826
Views
9
CrossRef citations to date
0
Altmetric
Review

Clinical trials and novel therapeutics in dermatomyositis

&
Pages 213-228 | Received 03 Apr 2020, Accepted 23 Jun 2020, Published online: 10 Jul 2020

References

  • Ceribelli A, De Santis M, Isailovic N, et al. The immune response and the pathogenesis of idiopathic inflammatory myositis: a critical review. Clin Rev Allergy Immunol. 2017 [cited 2016 Jan 19]:52:58–70.
  • Rider LG, Werth VP, Huber AM, et al. Measures of adult and juvenile dermatomyositis, polymyositis, and inclusion body myositis: physician and patient/parent global activity, manual muscle testing (MMT), health assessment questionnaire (HAQ)/childhood health assessment questionnaire (C-HAQ), childhood myositis assessment scale (CMAS), myositis disease activity assessment tool (MDAAT), disease activity score (DAS), short form 36 (SF-36), child health questionnaire (CHQ), physician global damage, myositis damage index (MDI), quantitative muscle testing (QMT), myositis functional index-2 (FI-2), myositis activities profile (MAP), inclusion body myositis functional rating scale (IBMFRS), cutaneous dermatomyositis disease area and severity index (CDASI), cutaneous assessment tool (CAT), dermatomyositis skin severity index (DSSI), skindex, and dermatology life quality index (DLQI). Arthritis Care Res (Hoboken). 2011 [cited 2012 May 25];63(Suppl 11):S118–157.
  • Aggarwal R, Rider LG, Ruperto N, et al. 2016 American College of Rheumatology/European league against rheumatism criteria for minimal, moderate, and major clinical response in adult dermatomyositis and polymyositis: an international myositis assessment and clinical studies group/paediatric rheumatology international trials organisation collaborative initiative. Arthritis Rheumatol. 2017 [cited 2017 Apr 07];69:898–910.
  • Rider LG, Aggarwal R, Pistorio A, et al. American College of Rheumatology/European league against rheumatism criteria for minimal, moderate, and major clinical response in juvenile dermatomyositis: an international myositis assessment and clinical studies group/paediatric rheumatology international trials organisation collaborative initiative. Arthritis Rheumatol. 2016;69:911–923.
  • Aggarwal R, Rider LG, Ruperto N, et al. 2016 American College of Rheumatology/European league against rheumatism criteria for minimal, moderate, and major clinical response in adult dermatomyositis and polymyositis: an international myositis assessment and clinical studies group/paediatric rheumatology international trials organisation collaborative initiative. Ann Rheum Dis. 2017 [2017 Apr 08];76:792–801.
  • Rider LG, Aggarwal R, Pistorio A, et al. 2016 American College of Rheumatology/European league against rheumatism criteria for minimal, moderate, and major clinical response in juvenile dermatomyositis: an international myositis assessment and clinical studies group/paediatric rheumatology international trials organisation collaborative initiative. Ann Rheum Dis. 2017 [cited 2017 Apr 08];76:782–791.
  • Rider LG, Ruperto N, Pistorio A, et al. 2016 ACR-EULAR adult dermatomyositis and polymyositis and juvenile dermatomyositis response criteria-methodological aspects. Rheumatology (Oxford). 2017 [cited 2017 Oct 05];56:1884–1893.
  • Rider LG, Giannini EH, Brunner HI, et al. International consensus on preliminary definitions of improvement in adult and juvenile myositis. Arthritis Rheum. 2004 [ 2004/07/13];50:2281–2290.
  • Ruperto N, Pistorio A, Ravelli A, et al. The paediatric rheumatology international trials organisation provisional criteria for the evaluation of response to therapy in juvenile dermatomyositis. Arthritis Care Res (Hoboken). 2010 [cited 2010 Jun 29];62:1533–1541.
  • Lundberg IE, Tjarnlund A, Bottai M, et al. 2017 European league against rheumatism/American College of Rheumatology classification criteria for adult and juvenile idiopathic inflammatory myopathies and their major subgroups. Ann Rheum Dis. 2017 [cited 2017Oct 29];76:1955–1964.
  • Bottai M, Tjarnlund A, Santoni G, et al. EULAR/ACR classification criteria for adult and juvenile idiopathic inflammatory myopathies and their major subgroups: a methodology report. RMD Open. 2017 [ 2017/ 11/28];3:e000507.
  • Anyanwu CO, Fiorentino DF, Chung L, et al. Validation of the cutaneous dermatomyositis disease area and severity index: characterizing disease severity and assessing responsiveness to clinical change. Br J Dermatol. 2015 [ 2015/ 05/23];173:969–974.
  • Rider LG, Aggarwal R, Machado PM, et al. Update on outcome assessment in myositis. Nat Rev Rheumatol. 2018 [cited 2018 Apr 14]:14:303–318.
  • Saygin D, Oddis C, Neiman N, et al. Reliability, validity and responsiveness of PROMIS PF-20 in patients with inflammatory myopathy [abstract]. Arthritis Rheumatol. 2019;71:5037-5039(suppl 10).
  • Ernste FC, Chong C, Crowson CS, et al. Functional index-3: a valid and reliable functional outcome assessment measure in dermatomyositis and polymyositis patients. J Rheumatol. 2020 [cited 2020 Apr 17]. DOI:10.3899/jrheum.191374
  • Brunner E, Tasan L, Torok KS, et al. Stepping it up: the use of physical activity monitors as an outcome measure in juvenile myositis [abstract]. Arthritis Rheumatol. 2018;70:516-517(suppl 10).
  • Chiu YE, Co DO. Juvenile dermatomyositis: immunopathogenesis, role of myositis-specific autoantibodies, and review of rituximab use. Pediatr Dermatol. 2011 [cited 2011 Jul 29];28:357–367.
  • Kikuchi Y, Koarada S, Tada Y, et al. Difference in B cell activation between dermatomyositis and polymyositis: analysis of the expression of RP105 on peripheral blood B cells. Ann Rheum Dis. 2001 [cited2001 Nov 16];60:1137–1140.
  • Oddis CV, Reed AM, Aggarwal R, et al. Rituximab in the treatment of refractory adult and juvenile dermatomyositis and adult polymyositis: a randomized, placebo-phase trial. Arthritis Rheum. 2013 [cited 2012 Nov 06]:65:314–324.
  • Levine TD. Rituximab in the treatment of dermatomyositis: an open-label pilot study. Arthritis Rheum. 2005 [ 2005/ 02/05];52:601–607.
  • Chung L, Genovese MC, Fiorentino DF. A pilot trial of rituximab in the treatment of patients with dermatomyositis. Arch Dermatol. 2007 [cited 2007 Jun 20];143:763–767.
  • Mahler EA, Blom M, Voermans NC, et al. Rituximab treatment in patients with refractory inflammatory myopathies. Rheumatology (Oxford). 2011 [cited 2011 May 17];50:2206–2213.
  • Noss EH, Hausner-Sypek DL, Weinblatt ME. Rituximab as therapy for refractory polymyositis and dermatomyositis. J Rheumatol. 2006 [cited 2006 Mar 17];33:1021–1026.
  • Chiappetta N, Steier J, Gruber B. Rituximab in the treatment of refractory dermatomyositis. J Clin Rheumatol. 2005 [cited 2005 Dec 17];11:264–266.
  • Lambotte O, Kotb R, Maigne G, et al. Efficacy of rituximab in refractory polymyositis. J Rheumatol. 2005 [cited 2005 Jul 05];32:1369–1370.
  • Dinh HV, McCormack C, Hall S, et al. Rituximab for the treatment of the skin manifestations of dermatomyositis: a report of 3 cases. J Am Acad Dermatol. 2007 [cited 2006 Nov 14];56:148–153.
  • Touma Z, Arayssi T, Kibbi L, et al. Successful treatment of cardiac involvement in dermatomyositis with rituximab. Joint Bone Spine. 2008 [cited 2007 Nov 09];75:334–337.
  • Feist E, Dorner T, Sorensen H, et al. Longlasting remissions after treatment with rituximab for autoimmune myositis. J Rheumatol. 2008 [cited 2008 Jun 06];35:1230–1232.
  • Sultan SM, Ng KP, Edwards JC, et al. Clinical outcome following B cell depletion therapy in eight patients with refractory idiopathic inflammatory myopathy. Clin Exp Rheumatol. 2008 [cited 2008 Nov 27];26:887–893.
  • Kaposztas Z, Etheridge WB, Kahan BD. Case report: successful treatment of posttransplant lymphoproliferative disorder and quiescence of dermatomyositis with rituximab and sirolimus. Transplant Proc. 2008 [cited 2008 Jul 01];40:1744–1746.
  • Haroon M, Devlin J. Rituximab as a first-line agent for the treatment of dermatomyositis. Rheumatol Int. 2012 [cited 2010 Mar 27];32:1783–1784.
  • Parziale N, Kovacs SC, Thomas CB, et al. Rituximab and mycophenolate combination therapy in refractory dermatomyositis with multiple autoimmune disorders. J Clin Neuromuscul Dis. 2011 [cited 2012 Mar 01];13:63–67.
  • Unger L, Kampf S, Luthke K, et al. Rituximab therapy in patients with refractory dermatomyositis or polymyositis: differential effects in a real-life population. Rheumatology (Oxford). 2014 [cited 2014 Apr 08];53:1630–1638.
  • Aggarwal R, Loganathan P, Koontz D, et al. Cutaneous improvement in refractory adult and juvenile dermatomyositis after treatment with rituximab. Rheumatology (Oxford). 2017 [cited 2016 Nov 12];56:247–254.
  • Aggarwal R, Bandos A, Reed AM, et al. Predictors of clinical improvement in rituximab-treated refractory adult and juvenile dermatomyositis and adult polymyositis. Arthritis Rheumatol. 2014 [cited 2014 Feb 28];66:740–749.
  • Tepper MA, Zurier RB, Burstein SH. Ultrapure ajulemic acid has improved CB2 selectivity with reduced CB1 activity. Bioorg Med Chem. 2014 [ 2014/05/27];22:3245–3251.
  • Bouaboula M, Rinaldi M, Carayon P, et al. Cannabinoid-receptor expression in human leukocytes. Eur J Biochem. 1993 [cited 1993 May 15];214:173–180.
  • Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993 [cited 1993 Sep 02];365:61–65.
  • Burstein SH. Ajulemic acid: potential treatment for chronic inflammation. Pharmacol Res Perspect. 2018 [cited 2018 Apr 12];6:e00394.
  • Kim JS, Bashir MM, Werth VP. Gottron’s papules exhibit dermal accumulation of CD44 variant 7 (CD44v7) and its binding partner osteopontin: a unique molecular signature. J Invest Dermatol. 2012 [cited 2012 Mar 30];132:1825–1832.
  • Nabatian AS, Bashir MM, Wysocka M, et al. Tumor necrosis factor alpha release in peripheral blood mononuclear cells of cutaneous lupus and dermatomyositis patients. Arthritis Res Ther. 2012 [cited 2012 Jan 06];14:R1.
  • Robinson ES, Alves P, Bashir MM, et al. Cannabinoid reduces inflammatory cytokines, tumor necrosis factor-alpha, and type i interferons in dermatomyositis in vitro. J Invest Dermatol. 2017 [cited 2017 Jun 28];137:2445–2447.
  • Kim HJ, Zeidi M, Bonciani D, et al. Itch in dermatomyositis: the role of increased skin interleukin-31. Br J Dermatol. 2018 [cited 2018 Mar 02];179(3):669–678.
  • Chen K, Zeidi M, Reddy N, et al. Fri0307 lenabasum, a cannabinoid type 2 receptor agonist, reduces Cd4 cell populations and downregulates type 1 and 2 interferon activities in lesional dermatomyositis skin. Ann Rheum Dis. 2019;78:835.
  • Werth V, Hejazi E, Pena S, et al. Phase 2 study of safety and efficacy of lenabasum (JBT-101), a cannabinoid receptor type 2 agonist, in refractory skin-predominant dermatomyositis. Ann Rheum Dis. 2018;77:763–764.
  • Werth VP, Pearson D, Okawa J, et al. Safety and efficacy of lenabasum in refractory skin-predominant dermatomyositis subjects treated on an open-label extension of trial JBT101-DM-001 [abstract]. Arthritis Rheumatol. 2018;70:2524-2526(suppl):10.
  • Thompson C, Piguet V, Choy E. The pathogenesis of dermatomyositis. Br J Dermatol. 2018 [ 2017/ 05/26];179:1256–1262.
  • Giris M, Durmus H, Yetimler B, et al. Elevated IL-4 and IFN-gamma levels in muscle tissue of patients with dermatomyositis. in Vivo. 2017 [cited 2017 Jun 28];31:657–660.
  • Charlton D, Moghadam-Kia S, Smith K, et al. Refractory cutaneous dermatomyositis with severe scalp pruritus responsive to apremilast. J Clin Rheumatol. 2019 [cited 2019 Feb 26]. DOI:10.1097/RHU.0000000000000999
  • Bitar C, Maghfour J, Ho-Pham H, et al. Apremilast as a potential treatment for moderate to severe dermatomyositis: a retrospective study of 3 patients. JAAD Case Rep. 2019 [cited 2019 Feb 12];5:191–194.
  • Bilgic H, Ytterberg SR, Amin S, et al. Interleukin-6 and type I interferon-regulated genes and chemokines mark disease activity in dermatomyositis. Arthritis Rheum. 2009;60(11):3436–3446. .
  • Greenberg SA, Pinkus JL, Pinkus GS, et al. Interferon-alpha/beta-mediated innate immune mechanisms in dermatomyositis. Ann Neurol. 2005 [cited 2005 Apr 27];57:664–678.
  • Sanner H, Schwartz T, Flato B, et al. Increased levels of eotaxin and MCP-1 in juvenile dermatomyositis median 16.8 years after disease onset; associations with disease activity, duration and organ damage. PLoS One. 2014 [cited 2014 Mar 22];9:e92171.
  • Crescioli C, Sottili M, Bonini P, et al. Inflammatory response in human skeletal muscle cells: CXCL10 as a potential therapeutic target. Eur J Cell Biol. 2012 [cited 2011 Dec 20];91:139–149.
  • Pinal-Fernandez I, Casal-Dominguez M, Derfoul A, et al. Identification of distinctive interferon gene signatures in different types of myositis. Neurology. 2019 [cited 2019 Oct 23];93:e1193–e1204.
  • Narain S, Furie R. Update on clinical trials in systemic lupus erythematosus. Curr Opin Rheumatol. 2016 [cited 2016 Jun 18];28:477–487.
  • Morand EF, Furie R, Tanaka Y, et al. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med. 2020 [cited 2019 Dec 19];382:211–221.
  • Higgs BW, Zhu W, Morehouse C, et al. A phase 1b clinical trial evaluating sifalimumab, an anti-IFN-alpha monoclonal antibody, shows target neutralisation of a type I IFN signature in blood of dermatomyositis and polymyositis patients. Ann Rheum Dis. 2014 [cited 2013 Feb 26];73:256–262.
  • Guo X, Higgs BW, Rebelatto M, et al. Suppression of soluble T cell-associated proteins by an anti-interferon-alpha monoclonal antibody in adult patients with dermatomyositis or polymyositis. Rheumatology (Oxford). 2014 [cited 2013 Dec 21];53:686–695.
  • Ogorevc E, Schiffrer ES, Sosic I, et al. A patent review of immunoproteasome inhibitors. Expert Opin Ther Pat. 2018 [cited 2018 Jun 06]:28:517–540.
  • Ghannam K, Martinez-Gamboa L, Spengler L, et al. Upregulation of immunoproteasome subunits in myositis indicates active inflammation with involvement of antigen presenting cells, CD8 T-cells and IFNGamma. PLoS One. 2014 [cited 2014 Aug 08];9:e104048.
  • Bhattarai S, Ghannam K, Krause S, et al. The immunoproteasomes are key to regulate myokines and MHC class I expression in idiopathic inflammatory myopathies. J Autoimmun. 2016 [cited 2016 Aug 16];75:118–129.
  • Muchamuel T, Basler M, Aujay MA, et al. A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nat Med. 2009 [cited 2009 Jun 16];15:781–787.
  • Ichikawa HT, Conley T, Muchamuel T, et al. Beneficial effect of novel proteasome inhibitors in murine lupus via dual inhibition of type I interferon and autoantibody-secreting cells. Arthritis Rheum. 2012 [cited 2011 Sep 10];64:493–503.
  • Basler M, Mundt S, Muchamuel T, et al. Inhibition of the immunoproteasome ameliorates experimental autoimmune encephalomyelitis. EMBO Mol Med. 2014 [cited 2014 Jan 09];6:226–238.
  • Basler M, Dajee M, Moll C, et al. Prevention of experimental colitis by a selective inhibitor of the immunoproteasome. J Immunol. 2010 [cited 2010 Jun 08];185:634–641.
  • Kalim KW, Basler M, Kirk CJ, et al. Immunoproteasome subunit LMP7 deficiency and inhibition suppresses Th1 and Th17 but enhances regulatory T cell differentiation. J Immunol. 2012 [cited 2012 Sep 18];189:4182–4193.
  • Nakamura K, Jinnin M, Kudo H, et al. The role of PSMB9 upregulated by interferon signature in the pathophysiology of cutaneous lesions of dermatomyositis and systemic lupus erythematosus. Br J Dermatol. 2016 [cited 2015 Dec 30];174:1030–1041.
  • Johnson HWB, Lowe E, Anderl JL, et al. Required immunoproteasome subunit inhibition profile for anti-inflammatory efficacy and clinical candidate KZR-616 ((2 S,3 R)- N-((S)-3-(Cyclopent-1-en-1-yl)-1-((R)-2-methyloxiran-2-yl)-1-oxopropan-2-yl)-3-hydroxy-3-(4-methoxyphenyl)-2-((S)-2-(2-morpholinoacetamido)propanamido)propenamide). J Med Chem. 2018 [cited 2018 Nov 02];61:11127–11143.
  • Furie R, Bomba D, Dall’Era M, et al. Treatment of SLE patients with the immunoproteasome inhibitor KZR-616: results from the first 3 cohorts of an open-label phase 1b dose escalation trial [abstract]. Arthritis Rheumatol. 2019;71:4468-4470(suppl 10).
  • Lickliter J, Anderl J, Kirk CJ, et al. KZR-616, a selective Inhibitor of the immunoproteasome, shows a promising safety and target inhibition profile in a phase I, double-blind, single (SAD) and multiple ascending dose (MAD) study in healthy volunteers [abstract]. Arthritis Rheumatol. 2017;69:3727-3728(suppl 10).
  • Muangchan C, Pope JE. Interleukin 6 in systemic sclerosis and potential implications for targeted therapy. J Rheumatol. 2012 [cited 2012 Apr 17];39:1120–1124.
  • Baechler EC, Bauer JW, Slattery CA, et al. An interferon signature in the peripheral blood of dermatomyositis patients is associated with disease activity. Mol Med. 2007 [cited 2007 May 23];13:59–68.
  • Gabay C. Interleukin-6 and chronic inflammation. Arthritis Res Ther. 2006 [ 2006/08/11];8(Suppl 2):S3.
  • Nam NH. Naturally occurring NF-kappaB inhibitors. Mini Rev Med Chem. 2006 [cited 2006 Aug 22];6:945–951.
  • Okiyama N, Sugihara T, Iwakura Y, et al. Therapeutic effects of interleukin-6 blockade in a murine model of polymyositis that does not require interleukin-17A. Arthritis Rheum. 2009 [cited 2009 Aug 01];60:2505–2512.
  • Beaumel A, Muis-Pistor O, Tebib JG, et al. Antisynthetase syndrome treated with tocilizumab. Joint Bone Spine. 2016 [cited 2015 Oct 17];83:361–362.
  • Narazaki M, Hagihara K, Shima Y, et al. Therapeutic effect of tocilizumab on two patients with polymyositis. Rheumatology (Oxford). 2011 [cited 2011 Apr 26];50:1344–1346.
  • Murphy SM, Lilleker JB, Helliwell P, et al. The successful use of tocilizumab as third-line biologic therapy in a case of refractory anti-synthetase syndrome. Rheumatology (Oxford). 2016 [cited 2016 Aug 24];55:2277–2278.
  • Kondo M, Murakawa Y, Matsumura T, et al. A case of overlap syndrome successfully treated with tocilizumab: a hopeful treatment strategy for refractory dermatomyositis? Rheumatology (Oxford). 2014 [cited 2014 May 27];53:1907–1908.
  • Rosengren S, Corr M, Firestein GS, et al. The JAK inhibitor CP-690,550 (tofacitinib) inhibits TNF-induced chemokine expression in fibroblast-like synoviocytes: autocrine role of type I interferon. Ann Rheum Dis. 2012 [cited 2011 Nov 29];71:440–447.
  • Ladislau L, Suarez-Calvet X, Toquet S, et al. JAK inhibitor improves type I interferon induced damage: proof of concept in dermatomyositis. Brain. 2018 [ 2018/ 05/10];141:1609–1621.
  • Kurtzman DJ, Wright NA, Lin J, et al. Tofacitinib citrate for refractory cutaneous dermatomyositis: an alternative treatment. JAMA Dermatol. 2016 [ 2016/ 04/28];152:944–945.
  • Moghadam-Kia S, Charlton D, Aggarwal R, et al. Management of refractory cutaneous dermatomyositis: potential role of Janus kinase inhibition with tofacitinib. Rheumatology (Oxford). 2019 [cited 2019 Jan 05];58:1011–1015.
  • Sabbagh S, Almeida de Jesus A, Hwang S, et al. Treatment of anti-MDA5 autoantibody-positive juvenile dermatomyositis using tofacitinib. Brain. 2019 [cited 2019 Oct 12];142:e59.
  • Chen Z, Wang X, Ye S. Tofacitinib in amyopathic dermatomyositis-associated interstitial lung disease. N Engl J Med. 2019 [cited 2019 Jul 18];381:291–293.
  • 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 [cited 2018 Jul 31];57:2114–2119.
  • Wendel S, Venhoff N, Frye BC, et al. Successful treatment of extensive calcifications and acute pulmonary involvement in dermatomyositis with the Janus-Kinase inhibitor tofacitinib – a report of two cases. J Autoimmun. 2019 [cited 2019 Mar 14];100:131–136.
  • Paik JJ, Christopher-Stine L. A case of refractory dermatomyositis responsive to tofacitinib. Semin Arthritis Rheum. 2017 [ 2016/ 09/20];46:e19.
  • Paik JJ, Albayda J, Tiniakou E, et al. Study of tofacitinib in refractory dermatomyositis (STIR): an open label pilot study in refractory dermatomyositis [abstract]. Arthritis Rheumatol. 2018;70(suppl 10). https://acrabstracts.org/abstract/study-of-tofacitinib-in-refractory-dermatomyositis-stir-an-open-label-pilot-study-in-refractory-dermatomyositis/
  • Syn NL, Teng MWL, Mok TSK, et al. De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol. 2017 [cited 2017 Dec 07];18:e731–e741.
  • Nagaraju K, Raben N, Villalba ML, et al. Costimulatory markers in muscle of patients with idiopathic inflammatory myopathies and in cultured muscle cells. Clin Immunol. 1999 [cited 1999 Aug 13];92:161–169.
  • Murata K, Dalakas MC. Expression of the costimulatory molecule BB-1, the ligands CTLA-4 and CD28, and their mRNA in inflammatory myopathies. Am J Pathol. 1999 [cited 1999 Aug 06];155:453–460.
  • Cutolo M, Soldano S, Montagna P, et al. CTLA4-Ig interacts with cultured synovial macrophages from rheumatoid arthritis patients and downregulates cytokine production. Arthritis Res Ther. 2009 [cited 2009 Nov 26];11:R176.
  • Musuruana JL, Cavallasca JA. Abatacept for treatment of refractory polymyositis. Joint Bone Spine. 2011 [cited 2011 May 10];78:431–432.
  • Kerola AM, Kauppi MJ. Abatacept as a successful therapy for myositis – a case-based review. Clin Rheumatol. 2015 [cited 2014 feb 05];34:609–612.
  • Arabshahi B, Silverman RA, Jones OY, et al. Abatacept and sodium thiosulfate for treatment of recalcitrant juvenile dermatomyositis complicated by ulceration and calcinosis. J Pediatr. 2012 [cited 2012 Jan 17];160:520–522.
  • Rodziewicz M, Kiely P. The successful use of subcutaneous abatacept in refractory anti- human transcriptional intermediary factor 1-gamma dermatomyositis skin and oesphagopharyngeal disease. Rheumatology (Oxford). 2018 [cited 2018 May 26];57:1866–1867.
  • Tjarnlund A, Tang Q, Wick C, et al. Abatacept in the treatment of adult dermatomyositis and polymyositis: a randomised, phase IIb treatment delayed-start trial. Ann Rheum Dis. 2018 [cited 2017 Oct 11];77(1):55–62.
  • Basta M, Dalakas MC. High-dose intravenous immunoglobulin exerts its beneficial effect in patients with dermatomyositis by blocking endomysial deposition of activated complement fragments. J Clin Invest. 1994;94:1729–1735.
  • Dalakas MCI, Illa JM, Dambrosia SA, et al. A controlled trial of high dose intravenous immunoglobulin infusions as treatment for dermatomyositis. N Engl J Med. 1993;329:1993–2000.
  • Amemiya K, Semino-Mora C, Granger RP, et al. Downregulation of TGF-beta1 mRNA and protein in the muscles of patients with inflammatory myopathies after treatment with high-dose intravenous immunoglobulin. Clin Immunol. 2000;94:99–104.
  • Raju R, Dalakas MC. Gene expression profile in the muscles of patients with inflammatory myopathies: effect of therapy with IVIg and biological validation of clinically relevant genes. Brain. 2005;128(Pt8):1887–1896.
  • Abdou NI, Wall H, Lindsley HB, et al. Network theory in autoimmunity. In vitro suppression of serum anti-DNA antibody binding to DNA by anti-idiotypic antibody in systemic lupus erythematosus. J Clin Invest. 1981;67:1297–1304.
  • Anderson CL. Human IgG Fc receptors. Clin Immunol Immunopathol. 1989 [cited 1989 Nov 01];53:S63–71.
  • Dalakas MC. Intravenous immune globulin therapy for neurologic diseases. Ann Intern Med. 1997;126:721–730.
  • Kurlander RJ. Reversible and irreversible loss of Fc receptor function of human monocytes as a consequence of interaction with immunoglobulin G. J Clin Invest. 1980 [cited 1980 Oct 01];66:773–781.
  • Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science. 2001 [cited 2001 Feb 13];291:484–486.
  • Anh-Tu Hoa S, Hudson M. Critical review of the role of intravenous immunoglobulins in idiopathic inflammatory myopathies. Semin Arthritis Rheum. 2017 [cited 2016 Dec 03];46:488–508.
  • Cherin P, Piette JC, Wechsler B, et al. Intravenous gamma globulin as first line therapy in polymyositis and dermatomyositis: an open study in 11 adult patients. J Rheumatol. 1994 [cited 1994 Jun 01];21:1092–1097.
  • Miyasaka N, Hara M, Koike T, et al. Effects of intravenous immuno-globulin therapy in Japanese patients with polymyositis and dermatomyositis resistant to corticosteroids: a randomized double-blind placebo-controlled trial. Mod Rheumatol. 2012;22(3):382–393.
  • Patwa HS, Chaudhry V, Katzberg H, et al. Evidence-based guideline: intravenous immunoglobulin in the treatment of neuromuscular disorders: report of the therapeutics and technology assessment subcommittee of the American Academy of Neurology. Neurology. 2012 [cited 2012 Mar 29];78:1009–1015.
  • Wang DX, Shu XM, Tian XL, et al. Intravenous immunoglobulin therapy in adult patients with polymyositis/dermatomyositis: a systematic literature review. Clin Rheumatol. 2012 [cited 2012 Jan 26];31:801–806.
  • Tian J, Gao JS, Chen JW, et al. Efficacy and safety of the combined treatment with intravenous immunoglobulin and oral glucocorticoid in the elderly with dermatomyositis. Chin J Geriatr. 2008;27:588–590.
  • Gottfried I, Seeber A, Anegg B, et al. High dose intravenous immunoglobulin (IVIG) in dermatomyositis: clinical responses and effect on sIL-2R levels. Eur J Dermatol. 2000 [cited 2000 Feb 29];10:29–35.
  • Cherin P, Pelletier S, Teixeira A, et al. Results and long-term followup of intravenous immunoglobulin infusions in chronic, refractory polymyositis: an open study with thirty-five adult patients. Arthritis Rheum. 2002 [cited 2002 Feb 13];46:467–474.
  • Danieli MG, Malcangi G, Palmieri C, et al. Cyclosporin A and intravenous immunoglobulin treatment in polymyositis/dermatomyositis. Ann Rheum Dis. 2002 [cited 2002 Jan 10];61:37–41.
  • Danieli MG, Gambini S, Pettinari L, et al. Impact of treatment on survival in polymyositis and dermatomyositis. A single-centre long-term follow-up study. Autoimmun Rev. 2014 [cited 2014 Sep 04];13:1048–1054.
  • Danieli MG, Pettinari L, Moretti R, et al. Subcutaneous immunoglobulin in polymyositis and dermatomyositis: a novel application. Autoimmun Rev. 2011 [cited 2010 Sep 23];10:144–149.
  • Danieli MG, Gelardi C, Pedini V, et al. Subcutaneous IgG in immune-mediate diseases: proposed mechanisms of action and literature review. Autoimmun Rev. 2014 [cited 2014 Aug 31];13:1182–1188.
  • Stiehm ER. Adverse effects of human immunoglobulin therapy. Transfus Med Rev. 2013 [cited 2013 Sep 10];27:171–178.
  • Catania A, Lonati C, Sordi A, et al. The melanocortin system in control of inflammation. ScientificWorldJournal. 2010 [cited 2010 Sep 21]:10:1840–1853.
  • Getting SJ, Christian HC, Flower RJ, et al. Activation of melanocortin type 3 receptor as a molecular mechanism for adrenocorticotropic hormone efficacy in gouty arthritis. Arthritis Rheum. 2002;46(10):2765–2775.
  • Catania A, Gatti S, Colombo G, et al. Targeting melanocortin receptors as a novel strategy to control inflammation. Pharmacol Rev. 2004 [cited 2004 Mar 06];56:1–29.
  • Dores RM. Adrenocorticotropic hormone, melanocyte-stimulating hormone, and the melanocortin receptors: revisiting the work of Robert Schwyzer: a thirty-year retrospective. Ann N Y Acad Sci. 2009 [cited 2009 May 22];1163:93–100.
  • Xing Y, Parker CR, Edwards M, et al. ACTH is a potent regulator of gene expression in human adrenal cells. J Mol Endocrinol. 2010 [cited 2010 May 13];45:59–68.
  • Levine T. Treating refractory dermatomyositis or polymyositis with adrenocorticotropic hormone gel: a retrospective case series. Drug Des Devel Ther. 2012;6:133–139.
  • Patel A, Seely G, Aggarwal R. Repository corticotropin injection for treatment of idiopathic inflammatory myopathies. Case Rep Rheumatol. 2016;2016:9068061.
  • Wolff M, Mancuso C, Lal K, et al. Paraneoplastic dermatomyositis with cutaneous and myopathic disease responsive to adrenocorticotropic hormone Therapy. J Clin Aesthet Dermatol. 2017;10(1):57–62.
  • Levine T, Malone M, Efthimiou P, et al. H.P. Acthar® gel in dermatomyositis and polymyositis treatment registry: an interim analysis. J Neurol Disord. 2016;4:292.
  • Aggarwal R, Marder G, Koontz DC, et al. Efficacy and safety of adrenocorticotropic hormone gel in refractory dermatomyositis and polymyositis. Ann Rheum Dis. 2018 [cited 2017 Dec 15]:77:720–727.
  • Saygin D, Oddis CV, Marder G, et al. Follow-up results of myositis patients treated with H. P. Acthar gel. Rheumatology (Oxford). 2020 [cited 2020 Mar 12]. DOI:10.1093/rheumatology/keaa076
  • Lal R, Bell S, Challenger R, et al. Pharmacodynamics and tolerability of repository corticotropin injection in healthy human subjects: a comparison with intravenous methylprednisolone. J Clin Pharmacol. 2016;56(2):195–202.
  • Furie R, Mitrane M, Zhao E, et al. Efficacy and tolerability of repository corticotropin injection in patients with persistently active SLE: results of a phase 4, randomised, controlled pilot study. Lupus Sci Med. 2016;3(1):e000180.
  • Fiechtner JJ, Montroy T. Treatment of moderately to severely active systemic lupus erythematosus with adrenocorticotropic hormone: a single-site, open-label trial. Lupus. 2014;23(9):905–912.
  • Li X, Golubovsky J, Hui-Yuen J, et al. Adrenocorticotropic hormone gel in the treatment of systemic lupus erythematosus: a retrospective study of patients. F1000Res. 2015;4:1103.
  • Brown AN. Repository corticotropin injection in patients with refractory psoriatic arthritis: a case series. Open Access Rheumatol. 2016;8:97–102.
  • Baughman RP, Barney JB, O’Hare L, et al. A retrospective pilot study examining the use of Acthar gel in sarcoidosis patients. Respir Med. 2016;110:66–72.
  • Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther. 2002;96(1):23–43.
  • Manna SK, Aggarwal BB. Alpha-melanocyte-stimulating hormone inhibits the nuclear transcription factor NF-kappa B activation induced by various inflammatory agents. J Immunol. 1998;161(6):2873–2880.
  • Fernandez A. Interim results of an open-label study assessing efficacy and safety of adrenocorticotropic hormone gel for treatment of refractory cutaneous manifestations of dermatomyositis [abstract]. Arthritis Rheumatol. 2018;70:2528-2529(suppl 10).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.