Therapeutic vaccination with TNF-Kinoid in TNF antagonist-resistant rheumatoid arthritis: a phase II randomized, controlled clinical trial

Patrick Durez, Pierre Vandepapeliere, Pedro Miranda, Antoaneta Toncheva, Alberto Berman, Tatjana Kehler, Eugenia Mociran, Bruno Fautrel, Xavier Mariette, Olivier Dhellin, Bernard Fanget, Stephane Ouary, Géraldine Grouard-Vogel, Marie-Christophe Boissier, Patrick Durez, Pierre Vandepapeliere, Pedro Miranda, Antoaneta Toncheva, Alberto Berman, Tatjana Kehler, Eugenia Mociran, Bruno Fautrel, Xavier Mariette, Olivier Dhellin, Bernard Fanget, Stephane Ouary, Géraldine Grouard-Vogel, Marie-Christophe Boissier

Abstract

Objectives: Active immunization, or vaccination, with tumor necrosis factor (TNF)-Kinoid (TNF-K) is a novel approach to induce polyclonal anti-TNF antibodies in immune-mediated inflammatory diseases. This study was performed to transfer the proof of concept obtained in mice model of rheumatoid arthritis (RA) into human. We designed a pilot study to demonstrate the feasibility of therapeutic vaccination in RA.

Methods: This was a phase IIa, placebo-controlled, multicenter study in adults with RA who previously experienced secondary failure of TNF antagonists. Patients were immunized intramuscularly with 2 or 3 doses of placebo (n = 10) or 90 (n = 6), 180 (n = 12), or 360 µg TNF-K (n = 12). The primary objective was to identify the best dose and schedule based on anti-TNF antibody titers. Clinical symptoms and safety were assessed during 12 months and solicited reactions for 7 days after each injection.

Results: The highest anti-TNF antibody response was detected in patients immunized with 360 µg TNF-K and with 3 injections, although this difference was not significant with all other groups. Similar proportions of patients receiving TNF-K and placebo reported adverse events up to month 12. Serious adverse events were reported by 4 patients treated with TNF-K (13.3%) and 3 treated with placebo (30.0%), all unrelated to treatment. At month 12, DAS28-CRP, tender and swollen joint counts, and HAQ scores decreased significantly more in patients who exhibited anti-TNF antibody response than in patients who did not.

Conclusions: TNF-K therapeutic vaccination induced dose- and schedule-dependent anti-TNF antibodies in RA patients and was well tolerated. Patients who developed anti-TNF antibodies showed a trend toward clinical improvement. Although the most aggressive dose and schedule, i.e. 360 mg dose administered 3 times, did show a strong trend of higher antibody response, further studies are warranted to examine even higher and more frequent doses in order to establish the best conditions for clinical improvement.

Trial registration: ClinicalTrials.gov NCT01040715.

Conflict of interest statement

Competing Interests: PV OD BF SO GGV are employees of Neovacs SA and own stock or stock options in Neovacs SA. Data collection and management was subcontracted to a CRO, Inferential (35 rue Godot de Mauroy, 75009 Paris, France). Inferential had a role in the data analysis of the manuscript. Inferential had no role in the study design, decision to publish, or preparation of the manuscript. All this work was GCP compliant. This does not alter the authors’ adherence to all the PLOS ONE policies on sharing data.

Figures

Figure 1. Study design.
Figure 1. Study design.
In the first stage, 8 patients were randomized 3∶1 to receive 90 µg TNF-K or placebo, in the second, 16 patients were randomized 3∶1 to receive 180 µg TNF-K or placebo, and in the third, 17 patients were randomized 3∶1 to receive 360 µg TNF-K or placebo. In each stage, patients were also randomized 1∶1 to receive 2 doses (day 0, 28) or 3 doses (day 0, 7, 28) (arrows). For stages 1 and 2, after 3 patients had been enrolled and no safety issues had been reported for at least 7 days, enrolment in the subsequent stage started in parallel. One patient randomized to receive 3 doses of 360 µg TNF-K withdrew consent prior to treatment. The principal analysis portion of the study continued up to day 84, and the follow-up portion continued up to month 12.
Figure 2. Humoral immune response to TNF.
Figure 2. Humoral immune response to TNF.
Patients were treated with 2 doses (days 0 and 28) or 3 doses (days 0, 7, and 28) of placebo or 90, 180, or 360 µg TNF-K. Anti-TNF antibody titers were determined by enzyme-linked immunosorbent assay. (A) GMTs. (B) Percent of patients in each treatment group with detectable anti-TNF antibodies (titer ≥200) up to month 3 (at study day 38, 56, or 84), at month 6, at month 12, or at any time up to month 12 (i.e., antibody responders).
Figure 3. T cell response to TNF.
Figure 3. T cell response to TNF.
Peripheral blood mononuclear cells were collected on days 0 and 56 and treated in vitro with medium (control) or with 10 µg/mL TNF-K, TNF, or KLH. Lymphoproliferation was assessed after 72 h by 3H-thymidine incorporation. Shown is the stimulation index (fold-increase in lymphoproliferation vs. control) for cells from patients treated with placebo (n = 6) or TNF-K (n = 10).
Figure 4. Difference from baseline for clinical…
Figure 4. Difference from baseline for clinical assessments in anti-TNF antibody responders and non-responders.
Post-hoc analyses: Mean changes in clinical assessments from baseline are shown at months 3, 6, and 12 for antibody responders (detectable anti-TNF antibodies at any time; gray bars) and for non-responders (white bars). (A) DAS28-CRP. (B) CRP. (C) Tender joints count. (D) Swollen joints count. (E) Patient’s global activity score. (F) Physician’s global activity score. (G) Patient assessment of pain. (H) Change in HAQ disability/function score. Error bars indicate standard error of the mean. P-values were determined by Wilcoxon rank-sum test. NS, not significant (p≥0.05). In responders, n = 19 at month 3, 16 at months 6 and 12 except at month 3 for Physician GAS n = 18 and for CRP n = 20. In non-responders, n = 17 at month 3 and 15 at months 6 and 12 except at month 6 n = 14 for CRP and DAS 28 score.

References

    1. Scott DL, Wolfe F, Huizinga TWJ (2010) Rheumatoid arthritis. Lancet 376:1094–1108 10.1016/S0140-6736(10)60826-4
    1. McInnes IB, Schett G (2011) The pathogenesis of rheumatoid arthritis. N Engl J Med 365:2205–2219 10.7748/phc2011.11.21.9.29.c8797
    1. Breedveld FC, Combe B (2011) Understanding emerging treatment paradigms in rheumatoid arthritis. Arthritis Res Ther 13 Suppl 1: S3 10.1186/1478-6354-13-S1-S3
    1. Sfikakis PP (2010) The first decade of biologic TNF antagonists in clinical practice: lessons learned, unresolved issues and future directions. Curr Dir Autoimmun 11:180–210 10.1159/000289205
    1. Hetland ML, Christensen IJ, Tarp U, Dreyer L, Hansen A, et al. (2010) Direct comparison of treatment responses, remission rates, and drug adherence in patients with rheumatoid arthritis treated with adalimumab, etanercept, or infliximab: results from eight years of surveillance of clinical practice in the nationwide Danish. Arthritis Rheum 62:22–32 10.1002/art.27227
    1. Russell AS (2008) Quality-of-life assessment in rheumatoid arthritis. Pharmacoeconomics 26:831–846.
    1. Rubbert-Roth A, Finckh A (2009) Treatment options in patients with rheumatoid arthritis failing initial TNF inhibitor therapy: a critical review. Arthritis Res Ther 11 Suppl 1: S1 10.1186/ar2666
    1. Van Schouwenburg PA, Rispens T, Wolbink GJ (2013) Immunogenicity of anti-TNF biologic therapies for rheumatoid arthritis. Nat Rev Rheumatol 9:164–172 10.1038/nrrheum.2013.4
    1. Mulleman D, Ducourau E, Paintaud G, Ternant D, Watier H, et al. (2012) Should anti-TNF-α drug levels and/or anti-drug antibodies be assayed in patients treated for rheumatoid arthritis? Joint Bone Spine 79:109–112 10.1016/j.jbspin.2011.11.004
    1. Semerano L, Assier E, Delavallée L, Boissier M-C (2011) Kinoid of human tumor necrosis factor-alpha for rheumatoid arthritis. Expert Opin Biol Ther 11:545–550 10.1517/14712598.2011.566856
    1. Biton J, Semerano L, Delavallée L, Lemeiter D, Laborie M, et al. (2011) Interplay between TNF and regulatory T cells in a TNF-driven murine model of arthritis. J Immunol 186:3899–3910 10.4049/jimmunol.1003372
    1. Harris JR, Markl J (1999) Keyhole limpet hemocyanin (KLH): a biomedical review. Micron 30:597–623.
    1. Le Buanec H, Delavallée L, Bessis N, Paturance S, Bizzini B, et al. (2006) TNFalpha kinoid vaccination-induced neutralizing antibodies to TNFalpha protect mice from autologous TNFalpha-driven chronic and acute inflammation. Proc Natl Acad Sci U S A 103:19442–19447 10.1073/pnas.0604827103
    1. Hayward MD, Jones BK, Saparov A, Hain HS, Trillat A-C, et al. (2007) An extensive phenotypic characterization of the hTNFalpha transgenic mice. BMC Physiol 7:13 10.1186/1472-6793-7-13
    1. Delavallée L, Le Buanec H, Bessis N, Assier E, Denys A, et al. (2008) Early and long-lasting protection from arthritis in tumour necrosis factor alpha (TNFalpha) transgenic mice vaccinated against TNFalpha. Ann Rheum Dis 67:1332–1338 10.1136/ard.2007.079137
    1. Delavallée L, Semerano L, Assier E, Vogel G, Vuagniaux G, et al. (2009) Active immunization to tumor necrosis factor-alpha is effective in treating chronic established inflammatory disease: a long-term study in a transgenic model of arthritis. Arthritis Res Ther 11:R195 10.1186/ar2897
    1. Vandepapeliere P, Malan F, Rogler G, van der Bijl A, Kruger FC, et al. (2011) Safety, Immunogenicity and Clinical Phase I-II Results of TNFα-Kinoid Immunotherapeutic in Crohn’s Disease Patients. Gastroenterology 140:S–123.
    1. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, et al. (1988) The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 31:315–324.
    1. Flick DA, Gifford GE (1984) Comparison of in vitro cell cytotoxic assays for tumor necrosis factor. J Immunol Methods 68:167–175.
    1. Aletaha D, Nell VPK, Stamm T, Uffmann M, Pflugbeil S, et al. (2005) Acute phase reactants add little to composite disease activity indices for rheumatoid arthritis: validation of a clinical activity score. Arthritis Res Ther 7:R796–806 10.1186/ar1740
    1. Van Gestel AM, Haagsma CJ, van Riel PL (1998) Validation of rheumatoid arthritis improvement criteria that include simplified joint counts. Arthritis Rheum 41:1845–1850 10.1002/1529-0131(199810)41:10<1845::AID-ART17>;2-K
    1. Gaujoux-Viala C, Mouterde G, Baillet A, Claudepierre P, Fautrel B, et al. (2012) Evaluating disease activity in rheumatoid arthritis: which composite index is best? A systematic literature analysis of studies comparing the psychometric properties of the DAS, DAS28, SDAI and CDAI. Joint Bone Spine 79:149–155 10.1016/j.jbspin.2011.04.008
    1. Lauwerys BR, Hachulla E, Spertini F, Lazaro E, Jorgensen C, et al. (2013) Down-regulation of interferon signature in systemic lupus erythematosus patients by active immunization with interferon α-kinoid. Arthritis Rheum 65:447–456 10.1002/art.37785
    1. Spohn G, Guler R, Johansen P, Keller I, Jacobs M, et al. (2007) A virus-like particle-based vaccine selectively targeting soluble TNF-alpha protects from arthritis without inducing reactivation of latent tuberculosis. J Immunol 178:7450–7457.
    1. Zuany-Amorim C, Manlius C, Dalum I, Jensen MR, Gautam A, et al. (2004) Induction of TNF-alpha autoantibody production by AutoVac TNF106: a novel therapeutic approach for the treatment of allergic diseases. Int Arch Allergy Immunol 133:154–163 10.1159/000076441
    1. Waterston AM, Salway F, Andreakos E, Butler DM, Feldmann M, et al. (2004) TNF autovaccination induces self anti-TNF antibodies and inhibits metastasis in a murine melanoma model. Br J Cancer 90:1279–1284 10.1038/sj.bjc.6601670
    1. Schneerson R, Barrera O, Sutton A, Robbins JB (1980) Preparation, characterization, and immunogenicity of Haemophilus influenzae type b polysaccharide-protein conjugates. J Exp Med 152:361–376.
    1. Saidenberg-Kermanac’h N, Semerano L, Naccache JM, Brauner M, Falgarone G, et al. (2012) Screening for latent tuberculosis in anti-TNF-α candidate patients in a high tuberculosis incidence setting. Int J Tuberc Lung Dis 16:1307–1314 10.5588/ijtld.12.0111
    1. Semerano L, Biton J, Delavallée L, Duvallet E, Assier E, et al. (2013) Protection from articular damage by passive or active anti-tumour necrosis factor (TNF)-α immunotherapy in human TNF-α transgenic mice depends on anti-TNF-α antibody levels. Clin Exp Immunol 172:54–62 10.1111/cei.12040
    1. Chatzidionysiou K, Askling J, Eriksson J, Kristensen LE, van Vollenhoven R, et al.. (2013) Effectiveness of TNF inhibitor switch in RA: results from the national Swedish register. Ann Rheum Dis. 2014 Jan 15. doi:[]10.1136/annrheumdis-2013-204714.Epubaheadofprint.
    1. Spinelli FR, Valesini G (2013) Immunogenicity of anti-tumour necrosis factor drugs in rheumatic diseases. Clin Exp Rheumatol 31:954–63.

Source: PubMed

3
Abonnieren