Autoantibody-Targeted Treatments for Acute Exacerbations of Idiopathic Pulmonary Fibrosis

Michael Donahoe, Vincent G Valentine, Nydia Chien, Kevin F Gibson, Jay S Raval, Melissa Saul, Jianmin Xue, Yingze Zhang, Steven R Duncan, Michael Donahoe, Vincent G Valentine, Nydia Chien, Kevin F Gibson, Jay S Raval, Melissa Saul, Jianmin Xue, Yingze Zhang, Steven R Duncan

Abstract

Background: Severe acute exacerbations (AE) of idiopathic pulmonary fibrosis (IPF) are medically untreatable and often fatal within days. Recent evidence suggests autoantibodies may be involved in IPF progression. Autoantibody-mediated lung diseases are typically refractory to glucocorticoids and nonspecific medications, but frequently respond to focused autoantibody reduction treatments. We conducted a pilot trial to test the hypothesis that autoantibody-targeted therapies may also benefit AE-IPF patients.

Methods: Eleven (11) critically-ill AE-IPF patients with no evidence of conventional autoimmune diseases were treated with therapeutic plasma exchanges (TPE) and rituximab, supplemented in later cases with intravenous immunoglobulin (IVIG). Plasma anti-epithelial (HEp-2) autoantibodies and matrix metalloproteinase-7 (MMP7) were evaluated by indirect immunofluorescence and ELISA, respectively. Outcomes among the trial subjects were compared to those of 20 historical control AE-IPF patients treated with conventional glucocorticoid therapy prior to this experimental trial.

Results: Nine (9) trial subjects (82%) had improvements of pulmonary gas exchange after treatment, compared to one (5%) historical control. Two of the three trial subjects who relapsed after only five TPE responded again with additional TPE. The three latest subjects who responded to an augmented regimen of nine TPE plus rituximab plus IVIG have had sustained responses without relapses after 96-to-237 days. Anti-HEp-2 autoantibodies were present in trial subjects prior to therapy, and were reduced by TPE among those who responded to treatment. Conversely, plasma MMP7 levels were not systematically affected by therapy nor correlated with clinical responses. One-year survival of trial subjects was 46+15% vs. 0% among historical controls. No serious adverse events were attributable to the experimental medications.

Conclusion: This pilot trial indicates specific treatments that reduce autoantibodies might benefit some severely-ill AE-IPF patients. These findings have potential implications regarding mechanisms of IPF progression, and justify considerations for incremental trials of autoantibody-targeted therapies in AE-IPF patients.

Trial registration: ClinicalTrials.gov NCT01266317.

Conflict of interest statement

Competing Interests: SRD received <$5,000 honorarium from Genentech. With respect to the one-time honorarium given to SRD by Genentech for visiting their facility and giving a lecture, which is reported here, this does not alter the authors' adherence to all the PLOS One policies on sharing data and materials. The other authors have declared that no competing interests exist.

Figures

Fig 1. Flowchart of subject recruitments for…
Fig 1. Flowchart of subject recruitments for these experimental trials.
Original Regimen denotes the first series of subjects who were treated with the initial, relatively more conservative regimen (#1–7). Augmented Regimen denotes the most recent four subjects (#8–11) who received a more aggressive therapeutic course, based on interval results in the first cohort (see text for details). TPE = therapeutic plasma exchange; IVIG = intravenous immunoglobulin; * denotes oral consent of patients, under auspices of innovative clinical practice, that were given by these patients after being fully informed of potential risks and yet-unproven efficiencies of the novel treatments.
Fig 2. Clinical Responses to Experimental Therapy.
Fig 2. Clinical Responses to Experimental Therapy.
A.) Decreases in Supplemental Oxygen Requirements. Oxygen requirements decreased in only one of the historical control subjects during their hospitalizations, whereas pulmonary gas exchange improved with experimental treatments among most of the trial cohort (see also Table 2). B.) Changes in Abilities to Ambulate. Trial subjects who responded to experimental therapy reported improved exercise tolerance, but maximal walk distances were added as a formal outcome assessment in latest subjects, identified here by subject number (see Tables 1 and 2). With the exception of Subject #10 who showed no response to the experimental treatment, the walk distances of these later patients increased substantially. (Note: The post-treatment >2 mile distance of Subject #9 was limited by boredom rather than exercise capacity).
Fig 3. Radiographic Changes with Experimental Treatment.
Fig 3. Radiographic Changes with Experimental Treatment.
In addition to better lung function and gas exchange, experimental treatments frequently improved the chest radiographs (CXR) of these subjects. a) Pretreatment CXR of Subject #5 during relapse shows diffuse infiltrates; b.) Radiographic improvement (and extubation) of this subject to 2nd TPE series. c.) CXR of Subject #7 immediately prior to treatment and d.) after first three TPE. These (and other) radiographic and clinical improvements in the subject population were not attributable to changes in intravascular volume status or infection.
Fig 4. Anti-HEp-2 Autoantibodies in Trial Patients.
Fig 4. Anti-HEp-2 Autoantibodies in Trial Patients.
A.) Indirect Immuno-fluorescence Assays (IFA). a.) Pretreatment anti-HEp-2 autoantibodies were present at plasma titrations of 1:80 in Subject #6. b.) Autoantibodies were diminished immediately following his TPE treatment (image here at 1:20 titration). c.) Normal plasma control specimen (1:20 titration). B.) IFA Titers. Anti-HEp-2 autoantibody titers were determined in the first eight subjects. Titers were reduced following the initial TPE series in all except Subject #1 (dotted line, solid circles), who was also the only subject among these that did not have a beneficial clinical response.
Fig 5. Matrix Metalloproteinase 7 (MMP7).
Fig 5. Matrix Metalloproteinase 7 (MMP7).
Plasma MMP7 levels did not appear to consistently decrease with TPE, nor correlate with clinical responses. In particular, three subjects (#3,4,7, open circles, dotted lines) had increases or no changes of MMP7 levels from pretreatment values despite having prolonged clinical remissions.
Fig 6. Survival Comparisons.
Fig 6. Survival Comparisons.
A.) Survival in the aggregate trial subject population (n = 11) was greater than among historical control AE-IPF patients (n = 20). Cross hatches and numbers in parentheses denote censored observations. Lung transplantation censoring is denoted with “T”. B.) Clinical responses may be more durable and survival may be further enhanced among the later trial subjects (n = 4) treated with a more aggressive regimen of autoantibody-targeted modalities (9 initial TPE + rituximab + IVIG).

References

    1. Travis WD, Costabel U, Hansell DM, King TE, Lynch DA, Nicholson AG, et al. ATS/ERS Committee on Idiopathic Interstitial Pneumonias. An official American Thoracic Society/ European Respiratory Society statement: Update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am J Resp Crit Care Med 2013; 188:733–748. 10.1164/rccm.201308-1483ST
    1. Kim DS. Acute exacerbations of idiopathic pulmonary fibrosis. Clin Chest Med 2012; 33:59–68. 10.1016/j.ccm.2012.01.001
    1. Campbell DA, Poulter LW, Janossy G, du Bois RM. Immunohistological analysis of lung tissue from patients with cryptogenic fibrosing alveolitis suggesting local expression of immune hypersensitivity. Thorax 1985; 40:405–11.
    1. Marchal-Somme J, Uzunhan Y, Marchand-Adam S, Valeyre D, Soumelis V, Crestani B, et al. Cutting edge: non-proliferating mature immune cells form a novel type of organizing lymphoid structure in idiopathic pulmonary fibrosis. J Immunol 2006; 176:5735–5739.
    1. Nuovo GJ, Hagood JS, Magro CM, Chin N, Kapil R, Davis L, et al. The distribution of immunomodulatory cells in the lungs of patients with idiopathic pulmonary fibrosis. Mod Pathol 2012; 25:416–33. 10.1038/modpathol.2011.166
    1. Dall Aglio PP, Pesci A, Bertorelli G, Brianti E, Scarpa S. Study of immune complexes in broncholaveolar lavage fluids. Respiration 1988; 54:36–41.
    1. Dobashi N, Fujita J, Murota M, Ohtsuki Y, Yamadori I, Yoshinouchi T, et al. Elevation of anti-cytokeratin 18 antibody and circulating cytokeratin 18: anti-cytokeratin 18 antibody immune complexes in sera of patients with idiopathic pulmonary fibrosis. Lung 2000; 178:171–179.
    1. Feghali-Bostwick CA, Tsai CG, Valentine VG, Kantrow S, Stoner MW, Pilewski JM, et al. Cellular and humoral autoreactivity in idiopathic pulmonary fibrosis. J Immunol 2007; 179:2592–2599.
    1. Magro CM, Waldman WJ, Knight DA, Allen JN, Nadasdy T, Frambach GE, et al. Idiopathic pulmonary fibrosis related to endothelial injury and antiendothelial cell antibodies. Hum Immunol 2006; 67:284–297.
    1. Ogushi F, Tani K, Endo T, Tada H, Kawano T, Asano T, et al. Autoantibodies to IL-1 in sera from rapidly progressive idiopathic pulmonary fibrosis. J Med Invest 2001; 48:181–189.
    1. Kurosu K, Takiguchi Y, Okada O, Yumoto N, Sakao S, Tada Y, et al. Identification of annexin 1 as a novel autoantigen in acute exacerbation of idiopathic pulmonary fibrosis. J Immunol 2008; 181:756–767.
    1. Kahloon RA, Xue J, Bhargava A, Csizmadia E, Otterbein L, Kass DJ, et al. Idiopathic pulmonary fibrosis patients with antibodies to heat shock protein 70 have poor prognoses. Am J Resp Crit Care Med 2013; 187:768–775. 10.1164/rccm.201203-0506OC
    1. Taillé C, Grootenboer-Mignot S, Boursier C, Michel L, Debray MP, Fagart J, et al. Identification of periplakin as a new target for autoreactivity in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2011; 183:759–766. 10.1164/rccm.201001-0076OC
    1. Wallace WHH, Howie SM. Upregulation of tenascin and TGF-β production in a type II alveolar epithelial cell line by antibody against a pulmonary auto-antigen. J Pathol 2001; 195:251–256
    1. Yang Y, Fujita J, Bandoh S, Ohtsuki Y, Yamadori I, Yoshinouchi T, et al. Detection of antivimentin antibody in sera of patients with idiopathic pulmonary fibrosis and non-specific interstitial pneumonia. Clin Exp Immunol 2002; 128:169–174.
    1. Shum AK, Alimohammadi M, Tan CL, Cheng MH, Metzger TC, Law CS, et al. BPIFB1 is a lung-specific autoantigen associated with interstitial lung disease. Sci Transl Med 2013; 5:1–10.
    1. Vittal R, Mickler EA, Fisher AJ, Zhang C, Rothhaar K, Gu H, et al. Type V collagen induced tolerance suppresses collagen deposition, TGF-β and associated transcripts in pulmonary fibrosis. PLoS One 2013; 8:e76451 10.1371/journal.pone.0076451
    1. Vuga LJ, Tedrow JR, Pandit KV, Tan J, Kass DJ, Xue J, et al. C-X-C motif chemokine 13 (CXCL13) is a prognostic biomarker of patients with idiopathic pulmonary fibrosis. Am J Resp Crit Care Med 2014; 189:966–974. 10.1164/rccm.201309-1592OC
    1. Xue J, Kass DJ, Bon JM, Vuga L, Tan J, Csizmadia E, et al. Plasma B-lymphocyte stimulator (BLyS) and B-cell differentiation in idiopathic pulmonary fibrosis. J Immunol 2013; 191:2089–2095. 10.4049/jimmunol.1203476
    1. DePianto DJ, Chandriani S, Abbas AR, Jia G, N’Diaye EN, Caplazi P, et al. Heterogeneous gene expression signatures correspond to distinct lung pathologies and biomarkers of disease severity in idiopathic pulmonary fibrosis. Thorax 2015; 70:48–56. 10.1136/thoraxjnl-2013-204596
    1. Francois A, Gombault A, Villeret B, Alsaleh G, Fanny M, Gasse P, et al. B cell activating factor is central to bleomycin- and IL-17-mediated experimental pulmonary fibrosis. J Autoimmun 2015; 56:1–11. 10.1016/j.jaut.2014.08.003
    1. Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory disease. Nature Rev 2006; 6:205–211.
    1. Browning JL. B cells move to center stage: novel opportunities for autoimmune disease treatment. Nat Rev 2006. 5:564–575.
    1. Corsiero E, Bombardieri M, Manzo A, Bugatti S, Uguccioni M, Pitzalis C. Role of lymphoid chemokines in the development of functional ectopic lymphoid structures in rheumatic autoimmune diseases. Immunol Lett 2012; 145:62–67. 10.1016/j.imlet.2012.04.013
    1. Rioja I, Hughes FJ, Sharp CH, Warnock LC, Montgomery DS, Akil M, et al. Potential novel biomarkers of disease activity in rheumatoid arthritis patients: CXCL13, CCL23, transforming growth factor alpha, tumor necrosis factor receptor superfamily member 9, and macrophage colony-stimulating factor. Arthritis Rheum 2008; 58:2257–67. 10.1002/art.23667
    1. Brettschneider J, Czerwoniak A, Senel M, Fang L, Kassubek J, Pinkhardt E, et al. The chemokine CXCL13 is a prognostic marker in clinically isolated syndrome (CIS). PLoS ONE 2010; 5:e11986 10.1371/journal.pone.0011986
    1. Lee HT, Shiao YM, Wu TH, Chen WS, Hsu YH, Tsai SF, et al. Serum BLC/CXCL13 concentrations and renal expression of CXCL13/CXCR5 in patients with systemic lupus erythematosus and lupus nephritis. J Rheum 2010; 37:45–52. 10.3899/jrheum.090450
    1. Mayadas TN, Tsokos GC, Tsuboi N. Mechanisms of immune complex-mediated neutrophil recruitment and tissue injury. Circulation 2009; 120:2012–2024. 10.1161/CIRCULATIONAHA.108.771170
    1. Jacobi AM, Reiter K, Mackay M, Aranow C, Hiepe F, Radbruch A, et al. Activated memory B cell subsets correlate with disease activity in systemic lupus erythematosus: delineation by expression of CD27, IgD, and CD95. Arth Rheum 2008; 58: 1762–1773.
    1. Souto-Carneiro MM, Mahadevan V, Takada K, Fritsch-Stork R, Nanki T, Brown M, et al. Alterations in peripheral blood memory B cells in patients with active rheumatoid arthritis are dependent on the action of tumour necrosis factor. Arthritis Res Ther 2009; 11: R84 10.1186/ar2718
    1. Cancro MP, D’Cruz DP, Khamashta MA. The role of B lymphocyte stimulator (BLyS) in systemic lupus erythematosus. J Clin Invest 2009; 119:1066–1073. 10.1172/JCI38010
    1. Bossen C, Schneider P. BAFF, APRIL and their receptors: structure, function and signaling. Semin. Immunol 2006; 18:263–275.
    1. Dillon SR, Harder B, Lewis KB, Moore MD, Liu H, Bukowski TR, et al. B-lymphocyte stimulator/a proliferation-inducing ligand heterotrimers are elevated in the sera of patients with autoimmune disease and are neutralized by atacicept and B-cell maturation antigen-immunoglobulin. Arthrtitis Res Ther 2010; 12: R48 10.1186/ar2959
    1. Solomon DH, Kavanaugh AJ, Schur PH. Evidence-based guidelines for the use of immunologic tests: antinuclear antibody testing. Arthritis Rheum 2002; 47:434–44.
    1. Erickson SB, Kurtz SB, Donadio JV, Holley KE, Wilson CB, Pineda AA. Use of combined plasmapharesis and immunosuppression in the treatment of Goodpasture’s syndrome. Mayo Clin Proc 1979; 54:714–720.
    1. Sem M, Molberg O, Lund MB, Gran JT. Rituximab treatment of the anti-synthetase syndrome: a retrospective case series. Rheumatology (Oxford) 2009; 48:968–971. 10.1093/rheumatology/kep157
    1. Borie R, Debray MP, Laine C, Aubier M, Crestani B. Rituximab therapy in autoimmune pulmonary alveolar proteinosis. Eur Respir J 2009; 33,1503–1506. 10.1183/09031936.00160908
    1. Keir GJ, Maher TM, Hansell DM, Denton CP, Ong VH, Singh S, et al. Severe interstitial lung disease in connective tissue disease: rituximab as rescue therapy. Eur Resp J 2012; 40:641–648. 10.1183/09031936.00163911
    1. Stone JH, Merkel PA, Spiera R, Seo P, Langford CA, Hoffman GS, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med 2010; 363:221–232. 10.1056/NEJMoa0909905
    1. Orlin JB, Berkman EM. Partial plasma exchange using albumin replacement: removal and recovery of normal plasma constituents. Blood 1980; 56:1055–1060.
    1. Reverberi R, Reverberi L. Removal kinetics of therapeutic apheresis. Blood Transfus 2007; 5:164–174. 10.2450/2007.0032-07
    1. Richards TJ, Kaminski N, Baribaud F, Flavin S, Brodmerkel C, Horowitz D, et al. Peripheral blood proteins predict mortality in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 185:67–76. 10.1164/rccm.201101-0058OC
    1. Franks AL, Slansky JE. Multiple associations between a broad spectrum of autoimmune diseases, chronic inflammatory diseases and cancer. Anticancer Res 2012;32:1119–1136.
    1. Xue J, Gochuico BR, Alawad AS, Feghali-Bostwick CA, Noth I, Nathan SD, et al. The HLA Class II allele DRB1*1501 is over-represented in patients with idiopathic pulmonary fibrosis. PLoS One 2011; 6:e14715 10.1371/journal.pone.0014715
    1. Gilani SR, Vuga LJ, Lindell KO, Gibson KF, Xue J, Kaminski N, et al. CD28 down-regulation on circulating CD4 T-cells is associated with poor prognoses of patients with idiopathic pulmonary fibrosis. PLoS One. 2010; 5:e8959 10.1371/journal.pone.0008959
    1. Herazo-Maya JD, Noth I, Duncan SR, Kim S, Ma SF, Tseng GC, et al. Peripheral blood mononuclear cell gene expression profiles may predict poor outcome in idiopathic pulmonary fibrosis. Sci Translat 2013; 5:205ra136.
    1. Lee SJ, Kavanaugh A. Autoimmunity, vasculitis and autoantibodies. J Allerg Clin Immunol 2006; 117:S445–450.
    1. Pordeus V, Szyper-Kravitz M, Levy RA, Vaz NM, Shoenfeld Y. Infections and autoimmunity: a panorama. Clinic Rev Allerg Immunol 2008; 34:283–299.
    1. Lindstrom TM, Robinson WH. Rheumatoid arthritis: a role for immunosenescence? J Am Geriatr Soc. 2010: 1565–1575. 10.1111/j.1532-5415.2010.02965.x
    1. Goronzy JJ, Li G, Yu M, Weyand CM. Signaling pathways in aged T cells—a reflection of T cell differentiation, cell senescence and host environment. Semin Immunol. 2012; 24:365–372. 10.1016/j.smim.2012.04.003
    1. Nydegger UE, Sturzenegger M. Treatment of autoimmune disease: synergy between plasma exchange and intravenous immunoglobulin. Therap Apher 2001;5:186–192.
    1. Gelfand EW. Intravenous immune globulin in autoimmune and inflammatory diseases. N Eng J Med 2012; 167:2015–2025.

Source: PubMed

3
Suscribir