Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Nov 26.
Published in final edited form as: Arthritis Rheumatol. 2024 Jul 8;76(8):1182–1200. doi: 10.1002/art.42861

2023 American College of Rheumatology (ACR)/American College of Chest Physicians (CHEST) Guideline for the Treatment of Interstitial Lung Disease in People with Systemic Autoimmune Rheumatic Diseases

Sindhu R Johnson 1,†,*, Elana J Bernstein 2,*, Marcy B Bolster 3, Jonathan H Chung 4, Sonye K Danoff 5, Michael D George 6, Dinesh Khanna 7, Gordon Guyatt 8, Reza D Mirza 8, Rohit Aggarwal 9, Aberdeen Allen Jr 10, Shervin Assassi 11, Lenore Buckley 12, Hassan A Chami 5, Douglas S Corwin 13, Paul F Dellaripa 14, Robyn T Domsic 9, Tracy J Doyle 14, Catherine Marie Falardeau 15, Tracy M Frech 16, Fiona K Gibbons 3, Monique Hinchcliff 12, Cheilonda Johnson 6, Jeffrey P Kanne 17, John S Kim 18, Sian Yik Lim 19, Scott Matson 20, Zsuzsanna H McMahan 5, Samantha J Merck 21, Kiana Nesbitt 22, Mary Beth Scholand 23, Lee Shapiro 24, Christine D Sharkey 17, Ross Summer 25, John Varga 7, Anil Warrier 26, Sandeep K Agarwal 27, Danielle Antin-Ozerkis 12, Bradford Bemiss 28, Vaidehi Chowdhary 12, Jane E Dematte D’Amico 28, Robert Hallowell 3, Alicia M Hinze 29, Patil A Injean 30, Nikhil Jiwrajka 6, Elena K Joerns 31, Joyce S Lee 32, Ashima Makol 29, Gregory C McDermott 14, Jake G Natalini 33, Justin M Oldham 7, Didem Saygin 9, Kimberly Showalter Lakin 34, Namrata Singh 35, Joshua J Solomon 36, Jeffrey A Sparks 14, Marat Turgunbaev 37, Samera Vaseer 38, Amy Turner 37, Stacey Uhl 39, Ilya Ivlev 39
PMCID: PMC12646471  NIHMSID: NIHMS2059290  PMID: 38978310

Abstract

Objective.

We provide evidence-based recommendations regarding the treatment of interstitial lung disease (ILD) in adults with systemic autoimmune rheumatic diseases (SARDs).

Methods.

We developed clinically relevant population, intervention, comparator, and outcomes questions. A systematic literature review was then performed, and the available evidence was rated using the Grading of Recommendations, Assessment, Development, and Evaluation methodology. A panel of clinicians and patients reached consensus on the direction and strength of the recommendations.

Results.

Thirty-five recommendations were generated (including two strong recommendations) for first-line SARD-ILD treatment, treatment of SARD-ILD progression despite first-line ILD therapy, and treatment of rapidly progressive ILD. The strong recommendations were against using glucocorticoids in systemic sclerosis–ILD as a first-line ILD therapy and after ILD progression. Otherwise, glucocorticoids are conditionally recommended for first-line ILD treatment in all other SARDs.

Conclusion.

This clinical practice guideline presents the first recommendations endorsed by the American College of Rheumatology and American College of Chest Physicians for the treatment of ILD in people with SARDs.

INTRODUCTION

Interstitial lung disease (ILD) is a significant cause of morbidity and mortality in people with systemic autoimmune rheumatic diseases (SARDs). The American College of Rheumatology (ACR) has developed guidelines for screening those at risk for ILD and for the monitoring and treatment of those who have developed SARD-ILD, reported separately.1 This guideline provides recommendations for the treatment of ILD in adults with those SARDs at greatest ILD risk, including systemic sclerosis (SSc), rheumatoid arthritis (RA), idiopathic inflammatory myopathies (IIM including polymyositis, dermatomyositis, antisynthetase syndrome, immune-mediated necrotizing myopathy), mixed connective tissue disease (MCTD), and Sjögren disease (SjD).2,3

METHODS

We followed the ACR guideline development process and policy for managing conflicts of interest and disclosures (https://rheumatology.org/clinical-practice-guidelines), including Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) methodology.46 Supplementary Materials 1 describes our methods. The core leadership team (including rheumatologists, methodologists, a pulmonologist, and a radiologist) drafted clinical population, intervention, comparator, and outcomes (PICO) questions (Supplementary Materials 2). The Literature Review Team conducted a systematic literature review, rated the quality of evidence (high, moderate, low, or very low), and produced the evidence report (Supplementary Materials 3). A Patient Panel was convened that included 21 people with SARDs (SSc n = 7, RA n = 5, IIM n = 7, MCTD n = 3, SjD n = 8, with some patients having more than one diagnosis) at risk for (n = 4 [19%]) or diagnosed with ILD (n = 17 [81%]); the median age was 53 years (range 33–73 years), and the panel included n = 16 women (71%), with n = 14 White individuals (67%), n = 7 Black or multiracial individuals (33%), and n = 2 Hispanic individuals (10%). They met virtually with three members of the Core Team (MBB, MDG, and RDM) and two ACR staff, providing their values and preferences related to ILD screening, monitoring, and treatment.7 An expert Voting Panel including rheumatologists (n = 19), pulmonologists (n = 4), a radiologist (n = 1), and Patient Panel representatives (n = 3) reviewed the evidence report and formulated and voted on the recommendations at virtual meetings (February to March 2023).

Consensus required ≥70% agreement on both direction and strength (strong or conditional) to make a recommendation. Strong recommendations indicate confidence that benefits of an intervention clearly outweigh the harms (or vice versa); conditional recommendations denote uncertainty regarding net benefit. The Panel voted on paired comparisons of treatment options resulting in a hierarchy of preferred treatments. Because of limitations in evidence and factors that might affect choices for specific patients (eg, extrapulmonary manifestations, provider comfort with medications and monitoring requirements, costs, access), we do not emphasize a mandatory treatment sequence but instead provide a range of “preferred” or “additional” ILD treatment options. Supplementary Materials 4 provides rosters of the Core Leadership Team, Literature Review Team, Voting Panel, and Patient Panel.

Scope

Patients.

This guideline applies only to adults with SSc, RA, MCTD, IIMs, and SjD-associated ILD, but not individuals who are pregnant or nursing.

Interventions.

This guideline addresses methotrexate, leflunomide, azathioprine, cyclophosphamide, mycophenolate, calcineurin inhibitors (CNIs; tacrolimus, cyclosporine), tumor necrosis factor inhibitors (TNFi; etanercept, adalimumab, infliximab, golimumab, certolizumab pegol), tocilizumab, rituximab, abatacept, JAK inhibitors (JAKi; tofacitinib, baricitinib, upadacitinib), antifibrotic agents (pirfenidone; nintedanib), glucocorticoids (oral prednisone, intravenous methylprednisolone), intravenous immunoglobulin (IVIG), plasma exchange, and referrals for stem cell and lung transplantation.

Outcomes.

Critical outcomes included death, disability, quality of life (QoL), and adverse events, including serious adverse events, toxicity leading to discontinuation, and other adverse reactions. Surrogate outcomes were disease activity/disease progression defined by forced vital capacity (FVC), diffusion capacity for carbon monoxide, and change in extent of ILD or ILD progression on high-resolution computed tomography of the chest (HRCT chest).

Progression of ILD.

Progression (as an outcome) was defined using the INBUILD trial criteria,8 ie, decline in FVC of ≥10% the predicted value, decline in FVC of 5% to <10% the predicted value and worsening of respiratory symptoms or increased fibrosis on HRCT chest, or worsening of respiratory symptoms and increased fibrosis all within 24 months. These criteria may be useful in assessments for disease progression but are not requisite for routine follow-up and clinical decision-making by practitioners and patients.

Rapidly progressive ILD.

Rapidly progressive ILD (RP-ILD) represents a subpopulation of ILD characterized by rapid progression from breathing room air or a patient’s baseline oxygen requirement to a high oxygen requirement or intubation within days to weeks without a documented alternative cause (eg, infection, heart failure), which can be seen in a diagnosis of acute interstitial pneumonia.9,10 Progressive pulmonary fibrosis (PPF) and progressive fibrosing ILD (PF-ILD) differ from RP-ILD, and these terms are not interchangeable.11

Recommendations related to RP-ILD were considered for monotherapy, dual therapy, and triple therapy. Monotherapy agents considered were oral/intravenous glucocorticoids, mycophenolate, azathioprine, CNI, rituximab, and cyclophosphamide. Dual combination therapy was defined as simultaneous use of glucocorticoids and one of the above therapies. Triple combination therapy was defined as simultaneous use of glucocorticoids and two of the above therapies.12

RESULTS/RECOMMENDATIONS

Thirty-five treatment recommendations (Table 1, Figures 1, 2, and 3) are based on 216 PICO questions. The literature search identified 5,235 records (including screening, monitoring, and treatment). After excluding 4,038 titles and abstracts, 1,197 full-text articles were reviewed, 1,083 excluded, and 117 included: screening (n = 10), monitoring (n = 17), and treatment (n = 90) (Supplementary Material 5 and 6). Table 2 summarizes Voting Panel decisions, PICO questions, certainty of evidence, and evidence that led to treatment recommendations. Table 3 provides guidance on medication toxicity and monitoring. Table 4 summarizes integrative and pharmacologic interventions beyond the scope of this guideline.

Table 1.

Summary of recommendations for management of SARD-ILD*

Recommendations for management of SARD-ILD: first ILD treatment

 For people with SARD-ILD other than SSc-ILD, we conditionally recommend glucocorticoids as a first-line ILD treatment.
 For people with SSc-ILD, we strongly recommend against glucocorticoids as a first-line ILD treatment.
 For people with SARD-ILD, we conditionally recommend mycophenolate, azathioprine, rituximab, and cyclophosphamide as first-line ILD treatment options.
 For people with SSc-ILD and MCTD-ILD, we conditionally recommend tocilizumab as a first-line ILD treatment option.
 For people with SARD-ILD, we conditionally recommend against leflunomide, methotrexate, TNFi, and abatacept as first-line ILD treatment options.
 For people with SSc-ILD, we conditionally recommend nintedanib as a first-line ILD treatment option.
 For people with SjD-ILD, IIM-ILD, and MCTD-ILD, we conditionally recommend against nintedanib as a first-line ILD treatment option.
 For people with RA-ILD, the Panel was not able to come to consensus on whether to recommend nintedanib as a first-line ILD treatment option.
 For people with SARD-ILD, we conditionally recommend against pirfenidone as a first-line ILD treatment option.
 For people with SARD-ILD receiving mycophenolate without evidence of ILD progression, we conditionally recommend against adding nintedanib or pirfenidone to mycophenolate.
 For people with SARD-ILD, we conditionally recommend against upfront combination of nintedanib or pirfenidone with mycophenolate over mycophenolate alone as first-line ILD treatment options.
 For people with IIM-ILD, we conditionally recommend JAKi as a first-line ILD treatment option.
 For people with SARD-ILD other than IIM-ILD, we conditionally recommend against JAKi as a first-line ILD treatment option.
 For people with IIM-ILD, we conditionally recommend CNIs as a first-line ILD treatment option.
 For people with SARD-ILD other than IIM-ILD, we conditionally recommend against CNIs as a first-line ILD treatment option.
 For people with SARD-ILD, we conditionally recommend against IVIG or plasma exchange as first-line ILD treatment options.
 For people with SARD-associated ILD, we conditionally recommend optimal medical management over referral for stem cell or lung transplantation as first-line ILD treatment options.

Recommendations for management of SARD-ILD progression despite first-line ILD treatment

 For people with SSc-ILD progression despite first ILD treatment, we strongly recommend against using long-term glucocorticoids, and in other SARD-ILD, we conditionally recommend against using long-term glucocorticoids.
 For people with SARD-ILD progression despite first ILD treatment, we conditionally recommend mycophenolate, rituximab, cyclophosphamide, and nintedanib as treatment options.
 For people with RA-ILD progression despite first ILD treatment, we conditionally recommend adding pirfenidone as a treatment option.
 For people with SARD-ILD other than RA-ILD progression despite first ILD treatment, we conditionally recommend against adding pirfenidone as a treatment option.
 For people with SSc-ILD, MCTD-ILD, or RA-ILD progression despite first ILD treatment, we conditionally recommend using tocilizumab as a treatment option.
 For people with SjD-ILD and IIM-ILD progression despite first ILD treatment, we conditionally recommend against using tocilizumab as a treatment option.
 For people with IIM-ILD progression despite first ILD treatments, we conditionally recommend using a CNI as a treatment option.
 For people with SARD-ILD other than IIM-ILD progression despite first ILD treatments, we conditionally recommend against using a CNI as a treatment option.
 For people with IIM-ILD progression despite first ILD treatment, we conditionally recommend using JAKi as a treatment option.
 For people with IIM-ILD and MCTD-ILD progression despite first ILD treatment, we conditionally recommend adding IVIG as a treatment option.
 For people with SARD-ILD progression despite first ILD treatment, we conditionally recommend against using plasma exchange as a treatment option.
 For people with SSc-ILD progression despite first ILD treatment, we conditionally recommend referral for stem cell transplantation and/or lung transplantation.

Recommendations for management of SARD with RP-ILD

 For people with SARD and RP-ILD, we conditionally recommend pulse intravenous methylprednisolone as a first-line RP-ILD treatment.
 For people with SARD and RP-ILD, we conditionally recommend rituximab, cyclophosphamide, IVIG, mycophenolate, CNI, and JAKi as first-line RP-ILD treatment options.
 For people with SARD and RP-ILD, we conditionally recommend against methotrexate, leflunomide, azathioprine, TNFi, abatacept, tocilizumab, nintedanib, pirfenidone, and plasma exchange as first-line RP-ILD treatment options.
 For people with RP-ILD, we conditionally recommend upfront combination therapy (triple therapy for those with confirmed or suspected MDA-5 and double or triple therapy for those without confirmed or suspected MDA-5) over monotherapy as first-line treatment.
 For people with SARD and RP-ILD, we conditionally recommend against referral for stem cell transplantation over optimal medical management as a first-line RP-ILD treatment.
 For people with SARD and RP-ILD, we conditionally recommend early referral for lung transplantation over later referral after progression on optimal medical management.
*

CNI, calcineurin inhibitor; IIM, idiopathic inflammatory myopathies; ILD, interstitial lung disease; IVIG, intravenous immunoglobulin; JAKi, JAK inhibitor; MCTD, mixed connective tissue disease; MDA-5, melanoma differentiation-associated protein 5; RA, rheumatoid arthritis; RP, rapidly progressive; SARD, systemic autoimmune rheumatic disease; SjD, Sjögren disease; SSc, systemic sclerosis; TNFi, tumor necrosis factor inhibitor.

Figure 1.

Figure 1.

Initial treatment options for the treatment of ILD associated with systemic autoimmune rheumatic diseases of interest. * Decisions on GC dose and use of oral versus intravenous therapy depend on severity of disease. GCs should be used cautiously in patients with MCTD with an SSc phenotype who may be at increased risk of renal crisis. Short-term is defined as 3 months or less. Treatments are listed in order based on a hierarchy established by head-to-head votes, although the panel noted that decisions on which first-line therapy to use were dependent on specific situations and patient factors. In all diseases, mycophenolate was conditionally recommended over the other listed therapies. Therapies are divided into “preferred” and “additional” options based on the rank-order hierarchy. CNI, calcineurin inhibitor; GC, glucocorticoid; ILD, interstitial lung disease; JAKi, JAK inhibitor; MCTD, mixed connective tissue disease; SSc, systemic sclerosis.

Figure 2.

Figure 2.

Management of SARD-ILD with progression of ILD despite first ILD therapy. * If intolerance leads to suboptimal dosing of first-line therapy, consider switching to an alternative first-line therapy. Therapies are generally listed in order based on a hierarchy established by head-to-head votes, but decisions depend on specific clinical situations. Decision on whether to switch therapy or add to current therapy depends on current therapy and on which therapy is being initiated. Cyclophosphamide is not typically used in combination with other therapies, whereas others may be used individually or in combination. Decision on use of nintedanib vs immunosuppression depends on pace of progression and amount of fibrotic disease or presence of a usual interstitial pneumonia pattern on CT chest. § JAKi conditionally recommended as an option particularly in patients with anti–MDA-5. Short-term glucocorticoids may be of use in some patients with disease flares or as a bridge when switching therapy. AHSCT, autologous hematopoietic stem cell transplantation; CNI, calcineurin inhibitor; CT, computed tomography; GC, glucocorticoid; ILD, interstitial lung disease; IVIG, intravenous immunoglobulin; JAKi, JAK inhibitor; MCTD, mixed connective tissue disease; MDA-5, melanoma differentiation-associated protein 5; SARD, systemic autoimmune rheumatic disease. Color figure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42861/abstract.

Figure 3.

Figure 3.

Management of SARD with rapidly progressive ILD. * In rare patients with systemic sclerosis with rapidly progressive ILD, there was no consensus on whether or not to use glucocorticoids; if used, patients should be monitored closely for evidence of renal crisis. Rituximab and cyclophosphamide recommended over IVIG, but IVIG may be preferred if there is high concern for infection. ILD, interstitial lung disease; IV, intravenous; IVIG, intravenous immune globulin; JAKi, JAK inhibitor; MDA-5, melanoma differentiation-associated protein 5; SARD, systemic autoimmune rheumatic disease. Color figure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42861/abstract.

Table 2.

Summary of Voting Panel decisions, certainty of evidence, PICO questions, and evidence that led to treatment recommendations*

Statement Certainty of evidence Based on the evidence reports of the following PICO(s) Evidence table in Supplementary Materials 3, page(s)
For people with SARD-ILD other than SSc-ILD, we conditionally recommend glucocorticoids as a first-line ILD treatment. Very low 36, 59, 60, 74, 75, 88, 89, 101, 102, 237 200, 201, 318–322, 371–375, 418–422, 465–469, 652
For people with SSc-ILD, we strongly recommend against daily glucocorticoids as a first-line ILD treatment. Very low 36, 59, 60, 74, 75, 88, 89, 101, 102, 237 200, 202, 318–322, 371–375, 418–422, 652
For people with SARD-ILD, we conditionally recommend mycophenolate, azathioprine, rituximab, and cyclophosphamide as first-line ILD treatment options. Very low 25, 33, 48, 49, 50, 52, 53, 55, 56, 60, 61, 65, 66, 68, 70 116, 179–189, 255–304, 309–313, 321–325, 335–349, 353–354, 359–361
For people with SSc-ILD and MCTD-ILD, we conditionally recommend tocilizumab as a first-line ILD treatment option. Very low 50, 52, 68, 82, 95 292–294, 298–300, 353–355, 401–403, 440–444
For people with SARD-ILD, we conditionally recommend against leflunomide, methotrexate, TNFi, and abatacept as first-line ILD treatment options. Very low 27, 28, 31, 32, 46, 47, 51, 54, 62, 63, 67, 69, 77, 78, 81, 83, 91, 92, 94, 96 143–152, 170–179, 249–254, 295–298, 305–308, 325–330, 350–352, 356–358, 378–384, 398–400, 404–406, 425–432, 436–439, 444–447
For people with SjD-ILD, IIM-ILD, and MCTD-ILD, we conditionally recommend against nintedanib as a first-line ILD treatment option. Very low 38, 56, 71, 85, 98, 104 203–223, 309–313, 362–366, 409–413, 450–454, 477–480
For people with SSc-ILD, we conditionally recommend nintedanib as a first-line ILD treatment option. Very low 38, 56, 71, 85, 98, 104 203–223, 309–313, 362–366, 409–413, 450–454, 477–480
For people with RA-ILD, the Panel was not able to come to consensus on whether to recommend nintedanib as a first-line ILD treatment option. Very low 38, 56, 71, 85, 98, 104 203–223, 309–313, 362–366, 409–413, 450–454, 477–480
For people with SARD-ILD, we conditionally recommend against pirfenidone as a first-line ILD treatment option. Very low 39, 57, 72, 86, 99 224–229, 313–315, 366–368, 413–415, 455–461
For people with SARD-ILD receiving mycophenolate without evidence of ILD progression, we conditionally recommend against adding nintedanib or pirfenidone to mycophenolate. Very low 42, 43 234–241
For people with SARD-ILD, we conditionally recommend against upfront combination of nintedanib or pirfenidone with mycophenolate over mycophenolate alone as first-line ILD treatment options. Very low 44, 45 242–248
For people with IIM-ILD, we conditionally recommend JAKi as a first-line ILD treatment option. Very low 35, 55, 70, 84, 97 197–199, 359–362, 407, 408, 448, 449
For people with IIM-ILD, we conditionally recommend CNIs as a first-line ILD treatment option. Very low 30, 50, 66, 80, 93 162–169, 292–294, 346–349, 390–397, 433–435
For people with SARD-ILD, we conditionally recommend against IVIG or plasma exchange as first-line ILD treatment options. Very low 40, 41, 58, 61, 73, 76, 87, 90, 100, 103 230–233, 316, 317, 323–325, 369, 370, 376–378, 416–418, 423, 424, 462–464
For people with SARD-associated ILD, we conditionally recommend optimal medical management over referral for stem cell or lung transplantation as first-line ILD treatment. Very low 105, 106 481–489
For people with SARD-ILD with progression despite first ILD therapy, we conditionally recommend against adding glucocorticoids, and in people with SSc-ILD, we strongly recommend against adding glucocorticoids. Very low 122, 123 515, 516
For people with SARD-ILD progression after first ILD therapy, we conditionally recommend using mycophenolate, rituximab, cyclophosphamide. Very low 111, 112, 113, 115, 116, 118, 119, 121, 127, 128, 130, 131, 133, 134, 136, 139, 142 497–502, 504–508, 510–512, 514, 521–524, 526–528, 530–532, 534, 537, 540
For people with SARD-ILD progression after first ILD therapy, we conditionally recommend adding nintedanib. Very low 107, 148, 161 490–493, 547, 561
For people with RA-ILD progression after first ILD therapy, we conditionally recommend adding pirfenidone. Very low 108, 120, 135, 149, 162 494, 513, 533, 548, 562
For people with SSc-ILD, MCTD-ILD, RA-ILD progression after first ILD therapy, we conditionally recommend adding tocilizumab. Very low 145 544
For people with SARD-ILD other than IIM-ILD, with ILD progression after first ILD therapy, we conditionally recommend against adding CNIs. Very low 143 541, 542
For people with IIM-ILD progression after first ILD therapy, we conditionally recommend adding CNIs. Very low 143, 156 541, 542, 555
For people with SARD-ILD progression after first ILD therapy, we conditionally recommend adding JAKi over adding azathioprine. Very low 147 546
For people with SARD-ILD progression after first ILD therapy, we conditionally recommend adding cyclophosphamide over adding JAKi. Very low 160 559, 560
For people with IIM-ILD and MCTD-ILD progression after first ILD therapy, we conditionally recommend adding IVIG. Very low 136, 150, 163 534, 549, 563
For people with SARD-ILD progression after first ILD therapy, we conditionally recommend against using plasma exchange. Very low 139, 153, 166 537, 552, 555
For people with SSc-ILD progression after first ILD therapy, we conditionally recommend referral for stem cell transplantation and/or lung transplantation. Very low 167, 168 568, 569
For people with SARD other than SSc and RP-ILD, we conditionally recommend pulse intravenous glucocorticoids as first-line RP-ILD treatment. Very low 169, 170, 190, 191, 205, 206, 232, 233 570, 571, 595, 596, 616, 617, 643, 644
For people with SSc and RP-ILD, the Panel could not come to consensus on glucocorticoids as first-line RP-ILD treatment. Very low 169, 170, 190, 191, 205, 206, 232, 233 570, 571, 595, 596, 616, 617, 643, 644
For people with SARD and RP-ILD, we conditionally recommend rituximab, cyclophosphamide, IVIG, mycophenolate, CNIs, and JAKi as first-line RP-ILD treatments. Very low 180, 181, 184, 186, 189, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 207, 210, 211, 215, 224, 228, 231 583–586, 589, 591, 594, 598–618, 621–623, 626, 635, 639, 642
For people with SARD and RP-ILD, we conditionally recommend against nintedanib or pirfenidone as a first-line RP-ILD treatment. Very low 171, 173, 175, 177, 178, 179, 182, 183, 185, 187, 195, 199, 202, 210, 211, 226, 229 572, 574, 576, 578–582, 587, 588, 590, 592, 600–601, 609, 613, 621, 622, 637, 640
For people with SARD-ILD and RP-ILD, we conditionally recommend against plasma exchange as first-line RP-ILD treatment. Very low 192, 221, 234 597, 634, 645
For people with SARD-ILD other than SSc-ILD, and RP-ILD, we conditionally recommend dual combination therapy over monotherapy as first-line RP-ILD treatment. Very low 235 646–648
For people with SSc-ILD and RP-ILD, we conditionally recommend against dual combination therapya over monotherapy as first-line RP-ILD treatment. Very low 235 646–648
For people with confirmed or suspected MDA-5 RP-ILD, we conditionally recommend triple combination therapy over monotherapy as first-line RP-ILD treatment. Very low 236 649–651
For people without high suspicion for MDA-5 RP-ILD, we conditionally recommend adding IVIG to monotherapy or dual combination therapy as first-line RP-ILD treatment. Very low 238 653–657
For people without high suspicion for MDA-5 RP-ILD, we conditionally recommend against adding IVIG to triple therapy as first-line RP-ILD treatment. Very low 238 653–657
For people without high suspicion for MDA-5 RP-ILD, we conditionally recommend adding IVIG to monotherapy, dual or triple combination therapy, as subsequent RP-ILD treatment. Very low 238 653–657
For people without high suspicion for MDA-5 RP-ILD, we conditionally recommend adding plasma exchange to monotherapy or dual combination therapy as first-line RP-ILD treatment. Very low 238 653–657
For people without high suspicion for MDA-5 RP-ILD, we conditionally recommend adding plasma exchange to monotherapy or dual combination therapy as subsequent RP-ILD treatment. Very low 238 653–657
For people with SARD and RP-ILD, we conditionally recommend against referral for stem cell transplantation over optimal medical management as first-line RP-ILD treatment. Very low 240 661
For people with SARD and RP-ILD, we conditionally recommend early referral for lung transplantation over later referral after progression on optimal medical management. Very low 241 662
*

CNI, calcineurin inhibitor; IIM, idiopathic inflammatory myopathies; ILD, interstitial lung disease; IVIG, intravenous immunoglobulin; JAKi, JAK inhibitor; MCTD, mixed connective tissue disease; MDA-5, melanoma differentiation-associated protein 5; PICO, population, intervention, comparator, and outcomes; RA, rheumatoid arthritis; RP, rapidly progressive; SARD, systemic autoimmune rheumatic disease; SjD, Sjögren disease; SSc, systemic sclerosis; TNFi, tumor necrosis factor inhibitor.

a

Monotherapy may include oral prednisone/intravenous methylprednisolone, mycophenolate, azathioprine, CNIs, rituximab, or cyclophosphamide. Dual combination therapy refers to simultaneous use of glucocorticoids and one of the above therapies. Triple combination therapy refers to simultaneous use of glucocorticoids and two of the above therapies.

Table 3.

Medication toxicity and monitoring guidance*

Medication Notable toxicities Starting doses and frequency Monitoring
Azathioprine Hepatotoxicity, leukopenia, rarely pancreatitis Starting at 50 mg per day PO and gradually increasing to a therapeutic dose of 2–3 mg/kg/day CBC with differential at baseline and LFTs 2–3 weeks after starting and 2–3 weeks after any dose increase; every 3 months once on a stable dose
Cyclophosphamide Marrow suppression, infertility, hemorrhagic cystitis, need for pregnancy avoidance; long term—malignancy, including bladder cancer Intravenous: 500–750 mg/m2 IV every 4 weeks for 6 months CBC with differential and urinalysis at baseline; with intravenous dosing, CBC with differential 10–14 days after administration and just before next dose
Oral: starting at 50–150 mg per day with target dose of 2 mg/kg per day (up to a maximum of 200 mg per day) for 6 months; some experts use a maximum dose of 100 mg daily With oral dosing, CBC with differential 10–14 days after starting and 10–14 days after any dose increase; every 4 weeks on stable dosing
Urinalysis every 4–8 weeks on stable dosing (IV or oral); once treated with cyclophosphamide, annual urine cytology
Cyclosporine Infection, hypertension, nephrotoxicity, hyperkalemia, hepatotoxicity, neurotoxicity, gingival hyperplasia, thrombotic microangiopathy, malignancy, hirsutism 3 mg/kg/day13 PO adjusted for the target trough levels between 100 and 150 ng/mL14 Baseline BP, serum creatinine, BUN, CBC, serum magnesium, potassium, uric acid, lipid profile; monitor BP, CBC, serum creatinine, and levels of BUN, uric acid, potassium, lipids, and magnesium every other week during the first 3 months of treatment, monthly monitoring after the first 3 months
Glucocorticoids: methylprednisolone and prednisone Hyperglycemia, hypertension, mood disturbances, osteoporosis, avascular necrosis Pulse: methylprednisolone 1 g IV daily for 3 days Blood pressure and serum glucose, DEXA scan if ≥3 months of glucocorticoids anticipated15
Prednisone 1 mg/kg/day PO
IVIG Aseptic meningitis, increased risk of VTE, renal insufficiency, hemolytic anemia 2 g/kg IV divided over 2–5 days every 4 weeks IgA level before initiation; renal function before first infusion and at regular intervals; monitoring for anaphylaxis during infusion, especially in patients with IgA-deficiency, and thromboembolic events
JAKi (tofacitinib, baricitinib, upadacitinib) Leukopenia, herpes zoster, black box warning regarding MACE, malignancy Tofacitinib: immediate release tablet, 5 mg PO twice daily; extended-release tablet, 11 mg PO daily Hepatitis B virus infection, hepatitis C virus infection, and latent TB screening before initiation; CBC with differential and CMP at baseline, 4–8 weeks after starting, and every 3 months thereafter; lipids at baseline, 4–8 weeks after starting, then annually
Baricitinib: 2 mg PO daily Upadacitinib: 15 mg PO daily
Mycophenolate Marrow suppression, hepatotoxicity; black box warning regarding pregnancy Mycophenolate mofetil: starting at 500 mg PO twice daily and gradually increasing to a therapeutic dose of 1,000–1,500 mg twice daily CBC with differential and CMP at baseline, 2–3 weeks after starting and 2–3 weeks after any dose increase, and every 3 months once on a stable dose; full body skin examination, preferably by a dermatologist, annually
Mycophenolic acid: starting at 360 mg PO twice daily and gradually increasing to a therapeutic dose of 720–1,080 mg twice daily
Nintedanib Hepatotoxicity, diarrhea, increased risk of CV events, potential increased risk of bleeding 100–150 mg PO every 12 hours LFTs every month for 3 months, then every 3 months; monitor for diarrhea and weight loss
Pirfenidone  Rash, diarrhea, nausea, abdominal pain, photosensitivity, weight loss, upper respiratory tract infection, dizziness Days 1 to 7: 267 mg 3 times per day; days 8 to 14: 534 mg 3 times per day; day 15 and after: 801 mg 3 times per day LFTs at baseline, every month for the first 6 months, then every 3 months thereafter; monitor for photosensitivity and weight loss
Rituximab Cytopenias, infection, hepatitis B reactivation; black box warning for PML 1 g IV every 2 weeks for 2 doses; may be repeated every 24 weeks as needed Hepatitis B virus infection, hepatitis C virus infection, and latent TB screening before initiation; CBC with differential at baseline and at 2- to 4-month intervals; monitor for infusion reactions
Tacrolimus Leukopenia, renal failure, neurotoxicity Dosing: 0.075 mg/kg/day adjusted for the target whole-blood trough levels between 5 and 10 ng/mL16; some experts use a maximum trough level of 6 ng/mL Trough tacrolimus level, CMP, magnesium, phosphorus monitored 1–2 times per week for the first month, monthly for 3 months, then every 2–3 months; monitor BP
Tocilizumab Transaminitis, hyperlipidemia, bowel perforation 162 mg subcutaneously weekly Latent TB screening before initiation; CBC with differential at baseline, 4–8 weeks after starting therapy, and every 3 months thereafter; ALT/AST, alkaline phosphatase, and total bilirubin at baseline, every 4–8 weeks after starting therapy for the first 6 months, and every 3 months thereafter; lipids at baseline, 4–8 weeks after starting therapy, 6 months after starting therapy, then annually
*

ALT, alanine transaminase; AST, aspartate transaminase; BP, blood pressure; BUN, blood urea nitrogen; CBC, complete blood cell count; CMP, comprehensive metabolic panel; CV, acrdiovascular; DEXA, dual-energy X-ray absorptiometry; IV, intravenous; IVIG, intravenous immunoglobulin; JAKi, JAK inhibitor; LFT, liver function test; MACE, major adverse cadiovascular events; PML, progressive multifocal leukoencephalopathy; PO, per os; TB, tuberculosis; VTE, venous thromboembolism.

Table 4.

Additional integrative and pharmacologic interventions that may be considered in the care of people with SARD-ILD*

Interventions Examples
Integrative
 Exercise Aerobic, resistance training, yoga, tai chi
 Palliative care Symptom treatment (eg, cough, pain, air hunger), end of life planning17,18
 Physiotherapy Chest physiotherapy, airway clearance, incentive spirometry
 Pulmonary rehabilitation Cardiopulmonary rehabilitation, resistance training19
 Smoking cessation Smoking cessation program
 Supplemental oxygen Oxygen administration by nasal prongs
Pharmacologic
 Gastroesophageal reflux management Proton pump inhibitors, H2 blockers20,21
 PJP prophylaxis Trimethoprim sulfamethoxazole
 Promotility agents Domperidone
 Vaccines MMR, influenza, COVID-19, pneumococcus, varicella zoster, RSV22,23
*

ILD, interstitial lung disease; MMR, measle, mumps, rubella; PJP, Pneumocystis jirovecii pneumonia; RSV, respiratory syncytial virus; SARD, systemic autoimmune rheumatic disease.

Recommendations for management of SARD-ILD: first-line ILD treatment

Certainty of all evidence was low to very low (Table 2) (Supplementary Material 3). Although PICOs included head-to-head comparisons of treatments to create a hierarchy, limited evidence on the differences among individual therapies led to grouping first-line ILD treatment options into “preferred” therapies (expected to be most prescribed) and “additional options” (may be appropriate in certain situations) (Figure 1).

For people with SARD-ILD other than SSc-ILD, we conditionally recommend glucocorticoids as a first-line ILD treatment.

Because glucocorticoids are generally used in combination with other immunosuppressive agents and there is value in short-term glucocorticoid exposure, they are presented separately from other immunosuppressives. Oral prednisone is commonly used in SARD-ILD other than SSc-ILD, and intravenous pulse methylprednisolone is typically reserved for acute onset or severe ILD.24,25 Glucocorticoids might not be used in people with predominantly fibrotic disease (eg, RA with a usual interstitial pneumonia [UIP] pattern), but not all panelists agreed. Glucocorticoids should be used with caution in people with MCTD-ILD with SSc features or IIM-ILD with SSc antibodies given the risk for scleroderma renal crisis (SRC).2629

For people with SSc-ILD, we strongly recommend against glucocorticoids as first-line ILD treatment.

Glucocorticoids have been associated with SRC, particularly in doses of prednisone (or its equivalent) >15 mg daily, although concerns remain at any dose.2629 If glucocorticoids are considered, the lowest effective dose should be used, ideally <15 mg/day. The moderate certainty of evidence for harm2629 without clear evidence for efficacy led to a strong recommendation.

For people with SARD-ILD, we conditionally recommend mycophenolate, rituximab, cyclophosphamide, and azathioprineas first-line ILD treatment options.

Mycophenolate.

Data on the use of mycophenolate in SSc-ILD suggest similar outcomes as cyclophosphamide but a more favorable adverse effect profile.3039 This and limited data in other diseases, combined with substantial clinical experience, led the Voting Panel to conditionally recommend mycophenolate as a preferred first-line ILD treatment option across SARDs.30,32,33,4044 However, alternative first-line ILD treatment options may be selected based on extrapulmonary manifestations or other factors, including patient preference.

Rituximab.

Four trials and observational studies in people with SSc, RA, and IIM suggest that rituximab results in improvement or stabilization in FVC.33,40,42,4554 Rituximab may be preferred in the setting of inflammatory arthritis, myositis, or Sjögren neuropathy because of potential effectiveness for treating these manifestations. The Voting Panel expressed caution related to risk of infection, cost, and impact on immunogenicity of vaccines.

Cyclophosphamide.

Five trials36,52,5456 in SSc-ILD and observational studies12,27,5765 in IIM-ILD, SSc-ILD, and RA-ILD suggest that cyclophosphamide results in FVC improvement or stabilization but with increased adverse events (eg, infection, cytopenias, hemorrhagic cystitis, infertility) compared with other agents. The Patient Panel noted a willingness to accept these risks in either life-threatening situations or for significant benefit. Although efficacy data resulted in recommending cyclophosphamide as a first-line ILD treatment option, the adverse effect profile led to cyclophosphamide being considered an “additional option” rather than preferred first-line treatment.

Azathioprine.

The Voting Panel supported use of azathioprine as a first-line option across diseases, although in SSc-ILD, azathioprine was considered an “additional option” rather than “preferred” therapy because of limited evidence of effectiveness relative to other therapies in SSc-ILD (Figure 1).30,33,42,66,68

For people with SSc-ILD and MCTD-ILD, we conditionally recommend tocilizumab as a first-line ILD treatment option.

Tocilizumab resulted in FVC stabilization in two SSc trials compared with placebo.69,70 Tocilizumab has a favorable adverse event profile relative to cyclophosphamide.36,56,5973 Thus, it is a preferred first-line treatment in SSc-ILD, specifically in those with an inflammatory phenotype (elevated C-reactive protein, progressive skin thickening) and early, diffuse disease. Tocilizumab was recommended as an additional option for first-line therapy in MCTD-ILD, particularly for patients with SSc features. The Voting Panel was unable to reach consensus on using tocilizumab in RA-ILD,67 SjD-ILD, or IIM-ILD.

For people with SARD-ILD, we conditionally recommend against leflunomide, methotrexate, TNFi, and abatacept as first-line ILD treatment options.

Leflunomide.

Leflunomide has been associated with development or worsening of ILD in rare cases. There is uncertain benefit on ILD outcomes.74,81,97

Methotrexate.

Although methotrexate can rarely be associated with idiosyncratic pneumonitis, observational data suggest that methotrexate is not associated with progression of existing ILD.75,79,80 Evidence on benefit for ILD, however, is lacking. Methotrexate may be continued in patients with ILD who are receiving it for extrapulmonary manifestations, but methotrexate should be stopped if there is concern for methotrexate pneumonitis, and some panelists would stop if ILD developed while on methotrexate.

Abatacept.

Studies suggest no worsening of ILD with abatacept,76,77,82 and discontinuation because of ILD is not necessary, but efficacy of abatacept for ILD is uncertain.77,78,8287

TNFi.

There are no data demonstrating a beneficial effect of TNFi on SARD-ILD, but observational data of low quality suggest harm or no effect.81,8890 TNFi can be used for extrapulmonary manifestations; however, some panelists would stop TNFi if ILD developed while on TNFi.

For people with SSc-ILD, we conditionally recommend nintedanib as a first-line ILD treatment option.

Trial evidence of low certainty supports nintedanib use in SSc-ILD.91 However, due to the attenuation of FVC decline rather than stabilization or improvement in published studies, lack of efficacy for non-ILD SARD manifestations, adverse effects (especially diarrhea), and cost, immunosuppressive medications were favored over nintedanib, with nintedanib recommended as an “additional” rather than a “preferred” option for first-line treatment.

For people with SjD-ILD, IIM-ILD, and MCTD-ILD, we conditionally recommend against nintedanib as a first-line ILD treatment option.

Evidence for efficacy of nintedanib in SjD-ILD, IIM-ILD, and MCTD-ILD is limited, and there are concerns regarding cost, adverse effects, and a preference for immunosuppressive therapies as first-line treatment. Some panelists would, however, consider using nintedanib for patients with a UIP pattern of ILD irrespective of SARD.

For people with RA-ILD, the Panel could not come to consensus on whether to recommend nintedanib as a first-line ILD treatment option.

Participants with RA-ILD in the INBUILD study were selected for progressive ILD and may not reflect most patients with RA-ILD in clinical practice.8 Enthusiasm for nintedanib was limited given cost and potential adverse effects. Some Panelists consider nintedanib as a first-line ILD therapy option in patients with RA-ILD with a fibrotic/UIP pattern.

For people with SARD-ILD, we conditionally recommend against pirfenidone as a first-line ILD treatment option.

Limited evidence, adverse effects, and cost led to this recommendation.9296

For people with SARD-ILD receiving mycophenolate without evidence of ILD progression, we conditionally recommend against adding nintedanib or pirfenidone to mycophenolate.

In considering the role of sequential combination therapy (ie, nintedanib or pirfenidone after starting mycophenolate, even in absence of ILD progression), cost, adverse effect profile, and limited data on efficacy in patients without evidence of progression on mycophenolate led to this recommendation.

For people with SARD-ILD, we conditionally recommend against upfront combination of nintedanib or pirfenidone with mycophenolate over mycophenolate alone as first-line ILD treatment options.

As with sequential therapy, there are concerns about cost and adverse effects from simultaneous use of both an antifibrotic and mycophenolate in those without progression on mycophenolate. The Patient Panel agreed, noting a willingness to tolerate adverse effects for crucial treatments but that gastrointestinal adverse effects could significantly impact QoL. Similar principles apply to upfront combinations of antifibrotics with other immunosuppressive therapies.

For people with IIM-ILD, we conditionally recommend JAKi as a first-line ILD treatment option.

For people with SARD-ILD other than IIM-ILD, we conditionally recommend against JAKi as a first-line ILD treatment option.

Indirect evidence from one observational study of anti-MDA-5 melanoma differentiation-associated protein 5 (MDA-5) associated ILD (not RP-ILD) suggested lower death rates with tofacitinib compared to tacrolimus.108 With emerging evidence and increasing experience using JAKi in MDA-5-ILD, JAKi were recommended as an additional option, although not a preferred first-line therapy. Given limited data and experience, we recommend against JAKi as first-line therapy for ILD associated with other SARDs.86

For people with IIM-ILD, we conditionally recommend CNIs as a first-line ILD treatment option.

For people with SARD-ILD other than IIM-ILD, we conditionally recommend against CNIs as a first-line ILD treatment option.

Observational studies suggest benefits of CNIs for IIM-ILD.12,16,103108 These data, rapid onset of action, and lower cost led to a recommendation for CNIs as a first-line ILD treatment option for IIM-ILD. CNIs may be used particularly in MD-A-5-ILD or more severe forms of ILD at presentation. CNIs have potential renal toxicity, and providers may have less experience with dosing and monitoring (Table 3). The Panel preferred use of tacrolimus over cyclosporine because of perceived improved effectiveness. Given limited data, experience, and dosing and toxicity concerns, we recommend against CNIs as first-line therapy for other SARD-ILDs.

For people with SARD-ILD, we conditionally recommend against IVIG or plasma exchange as first-line ILD treatment options.

IVIG is effective for myositis and dysphagia and may be used in the acute setting if infection is a concern. However, because of limited ILD efficacy data and experience, we recommend against IVIG98100 and plasma exchange.101,102

For people with SARD-associated ILD, we conditionally recommend optimal medical management over referral for stem cell or lung transplantation as first-line ILD treatment options.

Indirect evidence from three stem cell transplantation (SCT) trials in SSc provided only low-certainty evidence because of significant loss of follow-up, small sample size, limited number of events, and different SCT protocols. SCT should be prioritized for centers with strong SSc-ILD experience and demonstrated positive outcomes to minimize differences in adverse effects and death risk. Lung transplantation should be considered after progression on therapy. Early referral for lung transplantation evaluation should be reserved for patients if presenting with advanced disease.

Recommendations for management of SARD-ILD progression despite first-line ILD treatment

For each recommendation below, first ILD therapy refers to any prior therapy and excludes the mentioned potential intervention. The term “using” implies “adding or switching” unless otherwise specified. A hierarchy of treatment options (Figure 2) has been established, but the order of therapy may vary because of patient factors and preferences. RP-ILD will be considered separately.

For people with SSc-ILD progression despite first ILD treatment, we strongly recommend against using long-term glucocorticoids, and in other SARD-ILD, we conditionally recommend against using long-term glucocorticoids.

Addition of long-term glucocorticoids (eg, >3 to 6 months) should not be relied upon for treating progressive ILD. Glucocorticoids may have a short-term role in patients with flares or as a bridge for those warranting a more definitive therapy change. The recommendation against glucocorticoids in SSc-ILD is strong because of moderate-certainty evidence of risk of SRC and very low-certainty evidence of benefit.

For people with SARD-ILD progression despite first ILD treatment, we conditionally recommend mycophenolate, rituximab, cyclophosphamide, and nintedanib as treatment options.

Mycophenolate.

If there is progression on first-line therapy, switching to mycophenolate is recommended over other treatment options, based on two trials and observational studies in SSc-ILD and RA-ILD of low certainty3039 and concerns about cyclophosphamide toxicity.

Rituximab.

Indirect evidence from four trials and observational studies33,40,42,4554 suggests FVC stabilization or improvement in people with SSc-ILD, MCTD-ILD, IIM-ILD, RA-ILD, and SjD-ILD. Some panelists add rituximab to mycophenolate for SSc-ILD and IIM-ILD, while others switch to rituximab, depending on patients’ risk factors and preferences.

Cyclophosphamide.

Indirect evidence from five trials36,52,5456 in SSc-ILD and observational studies12,27,5765 in IIM-ILD, SSc-ILD, and RA-ILD suggests FVC stabilization or improvement with cyclophosphamide, making it a second-line option despite its side effect profile.

Nintedanib.

Nintedanib may be added, particularly for those with progressive fibrosing disease on HRCT chest. Concerns were raised that nintedanib only slows FVC decline, at best, and has frequent gastrointestinal side-effects and cost.

For people with RA-ILD progression despite first ILD treatment, we conditionally recommend adding pirfenidone.

For people with SARD-ILD other than RA-ILD progression despite first ILD treatment, we conditionally recommend against adding pirfenidone.

Trials of pirfenidone in RA-ILD and SSc-ILD are underpowered but suggest that pirfenidone may attenuate FVC progression.9296 Pirfenidone can be considered in RA-ILD, particularly with a UIP pattern.

For people with SSc-ILD, MCTD-ILD, and RA-ILD progression despite first ILD treatment, we conditionally recommend tocilizumab as a treatment option.

For people with SjD-ILD and IIM-ILD progression despite first ILD treatment, we conditionally recommend against tocilizumab as a treatment option.

Although there is randomized controlled trial evidence for tocilizumab in SSc-ILD, it was studied primarily in early SSc-ILD.69,70 Adding or switching to tocilizumab may be an option in early progressive SSc-ILD, diffuse skin involvement, and elevated acute phase reactants and in MCTD-ILD with an SSc phenotype. Tocilizumab was conditionally recommended in RA-ILD despite limited evidence, given its established use for articular disease and favorable safety profile compared to cyclophosphamide.

For people with IIM-ILD progression despite first ILD treatments, we conditionally recommend CNIs as a treatment option.

For people with SARD-ILD other than IIM-ILD progression despite first ILD treatments, we conditionally recommend against CNIs as a treatment option.

CNIs have been beneficial in refractory IIM-ILD, particularly those with anti-synthetase syndrome or MDA-5-ILD.12,103,104 Although the Panel preferred mycophenolate and rituximab over CNIs, CNIs are a treatment option for IIM-ILD with progression after first ILD treatment. Given limited data and experience, CNIs are not recommended in other SARD-ILDs.

For people with IIM-ILD progression despite first ILD treatment, we conditionally recommend JAKi as a treatment option.

Emerging evidence suggests that JAKi are a potential treatment for IIM-ILD.108 Given limited data, however, other treatments, including cyclophosphamide, were recommended over JAKi.

For people with IIM-ILD and MCTD-ILD progression despite first ILD treatment, we conditionally recommend adding IVIG as a treatment option.

Observational studies report IVIG use in IIM-ILD.98100 Panelists reported using IVIG in IIM-ILD and myositis predominant–MCTD-ILD, particularly. IVIG may be useful when rapid onset of action is desired, eg, presence of severe respiratory muscle weakness.

For people with SARD-ILD progression despite first ILD treatment, we conditionally recommend against using plasma exchange.

Data supporting use of plasma exchange are limited to small observational studies, and plasma exchange has potential risks.

For people with SSc-ILD progression despite first ILD treatment, we conditionally recommend referral for SCT and/or lung transplantation.

Although SCT has been evaluated in three trials of low evidence certainty, the associated harms are understudied. The Panel was concerned about toxicity and the small number of centers that are well-equipped for its safe performance. Patient Panelists would consider transplantation in carefully selected cases if there was potential benefit. Although referral for SCT may be considered in those with ILD progression despite one or more ILD medications, the Panel did not reach consensus on optimal referral timing. Because there are a limited number of centers offering lung transplantation, referral should occur before a patient deteriorates and is no longer eligible.

Recommendations for management of SARD with RP-ILD

For people with SARD and RP-ILD, we conditionally recommend pulse intravenous methylprednisolone as first-line RP-ILD treatment.

Intravenous pulse methylprednisolone is recommended as first-line RP-ILD treatment because of rapid onset of action. Usual practice is to use intravenous glucocorticoids followed by high-dose oral prednisone. Glucocorticoids are typically administered with other immunosuppressive agents. RP-ILD in SSc is rare and may have an overlap syndrome or MCTD. Generally, we strongly recommend against use of glucocorticoids in SSc, but in the rare occurrence of RP-ILD in a person with SSc, there was no Panel consensus on whether to recommend glucocorticoids. Therapy may be warranted given the life-threatening nature of RP-ILD, despite the potential risk for SRC with glucocorticoids. We suggest an individualized approach in rare instances of RP-ILD in SSc.

For people with SARD and RP-ILD, we conditionally recommend rituximab, cyclophosphamide, IVIG, mycophenolate, CNIs, and JAKi as first-line RP-ILD treatment options.

Rituximab.

The RECITAL trial, comparing rituximab and cyclophosphamide in severe and progressive IIM-ILD, SSc-ILD, and MCTD-ILD, demonstrated similar mortality rates and FVC but higher rates of gastrointestinal and nervous system disorders in the cyclophosphamide group with similar discontinuation rates in the two treatment groups.52 The peak efficacy of rituximab is after several months of treatment, with onset of effect beginning sooner. There was concern about infection risk with rituximab because immunosuppressive effects last six months compared with one month with cyclophosphamide. The Panel conditionally recommended rituximab over mycophenolate, azathioprine, CNIs, and JAKi for RP-ILD.

Cyclophosphamide.

The Panel preferred intravenous over oral administration and voted conditionally for cyclophosphamide over mycophenolate and azathioprine. There was no consensus on cyclophosphamide versus rituximab except for MDA-5 RP-ILD, for which rituximab was preferred over cyclophosphamide. Patient Panelists were willing to accept significant adverse effects in the case of life-threatening illness if risks were clearly communicated but acknowledged that specific patient concerns (eg, fertility) may vary substantially.

IVIG.

Panelists reported using IVIG with pulse methylprednisolone in RP-ILD, noting a lower infection risk, particularly important in patients who are critically ill or intubated.100 IVIG is in limited supply and costly; it should not be used long term without clear clinical need. The Panel voted conditionally for rituximab and cyclophosphamide over IVIG but noted that IVIG may be used initially when infection risk is of particular concern.

Mycophenolate.

Based on very low-certainty randomized trial evidence,3041 experience, and tolerability of mycophenolate in SARD-ILD, the Panel recommends its use in RP-ILD.

CNIs.

Tacrolimus is increasingly used in IIM RP-ILD based on observational studies suggesting improved survival in IIM-ILD (non–RP-ILD).12,16,103108 The Panel voted conditionally for rituximab, cyclophosphamide, and mycophenolate over CNIs but voted conditionally for CNIs over azathioprine. Some panelists would consider tacrolimus over mycophenolate in MDA-5 RP-ILD.

JAKi.

Observational data suggest possible effectiveness of JAKi in those with MDA-5-ILD but without RP-ILD, and small studies report a possible benefit of adding JAKi to other therapy in refractory cases.86,108110 Given the limited data and experience, the Panel conditionally voted for cyclophosphamide, rituximab, and mycophenolate over JAKi.

For people with SARD and RP-ILD, we conditionally recommend against methotrexate, leflunomide, azathioprine, TNFi, abatacept, tocilizumab, nintedanib, pirfenidone, and plasma exchange as first-line RP-ILD treatment options.

There was neither evidence nor experience to support the use of these therapies in RP-ILD. Azathioprine is not typically used for RP-ILD. Low-quality observational data suggest that combination therapy with plasma exchange may result in a reduction in antibodies, improved oxygen exchange, and survival.101,102 Concerns were raised about cost, and availability is limited to specialized centers. Plasma exchange should not be used as first-line RP-ILD therapy but reserved as salvage therapy. If used, it is important to avoid rituximab or IVIG removal by plasma exchange.

For people with RP-ILD, we conditionally recommend upfront combination therapy (triple therapy for those with confirmed or suspected MDA-5 and double or triple therapy for those without confirmed or suspected MDA-5) over monotherapy as first-line treatment.

RP-ILD carries a substantial risk of death, and several observational studies have suggested benefit from treatment with upfront combination therapy.12,100102,108,111113 A study of MDA-5-ILD evaluating combination cyclophosphamide, tacrolimus, and glucocorticoids demonstrated a beneficial effect on survival compared to a sequential step-up approach.12 One study found adding IVIG to standard immunosuppressive medications in MDA-5 RP-ILD resulted in lower all-cause death rate compared with standard therapy.100 There is insufficient evidence to recommend one specific treatment regimen, and treatment selections may depend on disease severity, whether the person has suspected MDA-5 RP-ILD, and infection. Typically, initial combination therapy involves glucocorticoids with one or two additional agents recommended for RP-ILD, especially rituximab, cyclophosphamide, IVIG, tacrolimus, mycophenolate, or JAKi (Figure 3). Adding other agents may be considered for those who are not responding to therapy. There are limited data on adding plasma exchange to combination therapy in refractory cases.12,101,102,112115

For people with SARD and RP-ILD, we conditionally recommend against referral for SCT over optimal medical management as first-line RP-ILD treatment.

There was indirect evidence from three trials. The Voting Panel had concerns about the ability of people with RP-ILD to tolerate SCT.

For people with SARD and RP-ILD, we conditionally recommend early referral for lung transplantation over later referral after progression on optimal medical management.

There is a limited opportunity for lung transplantation, and the pre-transplantation evaluation takes time. Options for medical therapy may be swayed by a patient’s candidacy for lung transplantation. The need for high-flow oxygen is a marker of severity that warrants transfer to a transplantation center. Early referral to a transplantation center is warranted even if not local for the patient. Patient Panelists preferred early referral for lung transplantation and were agreeable to traveling away from home for potentially life-saving measures.

DISCUSSION

We present recommendations for first-line ILD treatment, treatment for ILD progression despite first-line treatment, and treatment of RP-ILD. The Panel voted on paired comparisons of treatment options resulting in a hierarchy of “preferred” and “additional” options for ILD treatment based on Voting and Patient Panel deliberations. Although mycophenolate was generally favored as a first-line ILD treatment for all diseases, patient- and disease-specific factors may lead to selection of a different treatment within the “menu of options” provided. For example, in people with RA-ILD with active inflammatory arthritis, rituximab (also a preferred option) may be chosen, particularly considering potential benefits for death and possibly ILD by reducing disease activity. We provide this hierarchy to be transparent about how experts consider these options, particularly in the setting of low- to very low-certainty evidence. This hierarchy should not be used by insurers to mandate a specific order of prescribing. Clinicians must retain the latitude to prescribe recommended medications based on individual patient factors and preferences. This guideline emphasizes co-management of people with SARD-ILD by rheumatologists and pulmonologists.

Glucocorticoids should be considered separately from other treatments, and our two strong recommendations are against their use in SSc-ILD because of their association with SRC.2629 For the other diseases, short-term glucocorticoids may be considered; however, there are patients in whom glucocorticoids may not be necessary as a first-line therapy. In individuals with ILD progression, long-term glucocorticoid use should be avoided and reserved for short-term use, such as bridging to another therapy. The Panel advised against the use of methotrexate, leflunomide, TNFi, and abatacept for SARD-ILD treatment, although these treatments may be appropriate for extrapulmonary manifestations. Some panelists would stop these medications if ILD developed while using them.

Because most recommendations are conditional, shared decision-making that accounts for factors such as ILD severity, risk factors for progression, other disease manifestations, cost, and toxicity is crucial when choosing a medication within the range of recommended options. Co-management with pulmonologists is advised for initiation of ILD treatment, particularly to determine the need for treatment in asymptomatic patients with stable and mild ILD. Patient Panelists were willing to tolerate medication toxicity if there is potential for substantial benefit, particularly to prolong life.7 However, they requested clear communication about potential toxicities and close collaboration with their providers when starting a medication to work through any adverse effects that develop.7

RP-ILD is a descriptive term of the modern era, initially coined to describe patients who go from being well to respiratory failure (needing high-flow oxygen or mechanical ventilation) within days to weeks. RP-ILD has been used primarily in the context of MDA-5-IIM. Historically, it was termed acute interstitial pneumonitis, but because of overlapping terminology, the meaning has been less clear. The Panel conceptualized RP-ILD in its original context. Note that PPF and PF-ILD differ from RP-ILD, and these terms are not interchangeable.11

Research is needed to better understand fibrotic, inflammatory, or mixed lung phenotypes, given that HRCT patterns of ILD (eg, nonspecific interstitial pneumonia, UIP) are often used to guide therapeutic decision-making. Investigation of tissue-level effects of therapeutics or development of predictive biomarkers might lead to more informed management decisions. Higher-certainty evidence is needed on the impact of therapeutics on patient-important outcomes, including harms. Research is needed among individuals who may be disproportionately affected by the diseases or face barriers to accessing care (eg, those with low income, women, and Black and Hispanic individuals). Although our recommendations are based on the best available evidence, in some instances, practice is changing in advance of data. For example, several expert centers use tacrolimus and IVIG in critically ill patients with RP-ILD, although published data are limited.

A limitation of this guideline was our inability to include other important affected patient groups, interventions, and outcomes. The scope of the guideline needed to be focused on what we considered to be the most critical of these. Additional integrative and pharmacologic interventions are summarized in Table 4 and could be considered patient by patient. The inclusion of pharmacists and stem cell or lung transplantation specialists would have further informed the Voting Panel. Although trial data were available, the certainty of evidence was frequently downgraded for small sample size, use of surrogate outcomes (eg, FVC), and evaluations in different groups than the diseases of interest. In addition to evidence, the Voting Panel deliberations considered clinical experience, patient values and preferences, disease burden, and access considerations, including cost.

In summary, we present guidelines for first-line treatment of SARD-ILD, treatment for ILD progression despite first ILD therapy, and treatment of RP-ILD in people with SARDs. This ACR guideline provides recommendations for ILD treatment decisions frequently faced in clinical practice.

Supplementary Material

Supp1
Supp2
Supp4
Supp5
Supp6
Supp3

Additional supplementary information cited in this article can be found online in the Supporting Information section (http://onlinelibrary.wiley.com/doi/10.1002/art.42861).

ACKNOWLEDGMENTS

We thank the patients who (along with authors Aberdeen Allen Jr, Catherine Marie Falardeau, and Kiana Nesbitt) participated in the Patient Panel meeting: Alicia C. Beach, Linda J. B. Baum, Mariann Boyanowski, Sarah Dingivan, Shelley Fritz, Amy Gietzen, Joi Goodbread, Donald Legere, Katie S. Lewis, Ashleigh McGregor, Benita Moyers, Renee Roberson, Richard Frank Seiden, Barbara Shafranski, Roberto Damian Zapata, and three patients who wished to remain anonymous. We thank the ACR staff, including Regina Parker for assistance in coordinating the administrative aspects of the project and Cindy Force for assistance with manuscript preparation. We thank Janet Waters for her assistance in developing the literature search strategy and performing the initial literature search and Kathryn Vela for performing the update searches. We thank the following people for their assistance with recruiting patients to participate on the Patient Panel: Lisa Christopher-Stine, MD, MPH, Johns Hopkins University School of Medicine; Maria I. Danila, MD, MSc, MSPH, University of Alabama at Birmingham; Jamie Hillner, Pulmonary Fibrosis Foundation; Mary Elizabeth Kreider, MD, MSCE, and Jamie Lederer, MSN, CRNP, University of Pennsylvania; Vivek Nagaraja, MBBS, MD, Mayo Clinic; Ganesh Raghu, MD, University of Washington; David Roofeh, MD, Michigan Medicine University of Michigan; Robert W. Simms, MD, Boston University Medical Center; Shilpa Venkatachalam, Global Healthy Living Foundation; Mary J. Wheatley, IOM, CAE, National Scleroderma Foundation; Lynn Wilson, Myositis Support & Understanding.

Footnotes

Author disclosures are available at https://onlinelibrary.wiley.com/doi/10.1002/art.42861.

REFERENCES

  • 1.Johnson SR, Berstein EJ, Bolster MB, et al. 2023 American College of Rheumatology (ACR)/American College of Chest Physicians (CHEST) guideline for the screening and monitoring of interstitial lung disease in people with systemic autoimmune rheumatic diseases. Arthritis Rheumatol 2024;76(8):1201–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Fischer A, du Bois R. Interstitial lung disease in connective tissue disorders. Lancet 2012;380:689–698. [DOI] [PubMed] [Google Scholar]
  • 3.Fischer A, Strek ME, Cottin V, et al. Proceedings of the American College of Rheumatology/Association of Physicians of Great Britain and Ireland Connective Tissue Disease-Associated Interstitial Lung Disease Summit: a multidisciplinary approach to address challenges and opportunities. Arthritis Rheumatol 2019;71:182–195. [DOI] [PubMed] [Google Scholar]
  • 4.Andrews JC, Schünemann HJ, Oxman AD, et al. GRADE guidelines: 15. Going from evidence to recommendation-determinants of a recommendation’s direction and strength. J Clin Epidemiol 2013;66: 726–735. [DOI] [PubMed] [Google Scholar]
  • 5.Guyatt GH, Oxman AD, Vist GE, et al. ; GRADE Working Group. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008;336:924–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Johnson SR, Turner AS, Goodman SM. How the American College of Rheumatology develops guidelines. Rheum Dis Clin North Am 2022;48:579–588. [DOI] [PubMed] [Google Scholar]
  • 7.Mirza RD, Bolster MB, Johnson SR, et al. Assessing patient values and preferences to inform the American College of Rheumatology/American College of Chest Physicians interstitial lung disease guidelines. Arthritis Care Res (Hoboken) 2024;76(8):1083–1089. doi: 10.1002/acr.25346 [DOI] [PubMed] [Google Scholar]
  • 8.Flaherty KR, Wells AU, Cottin V, et al. Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med 2019;381: 1718–1727. [DOI] [PubMed] [Google Scholar]
  • 9.Fujisawa T, Hozumi H, Kono M, et al. Prognostic factors for myositis-associated interstitial lung disease. PLoS One 2014;9:e98824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Jablonski R, Bhorade S, Strek ME, et al. Recognition and management of myositis-associated rapidly progressive interstitial lung disease. Chest 2020;158:252–263. [DOI] [PubMed] [Google Scholar]
  • 11.Rajan SK, Cottin V, Dhar R, et al. Progressive pulmonary fibrosis: an expert group consensus statement. Eur Respir J 2023;61:2103187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tsuji H, Nakashima R, Hosono Y, et al. Multicenter prospective study of the efficacy and safety of combined immunosuppressive therapy with high-dose glucocorticoid, tacrolimus, and cyclophosphamide in interstitial lung diseases accompanied by anti-melanoma differentiation-associated gene 5-positive dermatomyositis. Arthritis Rheumatol 2020;72:488–498. [DOI] [PubMed] [Google Scholar]
  • 13.Cavagna L, Caporali R, Abdì-Alì L, et al. Cyclosporine in anti-Jo1-positive patients with corticosteroid-refractory interstitial lung disease. J Rheumatol 2013;40:484–492. [DOI] [PubMed] [Google Scholar]
  • 14.Homma S, Sakamoto S, Kawabata M, et al. Cyclosporin treatment in steroid-resistant and acutely exacerbated interstitial pneumonia. Intern Med 2005;44:1144–1150. [DOI] [PubMed] [Google Scholar]
  • 15.Buckley L, Guyatt G, Fink HA, et al. 2017 American College of Rheumatology guideline for the prevention and treatment of glucocorticoid-induced osteoporosis. Arthritis Rheumatol 2017;69: 1521–1537. [DOI] [PubMed] [Google Scholar]
  • 16.Takada K, Katada Y, Ito S, et al. Impact of adding tacrolimus to initial treatment of interstitial pneumonitis in polymyositis/dermatomyositis: a single-arm clinical trial. Rheumatology (Oxford) 2020;59:1084–1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bajwah S, Higginson IJ, Ross JR, et al. The palliative care needs for fibrotic interstitial lung disease: a qualitative study of patients, informal caregivers and health professionals. Palliat Med 2013;27: 869–876. [DOI] [PubMed] [Google Scholar]
  • 18.Bajwah S, Ross JR, Peacock JL, et al. Interventions to improve symptoms and quality of life of patients with fibrotic interstitial lung disease: a systematic review of the literature. Thorax 2013;68: 867–879. [DOI] [PubMed] [Google Scholar]
  • 19.Griffiths TL, Burr ML, Campbell IA, et al. Results at 1 year of outpatient multidisciplinary pulmonary rehabilitation: a randomised controlled trial. Lancet 2000;355:362–368. [DOI] [PubMed] [Google Scholar]
  • 20.Hershcovici T, Jha LK, Johnson T, et al. Systematic review: the relationship between interstitial lung diseases and gastro-oesophageal reflux disease. Aliment Pharmacol Ther 2011;34:1295–1305. [DOI] [PubMed] [Google Scholar]
  • 21.Kilduff CE, Counter MJ, Thomas GA, et al. Effect of acid suppression therapy on gastroesophageal reflux and cough in idiopathic pulmonary fibrosis: an intervention study. Cough 2014;10:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bass AR, Chakravarty E, Akl EA, et al. 2022 American College of Rheumatology guideline for vaccinations in patients with rheumatic and musculoskeletal diseases. Arthritis Care Res (Hoboken) 2023; 75:449–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Curtis JR, Johnson SR, Anthony DD, et al. American College of Rheumatology guidance for COVID-19 vaccination in patients with rheumatic and musculoskeletal diseases: version 5. Arthritis Rheumatol 2023;75:E1–E16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pérez Campos D, Estévez Del Toro M, Peña Casanovas A, et al. Are high doses of prednisone necessary for treatment of interstitial lung disease in systemic sclerosis? Reumatol Clin 2012;8:58–62. [DOI] [PubMed] [Google Scholar]
  • 25.Hozumi H, Fujisawa T, Nakashima R, et al. Efficacy of glucocorticoids and calcineurin inhibitors for anti-aminoacyl-tRNA synthetase antibody-positive polymyositis/dermatomyositis-associated interstitial lung disease: a propensity score-matched analysis. J Rheumatol 2019;46:509–517. [DOI] [PubMed] [Google Scholar]
  • 26.DeMarco PJ, Weisman MH, Seibold JR, et al. Predictors and outcomes of scleroderma renal crisis: the high-dose versus low-dose D-penicillamine in early diffuse systemic sclerosis trial. Arthritis Rheum 2002;46:2983–2989. [DOI] [PubMed] [Google Scholar]
  • 27.Penn H, Howie AJ, Kingdon EJ, et al. Scleroderma renal crisis: patient characteristics and long-term outcomes. QJM 2007;100: 485–494. [DOI] [PubMed] [Google Scholar]
  • 28.Steen VD, Medsger TA Jr. Case-control study of corticosteroids and other drugs that either precipitate or protect from the development of scleroderma renal crisis. Arthritis Rheum 1998;41:1613–1619. [DOI] [PubMed] [Google Scholar]
  • 29.Teixeira L, Mouthon L, Mahr A, et al. ; Group Français de Recherche sur le Sclérodermie (GFRS). Mortality and risk factors of scleroderma renal crisis: a French retrospective study of 50 patients. Ann Rheum Dis 2008;67:110–116. [DOI] [PubMed] [Google Scholar]
  • 30.Adler S, Huscher D, Allanore Y, et al. Use of immunosuppressants in SSc patients with interstitial lung disease -results of the deSScipher project of the EUSTAR group. Clin Exp Rheumatol 2014;32: S85–S86.24528649 [Google Scholar]
  • 31.Kim GHJ, Tashkin DP, Lo P, et al. Using transitional changes on high-resolution computed tomography to monitor the impact of cyclophosphamide or mycophenolate mofetil on systemic sclerosis-related interstitial lung disease. Arthritis Rheumatol 2020; 72:316–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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;40:207–216. [DOI] [PubMed] [Google Scholar]
  • 33.Matson SM, Baqir M, Moua T, et al. Treatment outcomes for rheumatoid arthritis-associated interstitial lung disease: a real-world, multisite study of the impact of immunosuppression on pulmonary function trajectory. Chest 2023;163:861–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Panopoulos ST, Bournia VK, Trakada G, et al. Mycophenolate versus cyclophosphamide for progressive interstitial lung disease associated with systemic sclerosis: a 2-year case control study. Lung 2013;191:483–489. [DOI] [PubMed] [Google Scholar]
  • 35.Shenoy PD, Bavaliya M, Sashidharan S, et al. Cyclophosphamide versus mycophenolate mofetil in scleroderma interstitial lung disease (SSc-ILD) as induction therapy: a single-centre, retrospective analysis. Arthritis Res Ther 2016;18:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tashkin DP, Roth MD, Clements PJ, et al. ; Scleroderma Lung Study II Investigators. 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;4:708–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Tashkin DP, Volkmann ER, Tseng CH, et al. Improved cough and cough-specific quality of life in patients treated for scleroderma-related interstitial lung disease: results of Scleroderma Lung Study II. Chest 2017;151:813–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Volkmann ER, Tashkin DP, LeClair H, et al. Treatment with mycophenolate and cyclophosphamide leads to clinically meaningful improvements in patient-reported outcomes in scleroderma lung disease: results of Scleroderma Lung Study II. ACR Open Rheumatol 2020;2:362–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Volkmann ER, Tashkin DP, Sim M, et al. ; SLS I and SLS II study groups. Short-term progression of interstitial lung disease in systemic sclerosis predicts long-term survival in two independent clinical trial cohorts. Ann Rheum Dis 2019;78:122–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Amlani B, Elsayed G, Barvalia U, et al. Treatment of primary Sjögren’s syndrome-related interstitial lung disease: a retrospective cohort study. Sarcoidosis Vasc Diffuse Lung Dis 2020;37:136–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.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;156:896–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kelly CA, Nisar M, Arthanari S, et al. Rheumatoid arthritis related interstitial lung disease - improving outcomes over 25 years: a large multicentre UK study. Rheumatology (Oxford) 2021;60:1882–1890. [DOI] [PubMed] [Google Scholar]
  • 43.Oldham JM, Lee C, Valenzi E, et al. Azathioprine response in patients with fibrotic connective tissue disease-associated interstitial lung disease. Respir Med 2016;121:117–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.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;69:1451–1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.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:S17–S22. [PubMed] [Google Scholar]
  • 46.Ebata S, Yoshizaki A, Oba K, et al. Safety and efficacy of rituximab in systemic sclerosis (DESIRES): a double-blind, investigator-initiated, randomised, placebo-controlled trial. Lancet Rheumatol 2021;3: e489–e497. [DOI] [PubMed] [Google Scholar]
  • 47.Daoussis D, Liossis SN, Tsamandas AC, et al. Experience with rituximab in scleroderma: results from a 1-year, proof-of-principle study. Rheumatology (Oxford) 2010;49:271–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Korsten P, Rademacher JG, Riedel L, et al. Antisynthetase syndrome-associated interstitial lung disease: monitoring of immunosuppressive treatment effects by chest computed tomography. Front Med (Lausanne) 2020;7:609595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.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;46:625–631. [DOI] [PubMed] [Google Scholar]
  • 50.Jordan S, Distler JH, Maurer B, et al. ; EUSTAR Rituximab study group. Effects and safety of rituximab in systemic sclerosis: an analysis from the European Scleroderma Trial and Research (EUSTAR) group. Ann Rheum Dis 2015;1188–1194. [DOI] [PubMed] [Google Scholar]
  • 51.Langlois V, Gillibert A, Uzunhan Y, et al. Rituximab and cyclophosphamide in antisynthetase syndrome-related interstitial lung disease: an observational retrospective study. J Rheumatol 2020;47:1678–1686. [DOI] [PubMed] [Google Scholar]
  • 52.Maher TM, Tudor VA, Saunders P, et al. RECITAL Investigators. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): a double-blind, double-dummy, randomised, controlled, phase 2b trial. Lancet Respir Med 2023;11:45–54. [DOI] [PubMed] [Google Scholar]
  • 53.Md Yusof MY, Kabia A, Darby M, et al. Effect of rituximab on the progression of rheumatoid arthritis-related interstitial lung disease: 10 years’ experience at a single centre. Rheumatology (Oxford) 2017;56:1348–1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sircar G, Goswami RP, Sircar D, et al. Intravenous cyclophosphamide vs rituximab for the treatment of early diffuse scleroderma lung disease: open label, randomized, controlled trial. Rheumatology (Oxford) 2018;57:2106–2113. [DOI] [PubMed] [Google Scholar]
  • 55.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;54:3962–3970. [DOI] [PubMed] [Google Scholar]
  • 56.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;354:2655–2666. [DOI] [PubMed] [Google Scholar]
  • 57.Adler S, Huscher D, Siegert E, et al. ; EUSTAR co-workers on behalf of the DeSScipher project research group within the EUSTAR network. Systemic sclerosis associated interstitial lung disease - individualized immunosuppressive therapy and course of lung function: results of the EUSTAR group. Arthritis Res Ther 2018;20:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Airò P, Danieli E, Rossi M, et al. Intravenous cyclophosphamide for interstitial lung disease associated to systemic sclerosis: results with an 18-month long protocol including a maintenance phase. Clin Exp Rheumatol 2007;25:293–296. [PubMed] [Google Scholar]
  • 59.Bruni C, Tashkin DP, Steen V, et al. ; EUSTAR, SLS I & SLS II centres collaborators. Intravenous versus oral cyclophosphamide for lung and/or skin fibrosis in systemic sclerosis: an indirect comparison from EUSTAR and randomised controlled trials. Clin Exp Rheumatol 2020;38 Suppl 125:161–168. [PubMed] [Google Scholar]
  • 60.Davas EM, Peppas C, Maragou M, et al. Intravenous cyclophosphamide pulse therapy for the treatment of lung disease associated with scleroderma. Clin Rheumatol 1999;18:455–461. [DOI] [PubMed] [Google Scholar]
  • 61.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;30:223–229. [DOI] [PubMed] [Google Scholar]
  • 62.Fu Q, Wang L, Li L, et al. Risk factors for progression and prognosis of rheumatoid arthritis-associated interstitial lung disease: single center study with a large sample of Chinese population. Clin Rheumatol 2019;38:1109–1116. [DOI] [PubMed] [Google Scholar]
  • 63.Grau JM, Miró O, Pedrol E, et al. Interstitial lung disease related to dermatomyositis. Comparative study with patients without lung involvement. J Rheumatol 1996;23:1921–1926. [PubMed] [Google Scholar]
  • 64.Nakamura K, Ohbe H, Ikeda K, et al. Intravenous cyclophosphamide in acute exacerbation of rheumatoid arthritis-related interstitial lung disease: a propensity-matched analysis using a nationwide inpatient database. Semin Arthritis Rheum 2021;51:977–982. [DOI] [PubMed] [Google Scholar]
  • 65.Steen VD, Lanz JK Jr, Conte C, et al. Therapy for severe interstitial lung disease in systemic sclerosis. A retrospective study. Arthritis Rheum 1994;37:1290–1296. [DOI] [PubMed] [Google Scholar]
  • 66.Dheda K, Lalloo UG, Cassim B, et al. Experience with azathioprine in systemic sclerosis associated with interstitial lung disease. Clin Rheumatol 2004;23:306–309. [DOI] [PubMed] [Google Scholar]
  • 67.Manfredi A, Cassone G, Furini F, et al. Tocilizumab therapy in rheumatoid arthritis with interstitial lung disease: a multicentre retrospective study. Intern Med J 2020;50:1085–1090. [DOI] [PubMed] [Google Scholar]
  • 68.Nadashkevich O, Davis P, Fritzler M, et al. A randomized unblinded trial of cyclophosphamide versus azathioprine in the treatment of systemic sclerosis. Clin Rheumatol 2006;25:205–212. [DOI] [PubMed] [Google Scholar]
  • 69.Khanna D, Denton CP, Jahreis A, et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial. Lancet 2016;387: 2630–2640. [DOI] [PubMed] [Google Scholar]
  • 70.Khanna D, Lin CJF, Furst DE, et al. ; focuSSced investigators. Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med 2020;8:963–974. [DOI] [PubMed] [Google Scholar]
  • 71.Kuster S, Jordan S, Elhai M, et al. ; EUSTAR collaborators. Effectiveness and safety of tocilizumab in patients with systemic sclerosis: a propensity score matched controlled observational study of the EUSTAR cohort. RMD Open 2022;8:e002477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Roofeh D, Lin CJF, Goldin J, et al. ; focuSSced Investigators. Tocilizumab prevents progression of early systemic sclerosis-associated interstitial lung disease. Arthritis Rheumatol 2021;73:1301–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Suleman Y, Clark KEN, Cole AR, et al. Real-world experience of tocilizumab in systemic sclerosis: potential benefit on lung function for anti-topoisomerase-positive patients. Rheumatology (Oxford) 2021; 60:3945–3946. [DOI] [PubMed] [Google Scholar]
  • 74.McLaren ZL, Dawson JK. Comment on: Factors associated with fatal outcome of leflunomide-induced lung injury in Japanese patients with rheumatoid arthritis. Rheumatology (Oxford) 2010;49: 1606; author reply 1607. [DOI] [PubMed] [Google Scholar]
  • 75.Kiely P, Busby AD, Nikiphorou E, et al. Is incident rheumatoid arthritis interstitial lung disease associated with methotrexate treatment? Results from a multivariate analysis in the ERAS and ERAN inception cohorts. BMJ Open 2019;9:e028466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.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 (Oxford) 2020;59:3906–3916. [DOI] [PubMed] [Google Scholar]
  • 77.Mena-Vázquez N, Rojas-Gimenez M, Fuego-Varela C, et al. Safety and effectiveness of abatacept in a prospective cohort of patients with rheumatoid arthritis-associated interstitial lung disease. Biomedicines 2022;10:1480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.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;9:277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.England BR, Sayles H, Michaud K, et al. Chronic lung disease in U.S. veterans with rheumatoid arthritis and the impact on survival. Clin Rheumatol 2018;37:2907–2915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Juge PA, Lee JS, Lau J, et al. Methotrexate and rheumatoid arthritis associated interstitial lung disease. Eur Respir J 2021;57:2000337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kim JW, Chung SW, Pyo JY, et al. Methotrexate, leflunomide, and tacrolimus use and the progression of rheumatoid arthritis-associated interstitial lung disease. Rheumatology (Oxford) 2023; 62:2377–2385. [DOI] [PubMed] [Google Scholar]
  • 82.Vicente-Rabaneda EF, Atienza-Mateo B, Blanco R, et al. Efficacy and safety of abatacept in interstitial lung disease of rheumatoid arthritis: a systematic literature review. Autoimmun Rev 2021;20: 102830. [DOI] [PubMed] [Google Scholar]
  • 83.Fernández-Díaz C, Atienza-Mateo B, Castañeda S, et al. ; Spanish Collaborative Group of Interstitial Lung Disease Associated with Rheumatoid Arthritis. Abatacept in monotherapy vs combined in interstitial lung disease of rheumatoid arthritis-multicentre study of 263 Caucasian patients. Rheumatology (Oxford) 2021;61:299–308. [DOI] [PubMed] [Google Scholar]
  • 84.Kang EH, Jin Y, Desai RJ, et al. Risk of exacerbation of pulmonary comorbidities in patients with rheumatoid arthritis after initiation of abatacept versus TNF inhibitors: a cohort study. Semin Arthritis Rheum 2020;50:401–408. [DOI] [PubMed] [Google Scholar]
  • 85.Nakashita T, Ando K, Takahashi K, et al. Possible effect of abatacept on the progression of interstitial lung disease in rheumatoid arthritis patients. Respir Investig 2016;54:376–379. [DOI] [PubMed] [Google Scholar]
  • 86.Tardella M, Di Carlo M, Carotti M, et al. A retrospective study of the efficacy of JAK inhibitors or abatacept on rheumatoid arthritis-interstitial lung disease. Inflammopharmacology 2022;30:705–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Tardella M, Di Carlo M, Carotti M, et al. Abatacept in rheumatoid arthritis-associated interstitial lung disease: short-term outcomes and predictors of progression. Clin Rheumatol 2021;40:4861–4867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Dixon WG, Hyrich KL, Watson KD, et al. ; BSRBR Control Centre Consortium; British Society for Rheumatology Biologics Register. Influence of anti-TNF therapy on mortality in patients with rheumatoid arthritis-associated interstitial lung disease: results from the British Society for Rheumatology Biologics Register. Ann Rheum Dis 2010;69:1086–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Huang Y, Lin W, Chen Z, et al. Effect of tumor necrosis factor inhibitors on interstitial lung disease in rheumatoid arthritis: angel or demon? Drug Des Devel Ther 2019;13:2111–2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ma J, Sun J, Wang Y, et al. Efficacy of infliximab plus conventional therapy in dermatomyositis/polymyositis with interstitial lung disease: a prospective cohort study. Int J Clin Exp Med 2017;10: 6819–6827. [Google Scholar]
  • 91.Distler O, Highland KB, Gahlemann M, et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. N Engl J Med 2019; 380:2518–2528. [DOI] [PubMed] [Google Scholar]
  • 92.Li T, Guo L, Chen Z, et al. Pirfenidone in patients with rapidly progressive interstitial lung disease associated with clinically amyopathic dermatomyositis. Sci Rep 2016;6:33226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Solomon JJ, Danoff S, Woodhead F, et al. A randomized, double-blinded, placebo-controlled, phase 2 study of safety, tolerability and efficacy of pirfenidone in patients with rheumatoid arthritis interstitial lung disease. medRxiv 2022:2022.2004.2001.22273270. [DOI] [PubMed] [Google Scholar]
  • 94.Wang J, Wang X, Qi X, et al. The efficacy and safety of pirfenidone combined with immunosuppressant therapy in connective tissue disease-associated interstitial lung disease: a 24-week prospective controlled cohort study. Front Med (Lausanne) 2022;9:871861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Acharya N, Sharma SK, Mishra D, et al. Efficacy and safety of pirfenidone in systemic sclerosis-related interstitial lung disease-a randomised controlled trial. Rheumatol Int 2020;40:703–710. [DOI] [PubMed] [Google Scholar]
  • 96.Behr J, Prasse A, Kreuter M, et al. ; RELIEF investigators. Pirfenidone in patients with progressive fibrotic interstitial lung diseases other than idiopathic pulmonary fibrosis (RELIEF): a double-blind, randomised, placebo-controlled, phase 2b trial. Lancet Respir Med 2021;9: 476–486. [DOI] [PubMed] [Google Scholar]
  • 97.Sato T, Inokuma S, Sagawa A, et al. ; Study Committee for Leflunomide-induced Lung Injury, Japan College of Rheumatology. Factors associated with fatal outcome of leflunomide-induced lung injury in Japanese patients with rheumatoid arthritis. Rheumatology (Oxford) 2009;48:1265–1268. [DOI] [PubMed] [Google Scholar]
  • 98.Hallowell RW, Amariei D, Danoff SK. Intravenous immunoglobulin as potential adjunct therapy for interstitial lung disease. Ann Am Thorac Soc 2016;13:1682–1688. [DOI] [PubMed] [Google Scholar]
  • 99.Huapaya JA, Hallowell R, Silhan L, et al. Long-term treatment with human immunoglobulin for antisynthetase syndrome-associated interstitial lung disease. Respir Med 2019;154:6–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wang LM, Yang QH, Zhang L, et al. Intravenous immunoglobulin for interstitial lung diseases of anti-melanoma differentiation-associated gene 5-positive dermatomyositis. Rheumatology (Oxford, England). [DOI] [PubMed] [Google Scholar]
  • 101.Bay P, de Chambrun MP, Rothstein V, et al. Efficacy of plasma exchange in patients with anti-MDA5 rapidly progressive interstitial lung disease. J Autoimmun 2022;133:102941. [DOI] [PubMed] [Google Scholar]
  • 102.Shirakashi M, Nakashima R, Tsuji H, et al. Efficacy of plasma exchange in anti-MDA5-positive dermatomyositis with interstitial lung disease under combined immunosuppressive treatment. Rheumatology (Oxford, England) 2020;59:3284–3292. [DOI] [PubMed] [Google Scholar]
  • 103.Wilkes MR, Sereika SM, Fertig N, et al. Treatment of antisynthetase-associated interstitial lung disease with tacrolimus. Arthritis Rheum 2005;52:2439–2446. [DOI] [PubMed] [Google Scholar]
  • 104.Wu W, Guo L, Fu Y, et al. Interstitial lung disease in Anti-MDA5 positive dermatomyositis. Clin Rev Allergy Immunol 2021;60:293–304. [DOI] [PubMed] [Google Scholar]
  • 105.Okamoto M, Fujimoto K, Sadohara J, et al. A retrospective cohort study of outcome in systemic sclerosis-associated interstitial lung disease. Respir Investig 2016;54:445–453. [DOI] [PubMed] [Google Scholar]
  • 106.Kurita T, Yasuda S, Oba K, et al. The efficacy of tacrolimus in patients with interstitial lung diseases complicated with polymyositis or dermatomyositis. Rheumatology (Oxford) 2015;54:39–44. [DOI] [PubMed] [Google Scholar]
  • 107.Lian L, Li M, Li Y, et al. Combination therapy of tacrolimus, high doses of glucocorticosteroids, and cyclophosphamide against existing historical treatment for patients in severe conditions of interstitial lung diseases complicated with dermatomyositis: a retrospective analysis. Medicine (Baltimore) 2022;101:e29108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Fan L, Lyu W, Liu H, et al. A retrospective analysis of outcome in melanoma differentiation-associated gene 5-related interstitial lung disease treated with tofacitinib or tacrolimus. J Rheumatol 2022; 49:1356–1364. [DOI] [PubMed] [Google Scholar]
  • 109.Chen Z, Wang X, Ye S. Tofacitinib in amyopathic dermatomyositis-associated interstitial lung disease. N Engl J Med 2019;381: 291–293. [DOI] [PubMed] [Google Scholar]
  • 110.Kurasawa K, Arai S, Namiki Y, et al. Tofacitinib for refractory interstitial lung diseases in anti-melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatology (Oxford) 2018; 57:2114–2119. [DOI] [PubMed] [Google Scholar]
  • 111.Chen Y, Bai Z, Zhang Z, et al. The efficacy and safety of tacrolimus on top of glucocorticoids in the management of IIM-ILD: a retrospective and prospective study. Front Immunol 2022;13:978429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Furuya H, Nakajima M, Ikeda K, et al. Prognosis and treatment of myositis-associated severe interstitial lung disease: a descriptive study using a nationwide inpatient database in Japan. Arthritis Care Res (Hoboken) 2022;74:478–483. [DOI] [PubMed] [Google Scholar]
  • 113.Vuillard C, Pineton de Chambrun M, de Prost N, et al. Clinical features and outcome of patients with acute respiratory failure revealing anti-synthetase or anti-MDA-5 dermato-pulmonary syndrome: a French multicenter retrospective study. Ann Intensive Care 2018; 8:87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Abe Y, Kusaoi M, Tada K, et al. Successful treatment of anti-MDA5 antibody-positive refractory interstitial lung disease with plasma exchange therapy. Rheumatology (Oxford) 2020;59:767–771. [DOI] [PubMed] [Google Scholar]
  • 115.Komai T, Iwasaki Y, Tsuchida Y, et al. Efficacy and safety of plasma exchange in interstitial lung diseases with anti-melanoma differentiation-associated 5 gene antibody positive clinically amyopathic dermatomyositis. Scand J Rheumatol 2023;52:77–83. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp1
Supp2
Supp4
Supp5
Supp6
Supp3

RESOURCES