553
Views
17
CrossRef citations to date
0
Altmetric
Review

Potential immunotherapies for sarcoidosis

&
Pages 399-407 | Received 09 Oct 2017, Accepted 10 Jan 2018, Published online: 17 Jan 2018

References

  • Cozier YC. Assessing the worldwide epidemiology of sarcoidosis: challenges and future directions. Eur Respir J. 2016;48:1545–1548.
  • Judson MA. The clinical features of sarcoidosis: a comprehensive review. Clin Rev Allergy Immunol. 2015;49:63–78.
  • Drent M, Lower EE, De Vries J. Sarcoidosis-associated fatigue. Eur Respir J. 2012;40:255–263.
  • Goracci A, Fagiolini A, Martinucci M, et al. Quality of life, anxiety and depression in sarcoidosis. Gen Hosp Psychiatry. 2008;30:441–445.
  • Iannuzzi MC, Fontana JR. Sarcoidosis clinical presentation, immunopathogenesis, and therapeutics. Jama. 2011;305:391–399.
  • Zissel G, Muller-Quernheim J. Cellular players in the immunopathogenesis of sarcoidosis. Clin Chest Med. 2015;36:549–560.
  • Timmermans WM, Van Laar JA, Van Hagen PM, et al. Immunopathogenesis of granulomas in chronic autoinflammatory diseases. Clin Transl Immunol. 2016;5:e118.
  • Noor A, Knox KS. Immunopathogenesis of sarcoidosis. Clin Dermatol. 2007;25:250–258.
  • Ramstein J, Broos CE, Simpson LJ, et al. IFN-gamma-producing T-helper 17.1 cells are increased in sarcoidosis and are more prevalent than T-helper type 1 cells. Am J Respir Crit Care Med. 2016;193:1281–1291.
  • Mortaz E, Rezayat F, Amani D, et al. The roles of T helper 1, T helper 17 and regulatory T cells in the pathogenesis of sarcoidosis. Iran J Allergy Asthma Immunol. 2016;15:334–339.
  • Miyara M, Amoura Z, Parizot C, et al. The immune paradox of sarcoidosis and regulatory T cells. J Exp Med. 2006;203:359–370.
  • Taflin C, Miyara M, Nochy D, et al. FoxP3+ regulatory T cells suppress early stages of granuloma formation but have little impact on sarcoidosis lesions. Am J Pathol. 2009;174:497–508.
  • Moller M, Aries SP, Dromann D, et al. Intracellular cytokine repertoire in different T cell subsets from patients with sarcoidosis. Thorax. 2001;56:487–493.
  • Patterson KC, Hogarth K, Husain AN, et al. The clinical and immunologic features of pulmonary fibrosis in sarcoidosis. Transl Res. 2012;160:321–331.
  • Cox CE, Donohue JF, Brown CD, et al. Health-related quality of life of persons with sarcoidosis. Chest. 2004;125:997–1004.
  • Chen ES, Song Z, Willett MH, et al. Serum amyloid A regulates granulomatous inflammation in sarcoidosis through Toll-like receptor-2. Am J Respir Crit Care Med. 2010;181:360–373.
  • Herrtwich L, Nanda I, Evangelou K, et al. DNA damage signaling instructs polyploid macrophage fate in granulomas. Cell. 2016;167:1264–80 e18.
  • Montesinos MC, Desai A, Delano D, et al. Adenosine A2A or A3 receptors are required for inhibition of inflammation by methotrexate and its analog MX-68. Arthritis Rheum. 2003;48:240–247.
  • Society AT. Statement on sarcoidosis. Am J Respir Crit Care Med. 1999;160:736–755.
  • Ferrante CJ, Pinhal-Enfield G, Elson G, et al. The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL-4Ralpha) signaling. Inflammation. 2013;36:921–931.
  • Hasko G, Cronstein B. Regulation of inflammation by adenosine. Front Immunol. 2013;4:85.
  • Isshiki T, Yamaguchi T, Yamada Y, et al. Usefulness of low-dose methotrexate monotherapy for treating sarcoidosis. Internal Medicine. 2013;52:2727–2732.
  • Lower EE, Baughman RP. Prolonged use of methotrexate for sarcoidosis. Arch Intern Med. 1995;155:846–851.
  • Chan ESL, Cronstein BN. Molecular action of methotrexate in inflammatory diseases. Arthritis Res. 2002;4:266–273.
  • Maltzman JS, Koretzky GA. Azathioprine: old drug, new actions. J Clin Invest. 2003;111:1122–1124.
  • Poppe D, Tiede I, Fritz G, et al. Azathioprine suppresses ezrin-radixin-moesin-dependent T cell-APC conjugation through inhibition of Vav guanosine exchange activity on Rac proteins. J Immunol. 2005;176:640–651.
  • Muller-Quernheim J, Kienast K, Held M, et al. Treatment of chronic sarcoidosis with an azathioprine/prednisolone regimen. Eur Respir J. 1999;14:1117–1122.
  • Vorselaars ADM, Wuyts WA, Vorselaars VMM, et al. Methotrexate vs azathioprine in second-line therapy of sarcoidosis. Chest. 2013;144:805–812.
  • Antoniu SA. Targeting the TNF-alpha pathway in sarcoidosis. Expert Opin Ther Targets. 2010;14:21–29.
  • Kreth S, Ledderose C, Luchting B, et al. Immunomodulatory properties of pentoxifylline are mediated via adenosine-dependent pathways. Shock. 2010;34:10–16.
  • Marques LJ, Zheng L, Poulaski N, et al. Pentoxifylline inhibits TNF-alpha production from human alveolar macrophages. Am J Respir Crit Care Med. 1999;159:508–511.
  • Tong Z, Dai H, Chen B, et al. Inhibition of cytokine release from alveolar macrophages in pulmonary sarcoidosis by pentoxifylline. Chest. 2003;124:1526–1532.
  • Park MK, Fontana JR, Babaali H, et al. Steroid-sparing effects of pentoxifylline in pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Disease. 2009;26:121–131.
  • Zabel P, Entzian P, Dalhoff K, et al. Pentoxifylline in treatment of sarcoidosis. Am J Respir Crit Care Med. 1997;155:1665–1669.
  • Moreira AL, Sampaio EP, Zmuidzinas A, et al. Thalidomide exerts its inhibitory action on tumor necrosis factor alpha by enhancing mRNA degradation. J Exp Med. 1993;177:1675–1680.
  • Tavares JL, Wangoo A, Dilworth P, et al. Thalidomide reduces tumour necrosis factor-alpha production by human alveolar macrophages. Respir Med. 1997;91:31–39.
  • Droitcourt C, Rybojad M, Porcher R, et al. A randomized, investigator-masked, double-blind, placebo-controlled trial on thalidomide in severe cutaneous sarcoidosis. Chest. 2014;146:1046–1054.
  • Judson MA, Silvestri J, Hartung C, et al. The effect of thalidomide on corticosteroid-dependent pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Disease. 2006;23:51–57.
  • Fazzi P, Manni E, Cristofani R, et al. Thalidomide for improving cutaneous and pulmonary sarcoidosis in patients resistant or with contraindications to corticosteroids. Biomed Pharmacother. 2012;66:300–307.
  • Schafer P. Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem Pharmacol. 2012;83:1583–1590.
  • Baughman RP, Judson MA, Ingledue R, et al. Efficacy and safety of apremilast in chronic cutaneous sarcoidosis. Arch Dermatol. 2012;148:262–264.
  • Baughman RP, Drent M, Kavuru M, et al. Infliximab therapy in patients with chronic sarcoidosis and pulmonary involvement. Am J Respir Crit Care Med. 2006;174:795–802.
  • Baughman RP, Judson MA, Lower EE, et al. Infliximab for chronic cutaneous sarcoidosis: a subset analysis from a double-blind randomized clinical trial. Sarcoidosis Vasc Diffuse Lung Disease. 2016;32:289–295.
  • Baughman RP, Shipley R, Desai S, et al. Changes in chest roentgenogram of sarcoidosis patients during a clinical trial of infliximab therapy: comparison of different methods of evaluation. Chest. 2009;136:526–535.
  • Judson MA, Baughman RP, Costabel U, et al. Efficacy of infliximab in extrapulmonary sarcoidosis: results from a randomised trial. Eur Respir J. 2008;31:1189–1196.
  • Saleh S, Ghodsian S, Yakimova V, et al. Effectiveness of infliximab in treating selected patients with sarcoidosis. Respir Med. 2006;100:2053–2059.
  • Erckens RJ, Mostard RL, Wijnen PA, et al. Adalimumab successful in sarcoidosis patients with refractory chronic non-infectious uveitis. Graefes Arch Clin Exp Ophthalmol. 2012;250:713–720.
  • Milman N, Graudal N, Loft A, et al. Effect of the TNF-alpha inhibitor adalimumab in patients with recalcitrant sarcoidosis: a prospective observational study using FDG-PET. Clin Respir J. 2012;6:238–247.
  • Crommelin HA, van der Burg LM, Vorselaars AD, et al. Efficacy of adalimumab in sarcoidosis patients who developed intolerance to infliximab. Respir Med. 2016;115:72–77.
  • Baughman RP, Lower EE, Bradley DA, et al. Etanercept for refractory ocular sarcoidosis. Chest. 2005;128:1062–1067.
  • Utz JP, Limper AH, Kalra S, et al. Etancercept for the treatment of stage II and III progressive pulmonary sarcoidosis. Chest. 2003;124:177–185.
  • Amber KT, Bloom R, Mrowietz U, et al. TNF-alpha: a treatment target or cause of sarcoidosis? J Eur Acad Dermatol Venereol. 2015;29:2104–2111.
  • Judson MA, Baughman RP, Costabel U, et al. Safety and efficacy of ustekinumab or golimumab in patients with chronic sarcoidosis. Eur Respir J. 2014;44:1296–1307.
  • Takahashi R, Shijubo R, Shigehara K, et al. T helper 1 inhibitor TAK-603 inhibits IFN-gamma and IL-12 production with no effect on IL-18: an observation in sarcoidosis patients. Sarcoidosis Vasc Diffuse Lung Disease. 2004;21:204–211.
  • Alwan W, Nestle FO. Pathogenesis and treatment of psoriasis: exploiting pathophysiological pathways for precision medicine. Clin Exp Rheumatol. 2015;33:S2–S6.
  • Baumgart DC, Sandborn WJ. Crohn’s disease. The Lancet. 2012;380:1590–1605.
  • Reinisch W, De Villiers W, Bene L, et al. Fontolizumab in moderate to severe Crohn’s disease: a phase 2, randomized, double-blind, placebo-controlled, multiple-dose study. Inflamm Bowel Dis. 2010;16:233–242.
  • Harden JL, Johnson-Huang LM, Chamian MF, et al. Humanized anti-IFN-gamma (HuZAF) in the treatment of psoriasis. J Allergy Clin Immunol. 2015;135:553–556.
  • Sah BP, Goyal S, Iannuzzi MC. Novel pharmacotherapy of sarcoidosis. Pharmacol Ther. 2016;157:1–9.
  • Jiang Z, Zhu L. Update on the role of alternatively activated macrophages in asthma. J Asthma Allergy. 2016;9:101–107.
  • Liu YC, Zou XB, Chai YF, et al. Macrophage polarization in inflammatory diseases. Int J Biol Sci. 2014;10:520–529.
  • Mantovani A, Sica A, Sozzani S, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:677–686.
  • Jiang H, Harris MB, Rothman P. IL-4/IL-13 signaling beyond JAK/STAT. J Allergy Clin Immunol. 2000;105:1063–1070.
  • Zeyda M, Poglitsch M, Geyeregger R, et al. Disruption of the interaction of T cells with antigen-presenting cells by the active leflunomide metabolite teriflunomide: involvement of impaired integrin activation and immunologic synapse formation. Arthritis Rheum. 2005;52:2730–2739.
  • Siemasko K, Chong ASF, Jack HM, et al. Inhibition of JAK3 and STAT6 tyrosine phosphorylation by the immunosuppressive drug leflunomide leads to a block in IgG1 production. J Immunol. 1998;160:1581–1588.
  • Sahoo DH, Bandyopadhyay D, Xu M, et al. Effectiveness and safety of leflunomide for pulmonary and extrapulmonary sarcoidosis. Eur Respir J. 2011;38:1145–1150.
  • He L, Marneros AG. Doxycycline inhibits polarization of macrophages to the proangiogenic M2-type and subsequent neovascularization. J Biol Chem. 2014;289:8019–8028.
  • Bachelez H, Senet P, Cadranel J, et al. The use of tetracyclines for the treatment of sarcoidosis. Arch Dermatol. 2001;137:69–73.
  • Simpson EL, Bieber T, Guttman-Yassky E, et al. Two phase 3 trials of dupilumab versus placebo in atopic dermatitis. N Engl J Med. 2016;375:2335–2348.
  • Wenzel S, Ford L, Pearlman D, et al. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med. 2013;368:2455–2466.
  • Eishi Y, Suga M, Ishige I, et al. Quantitative analysis of mycobacterial and propionibacterial DNA in lymph nodes of Japanese and European patients with sarcoidosis. J Clin Microbiol. 2002;40:198–204.
  • Song Z, Marzilli L, Greenlee BM, et al. Mycobacterial catalase-peroxidase is a tissue antigen and target of the adaptive immune response in systemic sarcoidosis. J Exp Med. 2005;201:755–767.
  • Rotsinger JE, Celada LJ, Polosukhin VV, et al. Molecular analysis of sarcoidosis granulomas reveals antimicrobial targets. Am J Respir Cell Mol Biol. 2016;55:128–134.
  • Drake WP, Oswald-Richter K, Richmond BW, et al. Oral antimycobacterial therapy in chronic cutaneous sarcoidosis: a randomized, single-masked, placebo-controlled study. JAMA Dermatol. 2013;149:1040–1049.
  • Drake WP, Richmond BW, Oswald-Richter K, et al. Effects of broad-spectrum antimycobacterial therapy on chronic pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Disease. 2014;30:201–211.
  • Ungprasert P, Crowson CS, Matteson EL. Smoking, obesity and risk of sarcoidosis: a population-based nested case-control study. Respir Med. 2016;120:87–90.
  • Newman LS, Rose CS, Bresnitz EA, et al. A case control etiologic study of sarcoidosis: environmental and occupational risk factors. Am J Respir Crit Care Med. 2004;170:1324–1330.
  • Valeyre D, Soler P, Clerici C, et al. Smoking and pulmonary sarcoidosis: effect of cigarette smoking on prevalence, clinical manifestations, alveolitis, and evolution of the disease. Thorax. 1988;43:516–524.
  • Sugano N, Shimida K, Ito K, et al. Nicotine inhibits the prodiction of inflammatory mediators in U937 cells through modulation of nuclear factor-kB activation. Biochemal Biophys Res Commun. 1998;252:25–28.
  • Zheng YS, Wu ZS, Zhang LY, et al. Nicotine ameliorates experimental severe acute pancreatitis via enhancing immunoregulation of CD4+ CD25+ regulatory T cells. Pancreas. 2015;44:500–506.
  • Julian MW, Shao G, Schlesinger LS, et al. Nicotine treatment improves Toll-like receptor 2 and Toll-like receptor 9 responsiveness in active pulmonary sarcoidosis. Chest. 2013;143:461–470.
  • Salin-Pascual RJ, Rosas M, Jimenez-Genchi A, et al. Antidepressant effect of transdermal nicotine patches in nonsmoking patients with major depression. J Clin Psychiatry. 1996;57:387–389.
  • Warburton DM, Mancuso G. Evaluation of the information processing and mood effects of a transdermal nicotine patch. Psychopharmacology. 1998;135:305–310.
  • Le Houezec J, Halliday R, Benowitz NL, et al. A low dose of subcutaneous nicotine improves information processing in non-smokers. Psychopharmacology. 1994;114:628–634.
  • Gentry MV, Hammersley JJ, Hale CR, et al. Nicotine patches improve mood and response speed in a lexical decision task. Addict Behav. 2000;25:549–557.
  • Braun NA, Celada LJ, Herazo-Maya JD, et al. Blockade of the programmed death-1 pathway restores sarcoidosis CD4(+) T-cell proliferative capacity. Am J Respir Crit Care Med. 2014;190:560–571.
  • Celada LJ, Rotsinger JE, Young A, et al. Programmed death-1 inhibition of phosphatidylinositol 3-kinase/AKT/mechanistic target of rapamycin signaling impairs sarcoidosis CD4(+) T cell proliferation. Am J Respir Cell Mol Biol. 2017;56:74–82.
  • Broos CE, van Nimwegen M, Kleinjan A, et al. Impaired survival of regulatory T cells in pulmonary sarcoidosis. Respir Res. 2015;16:108.
  • Broos CE, Veen JC, Hoogsteden HC. Decreased cytotoxic T-Lymphocyte antigen 4 expression on regulatory T cells and Th17 cells in sarcoidosis: double trouble? Am J Respir Crit Care Med. 2015;192:763–765.
  • Lomax AJ, McGuire HM, McNeil C, et al. Immunotherapy-induced sarcoidosis in patients with melanoma treated with PD-1 checkpoint inhibitors: case series and immunophenotypic analysis. Int J Rheum Dis. 2017;20:1277–1285.
  • Reddy SB, Possick JD, Kluger HM, et al. Sarcoidosis AA-PD-1 and anti-CTLA-4 therapy for metastatic melanoma. J Immunother. 2017;40:307–311.
  • Cousin S, Italiano A. Pulmonary sarcoidosis or post-immunotherapy granulomatous reaction induced by the anti-PD-1 monoclonal antibody pembrolizumab: the termilogy is not the key point. Ann Oncol. 2016;27:1974–1975.
  • Kamphuis LS, Van Zelm MC, Lam KH, et al. Perigranuloma localization and abnormal maturation of B cells: emerging key players in sarcoidosis? Am J Respir Crit Care Med. 2013;187:406–416.
  • Lee NS, Barber L, Akula SM, et al. Disturbed homeostasis and multiple signaling defects in the peripheral blood B-cell compartment of patients with severe chronic sarcoidosis. Clin Vaccine Immunol. 2011;18:1306–1316.
  • Saussine A, Tazi A, Feuillet S, et al. Active chronic sarcoidosis is characterized by increased transitional blood B cells, increased IL-10-producing regulatory B cells and high BAFF levels. PLoS One. 2012;7:e43588.
  • Ueda-Hayakawa I, Tanimura H, Osawa M, et al. Elevated serum BAFF levels in patients with sarcoidosis: association with disease activity. Rheumatology (Oxford). 2013;52:1658–1666.
  • Sweiss NJ, Lower EE, Mirsaedi M, et al. Rituximan in the treatment of refractory pulmonary sarcoidosis. Eur Respir J. 2014;43:1525–1528.
  • Hao W, Crouser ED, Friedman A. Mathematical model of sarcoidosis. Proc Natl Acad Sci U S A. 2014;111:16065–16070.
  • Crouser ED, White P, Caceres EG, et al. A novel in vitro human granuloma model of sarcoidosis and latent tuberculosis infection. Am J Respir Cell Mol Biol. 2017;57:487–498.
  • Maier LA, Crouser ED, Martin WJ, et al. Executive summary of the NHLBI workshop report: leveraging current scientific advances to better understand sarcoidosis variability and improve outcome. Ann Am Thorac Soc. 2017 Oct 19. Epub ahead of print. DOI:10.1513/AnnalsATS.201707-563OT
  • Crouser ED, Fingerlin TE, Yang IV, et al. Application of ‘Omics’ and systems biology to sarcoidosis research. Ann Am Thorac Soc. 2017 Oct 20. Epub ahead of press. DOI:10.1513/AnnalsATS.201707-567OT
  • Abrahamyan L, Feldman BM, Tomlinson G, et al. Alternative designs for clinical trials in rare diseases. Am J Med Genet C Semin Med Genet. 2016;172:313–331.

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.