assessed the role of MMF in SSc-ILD, providing encouraging results in terms of improvement or stability in lung function and symptoms as well as a good safety profile.

In 2015, Omair et al.76 performed a systematic review to evaluate the GI adverse events of MMF in patients with SSc. The secondary end-point was the evaluation of the effectiveness of the drug on lung disease in terms of pulmonary function tests (PFTs). Among 13 studies included, 7 observational studies reported improvement or stabilization in FVC.68,7376,8188 According to Omair et al.,76 the Australian Scleroderma Cohort Study and Japanese Respiratory Society observed stability or improvement in FVC in patients with SSc.89,90 In 2017, another retrospective study of 46 patients with SSc-ILD treated for at least 1 year showed that the use of MMF slowed the rate of decline of lung function, even at doses lower than 3 g/day.91 On the contrary, Panopoulos et al.92 cautioned about replacing CYC with MMF in SSc-associated ILD. A deterioration of lung HRCT findings at 2 years was noticed in the MMF group but not after CYC, even if the CYC group had more extended ILD at baseline. Recently, a network meta-analysis compared the efficacy and safety of different treatments in SSc-ILD. Compared to the placebo, MMF did not significantly reduce FVC decline nor DLCO.93

To date, only two RCTs have evaluated the safety and efficacy of MMF in patients with SSc-ILD.42,70 The Scleroderma Lung Study (SLS) II included 142 patients with moderate-to-severe SSc-ILD. It compared MMF 3 g daily for 2 years with oral CYC for 1 year finding no differences in efficacy but better tolerance to MMF.42 The average FVC presented a modest decline between 21 and 24 months of follow-up in both arms, and a complete loss of efficacy was not observed in the CYC arm, as occurred in SLS I. Notably, DLCO decreased in both groups, although this finding was significantly greater in the CYC versus the MMF group. The study also demonstrated a greater safety and tolerability of MMF compared with CYC, although no differences in the incidence of infection, bleeding, or death were recorded.42 Volkmann et al. compared the outcomes for the MMF arm of SLS II with the placebo arm of SLS I.70 Considering the limits inherent in the design of the study, MMF showed a long-lasting efficacy on PFT parameters and dyspnoea and showed a high safety profile.

Recently, a double-blind, randomized, placebo-controlled, pilot trial conducted at a tertiary care hospital in north India (NCT02896205) aimed to assess the efficacy and safety of MMF in patients with SSc-ILD and mildly impaired lung function (FVC ≥70% predicted); 41 patients were included in the study, and treatment with MMF did not result in a significant improvement of lung function over 6 months.71 Finally, SLS III (NCT03221257) is now ongoing to compare the efficacy of MMF alone or in combination with pirfenidone in patients with active and symptomatic SSc-ILD. The estimated study completion date is in March 2022 and no preliminary data are available yet.94

Primary Sjögren syndrome

ILD is the most frequent pulmonary manifestation of pSS. Although not frequent, lymphoid interstitial pneumonia is the radiological/histological pattern most closely associated with pSS. Recent studies indicated UIP as the most frequent pattern in pSS patients.95 Lung hematologic malignancies have also been described.96 Management strategies for Sjogren-associated lung diseases (pSS-ILD) remain empiric because no controlled studies have been performed.

The few available data about MMF in pSS are extrapolated from retrospective studies describing mixed cohorts of patients with CTD-ILD. These small case series suggest that MMF may be effective and safe on lung function. Moreover, it could have a glucocorticoid-sparing effect.39,40,67,68

Idiopathic inflammatory myopathies

ILD represents the most common non-muscoloskeletal manifestation of IIMs.97 The majority of patients with IIM who develop ILD have a clinical and histopathological pattern of NSIP or organizing pneumonia; thus, immunosuppressants are usually the first therapeutic choice.97,98 No evidence-based guidelines exist regarding IIM-ILD therapy regimens.99 Most patients follow a chronic, slowly progressive course that does not require specific treatment. However, early evidence suggests that some subtypes of antibodies correlate with the development of ILD and with worse severity,98,100102 as mentioned below. Immunosuppressants are usually the first therapeutic choice in those patients.

A retrospective review of treatment outcomes in IIM-ILD reported the same efficacy in stabilizing lung function and in glucocorticoid dose tapering for oral CYC, AZA, and MMF.103 In 2019, Hanaoka et al. evaluated the efficacy and tolerability of MMF alone (12 patients) or associated to calcineurin inhibitors (7 patients) in resistant inflammatory myopathy. No significant improvement in %FVC and HRCT images and no differences in death or ILD progression were found in patients with ILD in either group.104 In the largest cohort of IIM-ILD treated with MMF,105 44 patients showed improvement in FVC and in reaching lower prednisone dose, but no improvement of the DLCO was detected. Apart from these studies, only sporadic small case series and case reports are available on this topic.39,40,67,68,106110

Antisynthetase syndrome

ILD is a hallmark of antisynthetase syndrome (ASSD), with a prevalence ranging from 67% to 100% of cases.111 This IIM is characterized by the presence of antiaminoacyl-tRNA synthetase antibodies.112,113 Anti-Jo1 positivity has been shown to have a favourable prognostic value; on the contrary, anti-PL7 and anti-PL12 autoantibodies are often associated with a more aggressive ILD and a poor survival.100,101,114 In the study by Mira-Avendano et al.,103 approximately 50% of the patients were positive for anti-Jo1. As mentioned earlier, the use of CYC, AZA, or MMF was similarly associated with the stability of PFTs, a reduction in dyspnoea, and the steroid dose. Other anecdotal case reports showed similar results in patients with ASSD positive for anti-Jo1 antibodies.115117

No studies have directly evaluated the efficacy and safety of MMF in non-anti-Jo1 ASSD patients. Non-anti-Jo1 antisynthetase antibodies (anti-KS, anti-OJ, anti-EJ, anti-PL-7, anti-PL-12) are less frequent in ASSD. Therefore, evidence for these specific subgroups is very poor. However, many patients with these autoantibodies are included in studies on CTD-ILD or IIM-ILD. It has been suggested that patients with anti-KS and anti-OJ antibodies would be most likely to have a good response to CSs.118,119

Antimelanoma differentiation-associated gene 5 (MDA5) dermatomyositis

Anti-MDA5-positive dermatomyositis is characterized by an elevated risk of ILD with a rapidly progressive and potentially fatal course.120124 The 6-month survival rate in some studies is 40% despite therapies.125 In 2017, Hayashi et al.126 described the case of a patient with anti-MDA5-ILD successfully treated by the addition of MMF to an immunosuppressive therapy including CSs, oral cyclosporine, and intravenous CYC. The same year, Hisanaga et al.127 presented a case of worsening ILD despite treatment with high-dose prednisolone combined with cyclosporine and intravenous CYC in a patient positive for anti-MDA5. The addition of direct hemoperfusion with polymyxin-B, MMF, intravenous immunoglobulin, and rituximab (RTX) led to remission of the disease. On the other hand, Girard et al.128 reported a case of anti-MDA5-ILD treated with intravenous immunoglobulins, CYC, MMF, AZA, and RTX in combination with oral CSs, without any improvement in respiratory function. Gil et al.108 described a case series of patients with clinically amyopathic dermatomyositis-ILD, including a patient with anti-MDA-5 antibodies who received MMF and intravenous immunoglobulins. The patient died of pneumonia 30 months after initial presentation.

Systemic lupus erythematosus

ILD can rarely complicate SLE,129,130 and evidence for the treatment of SLE-ILD is of low quality, as no clinical trial or guidelines are available. A consensus conference on the management of SLE in 2015 proposed the use of MMF as induction therapy in association with corticosteroids in patients with lung involvement. MMF was also indicated as maintenance strategy or for the treatment of mild-to-moderate disease.131 Only one case report described the use of MMF for SLE-related diffuse alveolar haemorrhage, with some efficacy as a maintenance therapy (no further diffuse alveolar haemorrhage episodes).132 Only one of ten patients with CTD-ILD had a diagnosis of SLE in the case series by Saketkoo et al.,68 while Fisher et al.40 included four patients with SLE-ILD in their retrospective study.

Undifferentiated CTD

ILD can be a clinical manifestation of UCTD, even if the available classification criteria do not consider lung manifestations in UCTD.133 The management of ILD-UCTD is usually based on immunosuppression, including MMF. However, no evidence-based therapeutic regimens are available to date.

Interstitial pneumonia with autoimmune features

IPAF is a clinical condition characterized primarily by ILD associated to other features (clinical, serological, and/or morphological) suggestive of a CTD that does not meet established classification criteria for a given autoimmune disease.18 For IPAF, both the use of immunosuppressants134137 and antifibrotic agents138140 have been proposed. Unfortunately, to date, no controlled clinical trials are available to guide evidence-based therapeutic regimens. McCoy et al.135 recently described a retrospective case–control series of 28 patients with IPAF exposed to MMF. Changes in FVC% and DLCO% between the MMF-treated and -untreated groups were not significantly different. In patients treated with MMF, FVC and DLCO slightly improved after exposure to the drug but without statistical significance.

ANCA-associated vasculitis

Pulmonary involvement is frequently observed in AAV patients, and ILD is an emerging possible phenotype. The prevalence of ILD is higher in microscopic polyangiitis than in granulomatosis with polyangiitis, and anti-MPO antibodies are the main ANCA subtype associated to ILD. Lung fibrosis in oeosinophilic granulomatosis with polyangiitis or associated to anti-PR3 is rare.141 Cases of patients positive for ANCA, mainly MPO-ANCA, without vasculitis and concomitant ILD have also been reported in the literature. Only retrospective case series and a few case reports have been published and no controlled clinical trials are available to guide the treatment of AAV-ILD or ANCA-positive ILD.

Despite contrasting data, standard treatment of systemic vasculitis is also considered for ILD and includes systemic glucocorticoids with or without immunosuppressants.141,142 MMF is mainly used both as remission and maintenance therapy143 and its efficacy was demonstrated in all the clinical aspects of AAV. Immunosuppressive agents, including MMF, are also considered for the treatment of NSIP pattern, despite the presence of systemic vasculitis.141

Sarcoidosis

Sarcoidosis is a granulomatous systemic disease of unknown aetiology. Pulmonary involvement is present in about 90% of cases. Usually, pulmonary sarcoidosis is self-limited and does not require treatment. However, some patients may develop chronic progressive pulmonary involvement with fibrotic alterations, requiring long-term therapy with CS and/or other immunosuppressants as CS-sparing agents.144

The effect of MMF on chronic pulmonary sarcoidosis has been poorly investigated. A retrospective study evaluated the effectiveness and safety of MMF in eight patients with both pulmonary and extrapulmonary sarcoidosis. A statistically significant improvement in FVC was reported, and symptoms and chest radiological findings improved in all patients.145 On the contrary, another retrospective study of 37 patients with sarcoidosis found no statistically significant changes in PFT or DLCO measurements both before and after MMF therapy.146 Finally, Brill et al.147 retrospectively investigated the efficacy of MMF and systemic CSs in ten patients with biopsy-proven chronic pulmonary sarcoidosis. Pulmonary function, symptoms, and radiological signs improved in four patients, while six patients remained stable after 6 months.

Discussion

Although the pathogenesis of RD-related pulmonary disease is poorly understood, there is an assumption that it arises as a sequela of immune-mediated injury to the lung. As a result, immunosuppressive agents still represent the mainstay of treatment for RD-ILD.

MMF is an antimetabolite with a potent inhibitory effect on proliferation of T and B lymphocytes. MMF is well known and widely used since the early 1990s as immunosuppressant to treat oncologic disorders and to prevent acute allograft rejection after organ transplant.

As for other immunosuppressive agents in relation to RD-ILD, the use of MMF is supported by poor scientific evidence. Its efficacy has only been demonstrated by three RCTs in patients with SSc, with questionable results.42,70,71 Furthermore, MMF is often used in association with CSs with high heterogeneity in drug dosages and timing, making it difficult to extrapolate efficacy data on the single drug. Of note, all patients evaluated in retrospective studies describing the use of MMF in mixed RD-ILD were treated with concomitant CSs.39,40,67,68

Numerous other therapies have been proposed for RD-ILD, including novel agents such as antifibrotics, and biologic and non-biologic disease-modifying antirheumatic drugs. Clinical trials are ongoing.37,38 However, the manageable use of the ‘old drug’ MMF, its low rate of side-effects, the clinicians’ decades of experience in its use in real life, and the lack of other evidence make MMF a preferred therapeutic option for the treatment of severe forms of RD-ILD. The decision to start therapy in patients with RD-ILD should be evaluated in the single patient, balancing comorbidities, the possible adverse effects of treatment, and disease prognosis in each patient.

Moreover, the immunosuppressants historically used for the treatment of ILD, such as MMF as well as CYC and AZA, are usually of low efficacy for the articular manifestations of most RD. CYC is widely used in the treatment of RD-ILD, and it is usually the first choice in patients with rapidly progressive ILD. A recent systematic review found that a small benefit may be derived from the use of CYC in CTD-ILD when compared with placebo but not when compared with MMF.148 In particular, no significant impact on health-related quality of life, all-cause mortality, dyspnoea, or cough severity was found in the CYC group compared with the MMF group. Only four RCTs were included in the analysis, mostly on SSc, and the evidence was found to be of low quality as dropout rates were high in the intervention groups. Moreover, the risk of side-effects was increased with CYC versus MMF, in particular leukopenia and thrombocytopenia.

Among the disease-modifying antirheumatic drugs, the use of RTX has been suggested for RD-ILD, mainly in case reports and retrospective uncontrolled studies, showing encouraging short-term and long-term results with an acceptable safety profile.37,38 Recently, Atienza-Mateo et al.149 published a retrospective single centre study focusing on RTX in the treatment of RD-ILD. They found a sustained improvement in PFTs and a statistically significant increase in DLCO in patients treated with RTX, regardless of the radiological pattern or the underlying RD. However, only one RCT including eight patients with SSc-ILD150,151 and a nested case–control study152 have been published. No results from the RECOVER and RECITAL trials are yet available.153,154

The use of abatacept, tocilizumab, and Jak inhibitors for RD-ILD has also been proposed in anecdotal reports.37,38,155157

Another point to consider is the wide spectrum of clinical phenotypes and the heterogeneity in disease behaviour of RD as well as of the pulmonary involvement in RD that does not allow us to make generalized dissertations on this topic. Different diseases may benefit from different therapeutic approaches or different timing of treatment. The clinical behaviour of pulmonary and extrapulmonary manifestation, the presence of comorbidities, and the potential adverse effects of treatments globally influence the therapeutic approach to the patient. Therefore, treatment should be based on the balance between possible benefits and burden of disease in each single patient. As a result, a multidisciplinary evaluation including at least a rheumatologist and pulmonologist, and possibly a radiologist, with expertise in ILD, is always recommended. A deeper knowledge on how to treat such patients requires clear insights into the pathogenesis of RD-ILD and the availability of RCTs – both needs are still unmet.

Conclusion

Further prospective, randomized, controlled clinical studies are required to better define the long-term efficacy and safety of MMF in patients with ILD associated to RD. They should be adequately powered to compare outcomes specifically within different subgroups and different diseases and stratified for histological subtype, disease duration, and extent of pulmonary involvement. Researchers may consider comparing MMF (as other immunosuppressants) versus other drugs such as antifibrotic agents, or comparing both versus placebo, in particular for those patients with evidence of rapidly progressive fibrotic disease.

Acknowledgements

None.

Footnotes

Contributions: All authors contributed equally to the preparation of this review. All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Disclosure and potential conflicts of interest: The authors declare that they have no conflicts of interest relevant to this manuscript. The International Committee of Medical Journal Editors (ICMJE) Potential Conflicts of Interests form for the authors is available for download at: https://www.drugsincontext.com/wp-content/uploads/2020/12/dic.2020-8-8-COI.pdf

Funding declaration: There was no funding associated with the preparation of this article.

Correct attribution: Copyright © 2021 Cassone G, Sebastiani M, Vacchi C, Erre GL, Salvarani C, Manfredi A. https://doi.org/10.7573/dic.2020-8-8. Published by Drugs in Context under Creative Commons License Deed CC BY NC ND 4.0.

Provenance: Invited; externally peer reviewed.

Drugs in Context is published by BioExcel Publishing Ltd. Registered office: Plaza Building, Lee High Road, London, England, SE13 5PT.

BioExcel Publishing Limited is registered in England Number 10038393. VAT GB 252 7720 07.

For all manuscript and submissions enquiries, contact the Editorial office editorial@drugsincontext.com

For all permissions, rights and reprints, contact David Hughes david.hughes@bioexcelpublishing.com

References

  • 1.Lipsky JJ. Drug profile mycophenolate mofetil. Lancet. 1996;348:1357–1359. doi: 10.1016/S0140-6736(96)10310-X. [DOI] [PubMed] [Google Scholar]
  • 2.Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. Immunopharmacology. 2000;47(2–3):85–118. doi: 10.1016/S0162-3109(00)00188-0. [DOI] [PubMed] [Google Scholar]
  • 3.Broen JCA, van Laar JM. Mycophenolate mofetil, azathioprine and tacrolimus: mechanisms in rheumatology. Nat Rev Rheumatol. 2020;16(3):167–178. doi: 10.1038/s41584-020-0374-8. [DOI] [PubMed] [Google Scholar]
  • 4.Demedts M, Costabel U. American Thoracic Society American Thoracic Society/European Respiratory Society International Multidisciplinary Consensus Classification of the Idiopathic Interstitial Pneumonias. Eur Respir J. 2002;19(5):794–796. doi: 10.1183/09031936.02.00492002. [DOI] [PubMed] [Google Scholar]
  • 5.Kolb M, Vašáková M. The natural history of progressive fibrosing interstitial lung diseases. Respir Res. 2019;20:57. doi: 10.1186/s12931-019-1022-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cottin V, Hirani NA, Hotchkin DL, et al. Presentation, diagnosis and clinical course of the spectrum of progressive-fibrosing interstitial lung diseases. Eur Respir Rev. 2018;27(150) doi: 10.1183/16000617.0076-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wallis A, Spinks K. The diagnosis and management of interstitial lung diseases. BMJ. 2015;350:h2072. doi: 10.1136/bmj.h2072. [DOI] [PubMed] [Google Scholar]
  • 8.Cottin V, Hirani NA, Hotchkin DL, et al. Presentation, diagnosis and clinical course of the spectrum of progressive-fibrosing interstitial lung diseases. Eur Respir Rev. 2018;27:180076. doi: 10.1183/16000617.0076-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mira-Avendano I, Abril A, Burger CD, et al. Interstitial lung disease and other pulmonary manifestations in connective tissue diseases. Mayo Clin Proc. 2019;94(2):309–325. doi: 10.1016/j.mayocp.2018.09.002. [DOI] [PubMed] [Google Scholar]
  • 10.Mathai SC, Danoff SK. Management of interstitial lung disease associated with connective tissue disease. BMJ. 2016;352:h6819. doi: 10.1136/bmj.h6819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Fischer A, Distler J. Progressive fibrosing interstitial lung disease associated with systemic autoimmune diseases. Clin Rheumatol. 2019;38(10):2673–2681. doi: 10.1007/s10067-019-04720-0. [DOI] [PubMed] [Google Scholar]
  • 12.Spagnolo P, Lee JS, Sverzellati N, Rossi G, Cottin V. The lung in rheumatoid arthritis: focus on interstitial lung disease. Arthritis Rheumatol. 2018;70(10):1544–1554. doi: 10.1002/art.40574. [DOI] [PubMed] [Google Scholar]
  • 13.Brito Y, Glassberg MK, Ascherman DP. Rheumatoid arthritis-associated interstitial lung disease: current concepts. Curr Rheumatol Rep. 2017;19:79. doi: 10.1007/s11926-017-0701-5. [DOI] [PubMed] [Google Scholar]
  • 14.Bendstrup E, Møller J, Kronborg-White S, Prior TS, Hyldgaard C. Interstitial lung disease in rheumatoid arthritis remains a challenge for clinicians. J Clin Med. 2019;8(12):2038. doi: 10.3390/jcm8122038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Borie R, Crestani B. Antineutrophil cytoplasmic antibody-associated lung fibrosis. Semin Respir Crit Care Med. 2018;39(4):465–470. doi: 10.1055/s-0038-1669914. [DOI] [PubMed] [Google Scholar]
  • 16.Alba MA, Flores-Suárez LF, Henderson AG, et al. Interstital lung disease in ANCA vasculitis. Autoimmun Rev. 2017;16(7):722–729. doi: 10.1016/j.autrev.2017.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Atienza-Mateo B, Remuzgo-Martínez S, Mora Cuesta VM, et al. The spectrum of interstitial lung disease associated with autoimmune diseases: data of a 3.6-year prospective study from a referral center of interstitial lung disease and lung transplantation. J Clin Med. 2020;9(6):1606. doi: 10.3390/jcm9061606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fischer A, Antoniou KM, Brown KK, et al. An official European Respiratory Society/American Thoracic Society research statement: interstitial pneumonia with autoimmune features. Eur Respir J. 2015;46(4):976–987. doi: 10.1183/13993003.00150-2015. [DOI] [PubMed] [Google Scholar]
  • 19.Fischer A. Interstitial pneumonia with autoimmune features. Clin Chest Med. 2019;40(3):609–616. doi: 10.1016/j.ccm.2019.05.007. [DOI] [PubMed] [Google Scholar]
  • 20.Sebastiani M, Cassone G, De Pasquale L, et al. Interstitial pneumonia with autoimmune features: a single center prospective follow-up study. Autoimmun Rev. 2020;19:102451. doi: 10.1016/j.autrev.2019.102451. [DOI] [PubMed] [Google Scholar]
  • 21.Travis WD, Costabel U, Hansell DM, et al. An official American Thoracic Society/European Respiratory Society statement: update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med. 2013;188(6):733–748. doi: 10.1164/rccm.201308-1483ST. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ito Y, Arita M, Kumagai S, et al. Radiological fibrosis score is strongly associated with worse survival in rheumatoid arthritis-related interstitial lung disease. Mod Rheumatol. 2019;29(1):98–104. doi: 10.1080/14397595.2018.1442170. [DOI] [PubMed] [Google Scholar]
  • 23.Liu H, Xie S, Liang T, et al. Prognostic factors of interstitial lung disease progression at sequential HRCT in anti-synthetase syndrome. Eur Radiol. 2019;29(10):5349–5357. doi: 10.1007/s00330-019-06152-5. [DOI] [PubMed] [Google Scholar]
  • 24.Wells AU, Denton CP. Interstitial lung disease in connective tissue disease – mechanisms and management. Nat Rev Rheumatol. 2014;10(12):728–739. doi: 10.1038/nrrheum.2014.149. [DOI] [PubMed] [Google Scholar]
  • 25.Sgalla G, Iovene B, Calvello M, Ori M, Varone F, Richeldi L. Idiopathic pulmonary fibrosis: pathogenesis and management. Respir Res. 2018;19:32. doi: 10.1186/s12931-018-0730-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chatzidionisyou A, Catrina AI. The lung in rheumatoid arthritis, cause or consequence? Curr Opin Rheumatol. 2016;28(1):76–82. doi: 10.1097/BOR.0000000000000238. [DOI] [PubMed] [Google Scholar]
  • 27.Yamada H. ANCA-Associated lung fibrosis. Semin Respir Crit Care Med. 2011;32(3):322–327. doi: 10.1055/s-0031-1279828. [DOI] [PubMed] [Google Scholar]
  • 28.Alba MA, Jennette JC, Falk RJ. Pathogenesis of ANCA-associated pulmonary vasculitis. Semin Respir Crit Care Med. 2018;39(4):413–424. doi: 10.1055/s-0038-1673386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Paulin F, Doyle TJ, Fletcher EA, Ascherman DP, Rosas IO. Rheumatoid arthritis-associated interstitial lung disease and idiopathic pulmonary fibrosis: shared mechanistic and phenotypic traits suggest overlapping disease mechanisms. Rev Invest Clin. 2015;67(5):280. [PMC free article] [PubMed] [Google Scholar]
  • 30.Morisset J, Lee JS. New trajectories in the treatment of interstitial lung disease: treat the disease or treat the underlying pattern? Curr Opin Pulm Med. 2019;25(5):442–449. doi: 10.1097/MCP.0000000000000600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Juge PA, Lee JS, Ebstein E, et al. MUC5B promoter variant and rheumatoid arthritis with interstitial lung disease. N Engl J Med. 2018;379(23):2209–2219. doi: 10.1056/NEJMoa1801562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Putman RK, Gudmundsson G, Araki T, et al. The MUC5B promoter polymorphism is associated with specific interstitial lung abnormality subtypes. Eur Respir J. 2017;50:1700537. doi: 10.1183/13993003.00537-2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Namba N, Kawasaki A, Sada KE, et al. Association of MUC5B promoter polymorphism with interstitial lung disease in myeloperoxidase-antineutrophil cytoplasmic antibody-associated vasculitis. Ann Rheum Dis. 2019;78(8):1144–1146. doi: 10.1136/annrheumdis-2018-214263. [DOI] [PubMed] [Google Scholar]
  • 34.Seibold MA, Wise AL, Speer MC, et al. A common MUC5B promoter polymorphism and pulmonary fibrosis. N Engl J Med. 2011;364(16):1503–1512. doi: 10.1056/NEJMoa1013660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Dressen A, Abbas AR, Cabanski C, et al. Analysis of protein-altering variants in telomerase genes and their association with MUC5B common variant status in patients with idiopathic pulmonary fibrosis: a candidate gene sequencing study. Lancet Respir Med. 2018;6(8):603–614. doi: 10.1016/S2213-2600(18)30135-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Manfredi A, Sebastiani M, Cerri S, et al. Acute exacerbation of interstitial lung diseases secondary to systemic rheumatic diseases: a prospective study and review of the literature. J Thorac Dis. 2019;11(4):1621–1628. doi: 10.21037/jtd.2019.03.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vacchi C, Sebastiani M, Cassone G, et al. Therapeutic options for the treatment of interstitial lung disease related to connective tissue diseases. a narrative review. J Clin Med. 2020;9(2):407. doi: 10.3390/jcm9020407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Cassone G, Manfredi A, Vacchi C, et al. Treatment of rheumatoid arthritis-associated interstitial lung disease: lights and shadows. J Clin Med. 2020;9(4):1082. doi: 10.3390/jcm9041082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.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. doi: 10.1016/S0012-3692(15)50949-5. [DOI] [PubMed] [Google Scholar]
  • 40.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. doi: 10.3899/jrheum.121043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Saketkoo LA, Espinoza LR. Rheumatoid arthritis interstitial lung disease: mycophenolate mofetil as an antifibrotic and disease-modifying antirheumatic drug. Arch Intern Med. 2008;168(15):1718–1719. doi: 10.1001/archinte.168.15.1718. [DOI] [PubMed] [Google Scholar]
  • 42.Tashkin DP, Roth MD, Clements PJ, et al. Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): a randomised controlled, double-blind, parallel group trial. Lancet Respir Med. 2016;4(9):708–719. doi: 10.1016/S2213-2600(16)30152-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bullingham RES, Nicholls AJ, Kamm BR. Clinical pharmacokinetics of mycophenolate mofetil. Clin Pharmacokinet. 1998;34(6):429–455. doi: 10.2165/00003088-199834060-00002. [DOI] [PubMed] [Google Scholar]
  • 44.FDA. CellCept® (Mycophenolate Mofetil Capsules) (Mycophenolate Mofetil Tablets) CellCept® Oral Suspension (Mycophenolate Mofetil for Oral Suspension) CellCept® Intravenous (Mycophenolate Mofetil Hydrochloride for Injection) [Accessed July 20, 2020]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/050722s021,050723s019,050758s019,050759s024lbl.pdf.
  • 45.Allison AC. Mechanisms of action of mycophenolate mofetil. Lupus. 2005;14(Suppl 1):2–8. doi: 10.1191/0961203305lu2109oa. [DOI] [PubMed] [Google Scholar]
  • 46.Neff RT, Hurst FP, Falta EM, et al. Progressive multifocal leukoencephalopathy and use of mycophenolate mofetil after kidney transplantation. Transplantation. 2008;86(10):1474–1478. doi: 10.1097/TP.0b013e31818b62c8. [DOI] [PubMed] [Google Scholar]
  • 47.Vernino S, Salomao DR, Habermann TM, O’Neill BP. Primary CNS lymphoma complicating treatment of myasthenia gravis with mycophenolate mofetil. Neurology. 2005;65(4):639–641. doi: 10.1212/01.wnl.0000173031.56429.04. [DOI] [PubMed] [Google Scholar]
  • 48.Budde K, Drr M, Liefeldt L, Neumayer HH, Glander P. Enteric-coated mycophenolate sodium. Expert Opin Drug Saf. 2010;9(6):981–994. doi: 10.1517/14740338.2010.513379. [DOI] [PubMed] [Google Scholar]
  • 49.Salvadori M, Bertoni E, Budde K. Superior efficacy of enteric-coated mycophenolate vs mycophenolate mofetil in de novo transplant recipients: pooled analysis. TPS. 2010;42:1325–1328. doi: 10.1016/j.transproceed.2010.03.044. [DOI] [PubMed] [Google Scholar]
  • 50.Jung HY, Lee S, Jeon Y, et al. Mycophenolic acid trough concentration and dose are associated with hematologic abnormalities but not rejection in kidney transplant recipients. J Korean Med Sci. 2020;35(24):e185. doi: 10.3346/JKMS.2020.35.E185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Karampitsakos T, Tzilas V, Tringidou R, et al. Lung cancer in patients with idiopathic pulmonary fibrosis. Pulm Pharmacol Ther. 2017;45:1–10. doi: 10.1016/j.pupt.2017.03.016. [DOI] [PubMed] [Google Scholar]
  • 52.Liu Y, Zhu M, Geng J, et al. Incidence and radiologic-pathological features of lung cancer in idiopathic pulmonary fibrosis. Clin Respir J. 2018;12(4):1700–1705. doi: 10.1111/crj.12732. [DOI] [PubMed] [Google Scholar]
  • 53.Kingdon EJ, McLean AG, Psimenou E, et al. The safety and efficacy of MMF in lupus nephritis: a pilot study. Lupus. 2001;10(9):606–611. doi: 10.1191/096120301682430186. [DOI] [PubMed] [Google Scholar]
  • 54.Zhang Y, Zheng Y. Pneumocystis jirovecii pneumonia in mycophenolate mofetil-treated patients with connective tissue disease: analysis of 17 cases. Rheumatol Int. 2014;34(12):1765–1771. doi: 10.1007/s00296-014-3073-4. [DOI] [PubMed] [Google Scholar]
  • 55.Azevedo LS, Castro MCR, Paula FJ, Ianhez LE, David-Neto E. Mycophenolate mofetil may protect against Pneumocystis carinii pneumonia in renal transplanted patients. Rev Inst Med Trop Sao Paulo. 2005;47(3):143–145. doi: 10.1590/S0036-46652005000300005. [DOI] [PubMed] [Google Scholar]
  • 56.Totet A, Westeel PF, Nevez G, Azevedo LS. Severe Pneumocystis pneumonia in a renal transplant recipient after long term mycophenolate mofetil treatment (multiple letters) Rev Inst Med Trop Sao Paulo. 2005;47(5):303–305. doi: 10.1590/s0036-46652005000500012. [DOI] [PubMed] [Google Scholar]
  • 57.Iriart X, Le BM, Kamar N, Berry A. Pneumocystis pneumonia in solid-organ transplant recipients. J fungi (Basel, Switzerland) 2015;1(3):293–331. doi: 10.3390/jof1030293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Oz HS, Hughes WT. Novel anti-pneumocystis carinii effects of the immunosuppressant mycophenolate mofetil in contrast to provocative effects of tacrolimu sirolimus, and dexamethasone. J Infect Dis. 1997;175(4):901–904. doi: 10.1086/513988. [DOI] [PubMed] [Google Scholar]
  • 59.Husain S, Singh N. The impact of novel immunosuppressive agents on infections in organ transplant recipients and the interactions of these agents with antimicrobials. Clin Infect Dis. 2002;35(1):53–61. doi: 10.1086/340867. [DOI] [PubMed] [Google Scholar]
  • 60.Kobashigawa J, Miller L, Renlund D, et al. A randomized active-controlled trial of mycophenolate mofetil in heart transplant recipients. Transplantation. 1998;66(4):507–515. doi: 10.1097/00007890-199808270-00016. [DOI] [PubMed] [Google Scholar]
  • 61.Sarmiento JM, Dockrell DH, Schwab TR, Munn SR, Paya CV. Mycophenolate mofetil increases cytomegalovirus invasive organ disease in renal transplant patients. Clin Transplant. 2000;14(2):136–138. doi: 10.1034/j.1399-0012.2000.140206.x. [DOI] [PubMed] [Google Scholar]
  • 62.Renlund DG, Gopinathan SK, Kfoury AG, Taylor DO. Mycophenolate mofetil (MMF) in heart transplantation: rejection prevention and treatment. [Accessed May 12, 2020];Clin Transplant. 1996 10(1 Pt 2):136–139. http://www.ncbi.nlm.nih.gov/pubmed/8680051. [PubMed] [Google Scholar]
  • 63.Palmer SM, Baz MA, Sanders L, et al. Results of a randomized, prospective, multicenter trial of mycophenolate mofetil versus azathioprine in the prevention of acute lung allograft rejection. Transplantation. 2001;71(12):1772–1776. doi: 10.1097/00007890-200106270-00012. [DOI] [PubMed] [Google Scholar]
  • 64.Sebastiani M, Manfredi A, Cassone G, Sandri G, Cerri S, Ferri C. Interstitial lung disease is associated to infections of lower respiratory tract in immunocompromised rheumatoid arthritis patients. Clin Exp Rheumatol. 2017;35:542. [PubMed] [Google Scholar]
  • 65.Margaritopoulos GA, Kokosi MA, Wells AU. Diagnosing complications and co-morbidities of fibrotic interstitial lung disease. Expert Rev Respir Med. 2019;13(7):645–658. doi: 10.1080/17476348.2019.1632196. [DOI] [PubMed] [Google Scholar]
  • 66.Oldham JM, Lee C, Valenzi E, et al. Azathioprine response in patients with fibrotic connective tissue disease-associated interstitial lung disease. 2016;121:P117–122. doi: 10.1016/j.rmed.2016.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zhang G, Xu T, Zhang H, et al. [Randomized control multi-center clinical study of mycophenolate mofetil and cyclophosphamide in the treatment of connective tissue disease related interstitial lung disease]. Zhonghua Yi Xue Za Zhi. 2015;95(45):3641–3645. [PubMed] [Google Scholar]
  • 68.Saketkoo LA, Espinoza LR. Experience of mycophenolate mofetil in 10 patients with autoimmune-related interstitial lung disease demonstrates promising effects. Am J Med Sci. 2009;337(5):329–335. doi: 10.1097/MAJ.0b013e31818d094b. [DOI] [PubMed] [Google Scholar]
  • 69.Kelly C, Young A, Ahmad Y, et al. The effect of steroids, azathioprine and mycophenolate on the risk of death in rheumatoid arthritis. Rheumatology. 2016;55:i99–i100. doi: 10.1093/rheumatology/kew144.019. [DOI] [Google Scholar]
  • 70.Volkmann ER, Tashkin DP, Li N, et al. Mycophenolate mofetil versus placebo for systemic sclerosis–related interstitial lung disease: an analysis of scleroderma lung studies I and II. Arthritis Rheumatol. 2017;69(7):1451–1460. doi: 10.1002/art.40114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Naidu GSRSNK, Sharma SK, Adarsh MB, et al. Effect of mycophenolate mofetil (MMF) on systemic sclerosis-related interstitial lung disease with mildly impaired lung function: a double-blind, placebo-controlled, randomized trial. Rheumatol Int. 2020;40(2):207–216. doi: 10.1007/s00296-019-04481-8. [DOI] [PubMed] [Google Scholar]
  • 72.Perelas A, Silver RM, Arrossi AV, Highland KB. Systemic sclerosis-associated interstitial lung disease. Lancet Respir Med. 2020;8(3):304–320. doi: 10.1016/S2213-2600(19)30480-1. [DOI] [PubMed] [Google Scholar]
  • 73.Stratton RJ, Wilson H, Black CM. Pilot study of anti-thymocyte globulin plus mycophenolate mofetil in recent-onset diffuse scleroderma. Rheumatology (Oxford) 2001;40(1):84–88. doi: 10.1093/rheumatology/40.1.84. [DOI] [PubMed] [Google Scholar]
  • 74.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. doi: 10.1007/s10067-010-1498-z. [DOI] [PubMed] [Google Scholar]
  • 75.Le EN, Wigley FM, Shah AA, Boin F, Hummers LK. Long-term experience of mycophenolate mofetil for treatment of diffuse cutaneous systemic sclerosis. Ann Rheum Dis. 2011;70:1104–1107. doi: 10.1136/ARD.2010.142000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Omair MA, Alahmadi A, Johnson SR. Safety and effectiveness of mycophenolate in systemic sclerosis. A systematic review. PLoS One. 2015;10(5):e0124205. doi: 10.1371/journal.pone.0124205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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 doi: 10.1155/2012/143637. 143637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Nihtyanova SI, Brough GM, Black CM, Denton CP. Mycophenolate mofetil in diffuse cutaneous systemic sclerosis – a retrospective analysis. Rheumatology. 2007;46(3):442–445. doi: 10.1093/rheumatology/kel244. [DOI] [PubMed] [Google Scholar]
  • 79.Adler S, Huscher D, Siegert E, et al. Systemic sclerosis associated interstitial lung disease – individualized immunosuppressive therapy and course of lung function: results of the EUSTAR group. Arthritis Res Ther. 2018;20(1):17. doi: 10.1186/s13075-018-1517-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yilmaz N, Can M, Kocakaya D, Karakurt S, Yavuz S. Two-year experience with mycophenolate mofetil in patients with scleroderma lung disease: a case series. Int J Rheum Dis. 2014;17(8):923–928. doi: 10.1111/1756-185X.12399. [DOI] [PubMed] [Google Scholar]
  • 81.Mendoza FA, Nagle SJ, Lee JB, Jimenez SA. A prospective observational study of mycophenolate mofetil treatment in progressive diffuse cutaneous systemic sclerosis of recent onset. J Rheumatol. 2012;39(6):1241–1247. doi: 10.3899/jrheum.111229. [DOI] [PubMed] [Google Scholar]
  • 82.Derk CT, Grace E, Shenin M, Naik M, Schulz S, Xiong W. A prospective open-label study of mycophenolate mofetil for the treatment of diffuse systemic sclerosis. Rheumatology. 2009;48(12):1595–1599. doi: 10.1093/rheumatology/kep295. [DOI] [PubMed] [Google Scholar]
  • 83.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. doi: 10.1007/s10067-011-1823-1. [DOI] [PubMed] [Google Scholar]
  • 84.Herrick AL, Lunt M, Whidby N, et al. Observational study of treatment outcome in early diffuse cutaneous systemic sclerosis. J Rheumatol. 2010;37(1):116–124. doi: 10.3899/jrheum.090668. [DOI] [PubMed] [Google Scholar]
  • 85.Liossis SNC, Bounas A, Andonopoulos AP. Mycophenolate mofetil as first-line treatment improves clinically evident early scleroderma lung disease. Rheumatology. 2006;45(8):1005–1008. doi: 10.1093/rheumatology/kei211. [DOI] [PubMed] [Google Scholar]
  • 86.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. [PubMed] [Google Scholar]
  • 87.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. doi: 10.1378/chest.06-2861. [DOI] [PubMed] [Google Scholar]
  • 88.Zamora AC, Wolters PJ, Collard HR, et al. Use of mycophenolate mofetil to treat scleroderma-associated interstitial lung disease. Respir Med. 2008;102(1):150–155. doi: 10.1016/j.rmed.2007.07.021. [DOI] [PubMed] [Google Scholar]
  • 89.Owen C, Ngian GS, Elford K, et al. Mycophenolate mofetil is an effective and safe option for the management of systemic sclerosis-associated interstitial lung disease: results from the Australian Scleroderma Cohort Study. Clin Exp Rheumatol. 2016;34(100):170–176. [PubMed] [Google Scholar]
  • 90.Ueda T, Sakagami T, Kikuchi T, Takada T. Mycophenolate mofetil as a therapeutic agent for interstitial lung diseases in systemic sclerosis. Respir Investig. 2018;56(1):14–20. doi: 10.1016/j.resinv.2017.11.004. [DOI] [PubMed] [Google Scholar]
  • 91.Baqir M, Makol A, Osborn TG, Bartholmai BJ, Ryu JH. Mycophenolate mofetil for scleroderma-related interstitial lung disease: a real world experience. PLoS One. 2017;12(5):e0177107. doi: 10.1371/journal.pone.0177107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Panopoulos ST, Bournia VK, Trakada G, Giavri I, Kostopoulos C, Sfikakis PP. Mycophenolate versus cyclophosphamide for progressive interstitial lung disease associated with systemic sclerosis: a 2-year case control study. Lung. 2013;191(5):483–489. doi: 10.1007/s00408-013-9499-8. [DOI] [PubMed] [Google Scholar]
  • 93.Erre GL, Sebastiani M, Fenu MA, et al. Efficacy, safety, and tolerability of treatments for systemic sclerosis-related interstitial lung disease: a systematic review and network meta-analysis. J Clin Med. 2020;9(8):2560. doi: 10.3390/jcm9082560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Scleroderma Lung Study III – Combining pirfenidone with mycophenolate – Full Text View – ClinicalTrials.gov. [Accessed May 11, 2020]. https://clinicaltrials.gov/ct2/show/NCT03221257.
  • 95.Luppi F, Sebastiani M, Silva M, et al. Interstitial lung disease in Sjögren’s syndrome: a clinical review. Clin Exp Rheumatol. 2020;38(4):291–300. [PubMed] [Google Scholar]
  • 96.Luppi F, Sebastiani M, Sverzellati N, Cavazza A, Salvarani C, Manfredi A. Lung complications of Sjogren syndrome. Eur Respir Rev. 2020;29:200021. doi: 10.1183/16000617.0021-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hervier B, Uzunhan Y. Inflammatory myopathy-related interstitial lung disease: from pathophysiology to treatment. Front Med. 2020;6:326. doi: 10.3389/fmed.2019.00326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Long K, Danoff SK. Interstitial lung disease in polymyositis and dermatomyositis. Clin Chest Med. 2019;40(3):561–572. doi: 10.1016/j.ccm.2019.05.004. [DOI] [PubMed] [Google Scholar]
  • 99.Mecoli CA, Christopher-Stine L. Management of interstitial lung disease in patients with myositis specific autoantibodies. Curr Rheumatol Rep. 2018;20:27. doi: 10.1007/s11926-018-0731-7. [DOI] [PubMed] [Google Scholar]
  • 100.Hamaguchi Y, Fujimoto M, Matsushita T, et al. Common and distinct clinical features in adult patients with anti-aminoacyl-tRNA synthetase antibodies: heterogeneity within the syndrome. PLoS One. 2013;8(4):e60442. doi: 10.1371/journal.pone.0060442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Cavagna L, Trallero-Araguás E, Meloni F, et al. Influence of antisynthetase antibodies specificities on antisynthetase syndrome clinical spectrum time course. J Clin Med. 2019;8(11):2013. doi: 10.3390/jcm8112013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Jablonski R, Bhorade S, Strek ME, Dematte J. Recognition and management of myositis-associated rapidly progressive interstitial lung disease. Chest. 2020;158(1):252–263. doi: 10.1016/j.chest.2020.01.033. [DOI] [PubMed] [Google Scholar]
  • 103.Mira-Avendano IC, Parambil JG, Yadav R, et al. A retrospective review of clinical features and treatment outcomes in steroid-resistant interstitial lung disease from polymyositis/dermatomyositis. Respir Med. 2013;107(6):890–896. doi: 10.1016/j.rmed.2013.02.015. [DOI] [PubMed] [Google Scholar]
  • 104.Hanaoka H, Iida H, Kiyokawa T, Takakuwa Y, Kawahata K. Mycophenolate mofetil treatment with or without a calcineurin inhibitor in resistant inflammatory myopathy. Clin Rheumatol. 2019;38(2):585–590. doi: 10.1007/s10067-018-4294-9. [DOI] [PubMed] [Google Scholar]
  • 105.Huapaya JA, Silhan L, Pinal-Fernandez I, et al. Long-Term treatment with azathioprine and mycophenolate mofetil for myositis-related interstitial lung disease. Chest. 2019;156(5):896–906. doi: 10.1016/j.chest.2019.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Morganroth PA, Kreider ME, Werth VP. Mycophenolate mofetil for interstitial lung disease in dermatomyositis. Arthritis Care Res. 2010;62(10):1496–1501. doi: 10.1002/acr.20212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Koyama RVL, Braga TKK, da Silva Dias GA, Fujihara S, Fuzii HT, Yoshikawa GT. Hypomyopathic dermatomyositis associated with interstitial lung disease and good response to mycophenolate mofetil: case-based review. Clin Rheumatol. 2017;36(8):1919–1926. doi: 10.1007/s10067-017-3671-0. [DOI] [PubMed] [Google Scholar]
  • 108.Gil B, Merav L, Pnina L, Chagai G. Diagnosis and treatment of clinically amyopathic dermatomyositis (CADM): a case series and literature review. Clin Rheumatol. 2016;35(8):2125–2130. doi: 10.1007/s10067-015-2928-8. [DOI] [PubMed] [Google Scholar]
  • 109.Tsuchiya H, Tsuno H, Inoue M, et al. Mycophenolate mofetil therapy for rapidly progressive interstitial lung disease in a patient with clinically amyopathic dermatomyositis. Mod Rheumatol. 2014;24(4):694–696. doi: 10.3109/14397595.2013.874762. [DOI] [PubMed] [Google Scholar]
  • 110.Cozzani E, Cinotti E, Felletti R, Pelucco D, Rebora A, Parodi A. Amyopathic dermatomyositis with lung involvement responsive to mycophenolate mofetil. Immunopharmacol Immunotoxicol. 2013;35(6):687–692. doi: 10.3109/08923973.2013.833624. [DOI] [PubMed] [Google Scholar]
  • 111.Kiely PDW, Chua F. Interstitial lung disease in inflammatory myopathies: clinical phenotypes and prognosis. Curr Rheumatol Rep. 2013;15(9):359. doi: 10.1007/s11926-013-0359-6. [DOI] [PubMed] [Google Scholar]
  • 112.Cavagna L, Castañeda S, Sciré C, Gonzalez-Gay MA. Antisynthetase syndrome or what else? Different perspectives indicate the need for new classification criteria. Ann Rheum Dis. 2018;77(8):e50. doi: 10.1136/annrheumdis-2017-212368. [DOI] [PubMed] [Google Scholar]
  • 113.Chatterjee S, Prayson R, Farver C. Antisynthetase syndrome: not just an inflammatory myopathy. Cleve Clin J Med. 2013;80(10):655–666. doi: 10.3949/ccjm.80a.12171. [DOI] [PubMed] [Google Scholar]
  • 114.Mimori T, Nakashima R, Hosono Y. Interstitial lung disease in myositis: clinical subsets, biomarkers, and treatment. Curr Rheumatol Rep. 2012;14(3):264–274. doi: 10.1007/s11926-012-0246-6. [DOI] [PubMed] [Google Scholar]
  • 115.Rüegg CA, Maurer B, Laube I, Scholtze D. Jo1-antisynthetase syndrome and severe interstitial lung disease with organising pneumonia on histopathology with favourable outcome on early combined treatment with corticosteroids, mycophenolate mofetil and rituximab. BMJ Case Rep. 2019;12(9):10–14. doi: 10.1136/bcr-2019-231006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sundaragiri PR, Vallabhajosyula S, Kanaan JP. Interstitial lung disease in antisynthetase syndrome without clinical myositis. BMJ Case Rep. 2014;2014 doi: 10.1136/bcr-2014-204296. bcr2014204296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kulkarni HS, Gutierrez FR, Despotovic V, Russell TD. A 43-year-old man with antisynthetase syndrome presenting with acute worsening of dyspnea. Chest. 2015;147(6):e215–e219. doi: 10.1378/chest.14-2402. [DOI] [PubMed] [Google Scholar]
  • 118.Hirakata M, Suwa A, Takada T, et al. PM-DM_antiKS_A&R2007. Arthritis Rheum. 2007;56(4):1295–1303. doi: 10.1002/art.22506. [DOI] [PubMed] [Google Scholar]
  • 119.Kunimasa K, Arita M, Nakazawa T, et al. The clinical characteristics of two anti-OJ (anti-isoleucyl-tRNA synthetase) autoantibody-positive interstitial lung disease patients with polymyositis/dermatomyositis. Intern Med. 2012;51(24):3405–3410. doi: 10.2169/internalmedicine.51.7452. [DOI] [PubMed] [Google Scholar]
  • 120.Koguchi-Yoshioka H, Okiyama N, Iwamoto K, et al. Intravenous immunoglobulin contributes to the control of antimelanoma differentiation-associated protein 5 antibody-associated dermatomyositis with palmar violaceous macules/papules. Br J Dermatol. 2017;177(5):1442–1446. doi: 10.1111/bjd.15499. [DOI] [PubMed] [Google Scholar]
  • 121.Moghadam-Kia S, Oddis CV, Sato S, Kuwana M, Aggarwal R. Antimelanoma differentiation-associated gene 5 antibody: expanding the clinical spectrum in North American patients with dermatomyositis. J Rheumatol. 2017;44(3):319–325. doi: 10.3899/jrheum.160682. [DOI] [PubMed] [Google Scholar]
  • 122.Allenbach Y, Leroux G, Suárez-Calvet X, et al. Dermatomyositis with or without anti-melanoma differentiation-associated gene 5 antibodies common interferon signature but distinct NOS2 expression. Am J Pathol. 2016;186(3):691–700. doi: 10.1016/j.ajpath.2015.11.010. [DOI] [PubMed] [Google Scholar]
  • 123.Chen Z, Cao M, Plana MN, et al. Utility of anti-melanoma differentiation-associated gene 5 antibody measurement in identifying patients with dermatomyositis and a high risk for developing rapidly progressive interstitial lung disease: a review of the literature and a meta-analysis. Arthritis Care Res (Hoboken) 2013;65(8):1316–1324. doi: 10.1002/acr.21985. [DOI] [PubMed] [Google Scholar]
  • 124.Sakamoto S, Okamoto M, Kaieda S, et al. Low positive titer of anti-melanoma differentiation-associated gene 5 antibody is not associated with a poor long-term outcome of interstitial lung disease in patients with dermatomyositis. Respir Investig. 2018;56(6):464–472. doi: 10.1016/j.resinv.2018.07.007. [DOI] [PubMed] [Google Scholar]
  • 125.Ye S, Chen XX, Lu XY, et al. Adult clinically amyopathic dermatomyositis with rapid progressive interstitial lung disease: a retrospective cohort study. Clin Rheumatol. 2007;26(10):1647–1654. doi: 10.1007/s10067-007-0562-9. [DOI] [PubMed] [Google Scholar]
  • 126.Hayashi M, Kikuchi T, Takada T. Mycophenolate mofetil for the patients with interstitial lung diseases in amyopathic dermatomyositis with anti-MDA-5 antibodies. Clin Rheumatol. 2017;36(1):239–240. doi: 10.1007/s10067-016-3443-2. [DOI] [PubMed] [Google Scholar]
  • 127.Hisanaga J, Kotani T, Fujiki Y, Yoshida S, Takeuchi T, Makino S. Successful multi-target therapy including rituximab and mycophenolate mofetil in anti-melanoma differentiation-associated gene 5 antibody-positive rapidly progressive interstitial lung disease with clinically amyopathic dermatomyositis. Int J Rheum Dis. 2017;20(12):2182–2185. doi: 10.1111/1756-185X.13136. [DOI] [PubMed] [Google Scholar]
  • 128.Girard C, Vincent T, Bessis D. Dermatomyosite et pneumopathie interstitielle rapidement évolutive associées aux auto-anticorps anti-MDA-5: une présentation clinique atypique. Ann Dermatol Venereol. 2013;140(10):628–634. doi: 10.1016/j.annder.2013.04.083. [DOI] [PubMed] [Google Scholar]
  • 129.Toyoda Y, Koyama K, Kawano H, et al. Clinical features of interstitial pneumonia associated with systemic lupus erythematosus. Respir Investig. 2019;57(5):435–443. doi: 10.1016/j.resinv.2019.04.005. [DOI] [PubMed] [Google Scholar]
  • 130.Enomoto N, Egashira R, Tabata K, et al. Analysis of systemic lupus erythematosus-related interstitial pneumonia: a retrospective multicentre study. Sci Rep. 2019;9:7355. doi: 10.1038/s41598-019-43782-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Muangchan C, Van Vollenhoven RF, Bernatsky SR, et al. Treatment algorithms in systemic lupus erythematosus. Arthritis Care Res. 2015;67(9):1237–1245. doi: 10.1002/acr.22589. [DOI] [PubMed] [Google Scholar]
  • 132.Rashidi A, Alajmi M, Hegazi MO. Mycophenolate mofetil as a maintenance therapy for lupus-related diffuse alveolar hemorrhage: a case report. Lupus. 2011;20(14):1551–1553. doi: 10.1177/0961203311411353. [DOI] [PubMed] [Google Scholar]
  • 133.Mosca M, Tani C, Vagnani S, Carli L, Bombardieri S. The diagnosis and classification of undifferentiated connective tissue diseases. J Autoimmun. 2014;48–49:50–52. doi: 10.1016/j.jaut.2014.01.019. [DOI] [PubMed] [Google Scholar]
  • 134.Vivero F, Campins F, Lancellotti D, et al. Autoimmune interstitial lung disease in Latin-America. Clin Immunol. 2019;199:52–56. doi: 10.1016/j.clim.2018.12.013. [DOI] [PubMed] [Google Scholar]
  • 135.McCoy SS, Mukadam Z, Meyer KC, et al. Mycophenolate therapy in interstitial pneumonia with autoimmune features: a cohort study. Ther Clin Risk Manag. 2018;14:2171–2181. doi: 10.2147/TCRM.S173154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Novikov P, Shchegoleva E, Akulkina L, Bulanov N, Vinogradova E, Moiseev S. Diagnostic pitfalls and treatment challenges in interstitial pneumonia with autoimmune features: comment on the article by Wilfong et al. Arthritis Rheumatol. 2019;71(4):651–652. doi: 10.1002/art.40783. [DOI] [PubMed] [Google Scholar]
  • 137.Wilfong EM, Lentz RJ, Guttentag A, et al. Interstitial pneumonia with autoimmune features: an emerging challenge at the intersection of rheumatology and pulmonology. Arthritis Rheumatol. 2018;70(12):1901–1913. doi: 10.1002/art.40679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Maher TM, Corte TJ, Fischer A, et al. Pirfenidone in patients with unclassifiable progressive fibrosing interstitial lung disease: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Respir Med. 2020;8(2):147–157. doi: 10.1016/S2213-2600(19)30341-8. [DOI] [PubMed] [Google Scholar]
  • 139.Torrisi SE, Kahn N, Wälscher J, et al. Possible value of antifibrotic drugs in patients with progressive fibrosing non-IPF interstitial lung diseases. BMC Pulm Med. 2019;19(1):213. doi: 10.1186/s12890-019-0937-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Huang H, Sun YX, Li S, Shao C, Xu K, Xu ZJ. [The clinical experience of pirfenidone based on corticosteroids and immunosuppressant treatment for interstitial pneumonia with autoimmune features]. Zhonghua Jie He He Hu Xi Za Zhi. 2019;42(9):700–704. doi: 10.3760/cma.j.issn.1001-0939.2019.09.009. [DOI] [PubMed] [Google Scholar]
  • 141.Sebastiani M, Manfredi A, Vacchi C, et al. Epidemiology and management of interstitial lung disease in ANCA-associated vasculitis. 2020;38(Suppl 124):221–231. [PubMed] [Google Scholar]
  • 142.Thompson GE, Specks U. Update on the management of respiratory manifestations of the antineutrophil cytoplasmic antibodies-associated vasculitides. Clin Chest Med. 2019;40(3):573–582. doi: 10.1016/j.ccm.2019.05.012. [DOI] [PubMed] [Google Scholar]
  • 143.Yates M, Watts RA, Bajema IM, et al. EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis. Ann Rheum Dis. 2016;75(9):1583–1594. doi: 10.1136/annrheumdis-2016-209133. [DOI] [PubMed] [Google Scholar]
  • 144.Jain R, Yadav D, Puranik N, Guleria R, Jin J-O. Sarcoidosis: causes, diagnosis, clinical features, and treatments. J Clin Med. 2020;9(4):1081. doi: 10.3390/jcm9041081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Papiris S, Stagaki E, Papadaki G, et al. Mycophenolate mofetil as an alternative treatment in sarcoidosis. Pulm Pharmacol Ther. 2019;58:101840. doi: 10.1016/j.pupt.2019.101840. [DOI] [PubMed] [Google Scholar]
  • 146.Hamzeh N, Voelker A, Forssén A, et al. Efficacy of mycophenolate mofetil in sarcoidosis. Respir Med. 2014;108(11):1663–1669. doi: 10.1016/j.rmed.2014.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Brill AK, Ott SR, Geiser T. Effect and safety of mycophenolate mofetil in chronic pulmonary sarcoidosis: a retrospective study. Respiration. 2013;86(5):376–383. doi: 10.1159/000345596. [DOI] [PubMed] [Google Scholar]
  • 148.Barnes H, Holland AE, Westall GP, Goh NSL, Glaspole IN. Cyclophosphamide for connective tissue disease-associated interstitial lung disease. Cochrane Database Syst Rev. 2018;1:CD010908. doi: 10.1002/14651858.CD010908.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Atienza-Mateo B, Remuzgo-Martínez S, Prieto-Peña D, et al. Rituximab in the treatment of interstitial lung disease associated with autoimmune diseases: experience from a single referral center and literature review. J Clin Med. 2020;9(10):3070. doi: 10.3390/jcm9103070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Daoussis D, Liossis S-NC, Tsamandas AC, et al. Experience with rituximab in scleroderma: results from a 1-year, proof-of-principle study. Rheumatol. 2010;49(2):217–280. doi: 10.1093/rheumatology/kep093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Daoussis D, Melissaropoulos K, Sakellaropoulos G, et al. A multicenter, open-label, comparative study of B-cell depletion therapy with Rituximab for systemic sclerosis-associated interstitial lung disease. Semin Arthritis Rheum. 2017;46(5):625–631. doi: 10.1016/j.semarthrit.2016.10.003. [DOI] [PubMed] [Google Scholar]
  • 152.Jordan S, Distler JHW, Maurer B, et al. Effects and safety of rituximab in systemic sclerosis: an analysis from the European Scleroderma Trial and Research (EUSTAR) group. Ann Rheum Dis. 2015;74(6):1188–1194. doi: 10.1136/annrheumdis-2013-204522. [DOI] [PubMed] [Google Scholar]
  • 153.Rituximab in Systemic Sclerosis (RECOVER) Clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/study/NCT01748084.
  • 154.Saunders P, Tsipouri V, Keir GJ, et al. Rituximab versus cyclophosphamide for the treatment of connective tissue diseaseassociated interstitial lung disease (RECITAL): study protocol for a randomised controlled trial. Trials. 2017;18:275. doi: 10.1186/s13063-017-2016-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Cassone G, Manfredi A, Atzeni F, et al. Safety of abatacept in Italian Patients with rheumatoid arthritis and interstitial lung disease: a multicenter retrospective study. J Clin Med. 2020;9(1):277. doi: 10.3390/jcm9010277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Manfredi A, Cassone G, Furini F, et al. Tocilizumab therapy in rheumatoid arthritis with interstitial lung disease: a multicenter retrospective study. Intern Med J. 2020;50(9):1085–1090. doi: 10.1111/imj.14670. [DOI] [PubMed] [Google Scholar]
  • 157.Fernández-Díaz C, Castañeda S, Melero-González RB, et al. Abatacept in interstitial lung disease associated with rheumatoid arthritis: national multicenter study of 263 patients. Rheumatology. 2020;59(12):3906–3916. doi: 10.1093/rheumatology/keaa621. [DOI] [PubMed] [Google Scholar]

Articles from Drugs in Context are provided here courtesy of BioExcel Publishing Ltd

RESOURCES