Epitope-Specific Immunotherapy Targeting CD4-Positive T Cells in Celiac Disease: Safety, Pharmacokinetics, and Effects on Intestinal Histology and Plasma Cytokines with Escalating Dose Regimens of Nexvax2 in a Randomized, Double-Blind, Placebo-Controlled Phase 1 Study

A James M Daveson, Hooi C Ee, Jane M Andrews, Timothy King, Kaela E Goldstein, John L Dzuris, James A MacDougall, Leslie J Williams, Anita Treohan, Michael P Cooreman, Robert P Anderson, A James M Daveson, Hooi C Ee, Jane M Andrews, Timothy King, Kaela E Goldstein, John L Dzuris, James A MacDougall, Leslie J Williams, Anita Treohan, Michael P Cooreman, Robert P Anderson

Abstract

Background: Nexvax2® is a novel, peptide-based, epitope-specific immunotherapy intended to be administered by regular injections at dose levels that increase the threshold for clinical reactivity to natural exposure to gluten and ultimately restore tolerance to gluten in patients with celiac disease. Celiac disease patients administered fixed intradermal doses of Nexvax2 become unresponsive to the HLA-DQ2·5-restricted gluten epitopes in Nexvax2, but gastrointestinal symptoms and cytokine release mimicking gluten exposure, that accompany the first dose, limit the maximum tolerated dose to 150μg. Our aim was to test whether stepwise dose escalation attenuated the first dose effect of Nexvax2 in celiac disease patients.

Methods: We conducted a randomized, double-blind, placebo-controlled trial at four community sites in Australia (3) and New Zealand (1) in HLA-DQ2·5 genotype positive adults with celiac disease who were on a gluten-free diet. Participants were assigned to cohort 1 if they were HLA-DQ2·5 homozygotes; other participants were assigned to cohort 2, or to cohort 3 subsequent to completion of cohort 2. Manual central randomization without blocking was used to assign treatment for each cohort. Initially, Nexvax2-treated participants in cohorts 1 and 2 received an intradermal dose of 30μg (consisting of 10μg of each constituent peptide), followed by 60μg, 90μg, 150μg, and then eight doses of 300μg over six weeks, but this was amended to include doses of 3μg and 9μg and extended over a total of seven weeks. Nexvax2-treated participants in cohort 3 received doses of 3μg, 9μg, 30μg, 60μg, 90μg, 150μg, 300μg, 450μg, 600μg, 750μg, and then eight of 900μg over nine weeks. The dose interval was 3 or 4days. Participants, care providers, data managers, sponsor personnel, and study site personnel were blinded to treatment assignment. The primary outcome was the number of adverse events and percentage of participants with adverse events during the treatment period. This completed trial is registered with ClinicalTrials.gov, number NCT02528799.

Findings: From the 73 participants who we screened from 19 August 2015 to 31 October 2016, 24 did not meet eligibility criteria, and 36 were ultimately randomized and received study drug. For cohort 1, seven participants received Nexvax2 (two with the starting dose of 30μg and then five at 3μg) and three received placebo. For cohort 2, 10 participants received Nexvax2 (four with starting dose of 30μg and then six at 3μg) and four received placebo. For cohort 3, 10 participants received Nexvax2 and two received placebo. All 36 participants were included in safety and immune analyses, and 33 participants completed treatment and follow-up; in cohort 3, 11 participants were assessed and included in pharmacokinetics and duodenal histology analyses. Whereas the maximum dose of Nexvax2 had previously been limited by adverse events and cytokine release, no such effect was observed when dosing escalated from 3μg up to 300μg in HLA-DQ2·5 homozygotes or to 900μg in HLA-DQ2.5 non-homozygotes. Adverse events with Nexvax2 treatment were less common in cohorts 1 and 2 with the starting dose of 3μg (72 for 11 participants) than with the starting dose of 30μg (91 for six participants). Adverse events during the treatment period in placebo-treated participants (46 for nine participants) were similar to those in Nexvax2-treated participants when the starting dose was 3μg in cohort 1 (16 for five participants), cohort 2 (56 for six participants), and cohort 3 (44 for 10 participants). Two participants in cohort 2 and one in cohort 3 who received Nexvax2 starting at 3μg did not report any adverse event, while the other 33 participants experienced at least one adverse event. One participant, who was in cohort 1, withdrew from the study due to adverse events, which included abdominal pain graded moderate or severe and associated with nausea after receiving the starting dose of 30μg and one 60μg dose. The most common treatment-emergent adverse events in the Nexvax2 participants were headache (52%), diarrhoea (48%), nausea (37%), abdominal pain (26%), and abdominal discomfort (19%). Administration of Nexvax2 at dose levels from 150μg to 900μg preceded by dose escalation was not associated with elevations in plasma cytokines at 4h. Nexvax2 treatment was associated with trends towards improved duodenal histology. Plasma concentrations of Nexvax2 peptides were dose-dependent.

Interpretation: We show that antigenic peptides recognized by CD4-positive T cells in an autoimmune disease can be safely administered to patients at high maintenance dose levels without immune activation if preceded by gradual dose escalation. These findings facilitate efficacy studies that test high-dose epitope-specific immunotherapy in celiac disease.

Keywords: Celiac disease; Immunotherapy; Nexvax2; Peptide; Randomized controlled trial; T cell.

Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Study schematic. *Escalation was amended for all cohorts by including 3 μg and 9 μg doses when one participant in Cohort 1 withdrew with gastrointestinal adverse events graded moderate or severe after 30 μg and 60 μg doses. V14 was 1 week after V12. EOS, end of study; EOT, end of treatment; V, visit.
Fig. 2
Fig. 2
Trial profile. For cohort 1 and cohort 2, the Nexvax2 starting dose was 30 μg; for cohort 1′ and cohort 2′, the Nexvax2 starting dose was 3 μg.
Fig. 3
Fig. 3
Incidence, severity, and organ class of treatment-emergent adverse events after each dose. Treatment-emergent adverse events after each dose of Nexvax2 or placebo are shown as the number of participants who experienced no, mild, moderate, severe, or serious treatment-emergent adverse events in (A), (C), (E), (G), (I), and (K) and as the total number of treatment-emergent adverse events classified by organ system in (B), (D), (F), (H), (J), and (L). PT, post-treatment.
Fig. 4
Fig. 4
Median fold change in plasma cytokines and chemokines following administration of Nexvax2. Assessments were made during the escalation phase, at 150 μg of Nexvax2 (previously defined maximum tolerated dose), and after the first, second, forth, and eighth administrations at the 300 μg and 900 μg maintenance doses. Plasma cytokines and chemokines were measured pre-treatment, and at 4, 6, and 10 h post-treatment.
Fig. 5
Fig. 5
Plasma concentrations of Nexvax2 peptides. Plasma concentrations of NPL001, NPL002, and NPL003 peptides at 45 min after intradermal administration of Nexvax2 in cohort 3 (n = 10). Mean (95% CI) concentrations are shown for NPL001 (A), NPL002 (B), and NPL003 (C) after escalating doses of Nexvax2, and at the maintenance dose of 900 μg. The lower level of quantitation (LLOQ) for each peptide was 2 ng/mL indicated by the dashed line; readings below the LLOQ were assigned 2 ng/mL. Pre-treatment plasma concentrations of Nexvax2 peptides were below the LLOQ for each of the indicated doses in all participants.

References

    1. Alhadj A.M., Liu Y.F., Arif S., Tatovic D., Shariff H., Gibson V.B., Yusuf N., Baptista R., Eichmann M., Petrov N., Heck S., Yang J.H.M., Tree T.I.M., Pujol-Autonell I., Yeo L., Baumard L.R., Stenson R., Howell A., Clark A., Boult Z., Powrie J., Adams L., Wong F.S., Luzio S., Dunseath G., Green K., O'Keefe A., Bayly G., Thorogood N., Andrews R., Leech N., Joseph F., Nair S., Seal S., Cheung H., Beam C., Hills R., Peakman M., Dayan C.M. Metabolic and immune effects of immunotherapy with proinsulin peptide in human new-onset type 1 diabetes. Sci. Transl. Med. 2017;9 (pii, eaaf7779)
    1. Anderson R.P., Jabri B. Vaccine against autoimmune disease: antigen-specific immunotherapy. Curr. Opin. Immunol. 2013;25:410–417.
    1. Brown G.J., Daveson J., Marjason J.K., Ffrench R.A., Smith D., Sullivan M., Tye-Din J.A., Anderson R.P. A phase I study to determine safety, tolerability and bioactivity of Nexvax2 in HLA DQ2 + volunteers with celiac disease following a long-term, strict gluten-free diet. Gastroenterology. 2011;140:S437–8.
    1. Burks A.W., Calderon M.A., Casale T., Cox L., Demoly P., Jutel M., Nelson H., Akdis C.A. Update on allergy immunotherapy: American Academy of Allergy, Asthma & Immunology/European Academy of Allergy and Clinical Immunology/PRACTALL consensus report. J. Allergy Clin. Immunol. 2013;131 (1288-96.e3)
    1. Burton B.R., Britton G.J., Fang H., Verhagen J., Smithers B., Sabatos-Peyton C.A., Carney L.J., Gough J., Strobel S., Wraith D.C. Sequential transcriptional changes dictate safe and effective antigen-specific immunotherapy. Nat. Commun. 2014;5:4741.
    1. Chatenoud L., Ferran C., Legendre C., Thouard I., Merite S., Reuter A., Gevaert Y., Kreis H., Franchimont P., Bach J.F. In vivo cell activation following OKT3 administration. Systemic cytokine release and modulation by corticosteroids. Transplantation. 1990;49:697–702.
    1. Goel G., Mayassi T., Qiao S.-W., Ciszewski C., King T., Daveson A.J., Andrews J.M., Krishnarajah J., Krause R., Brown G.J., Fogel R., Barish C.F., Epstein R., Kinney T., Miner P.B., Tye-Din J.A., Girardin A., Goldstein K., Dzuris J.L., Williams L.J., Xavier R., Sollid L.M., Jabri B., Anderson R.P. A single intradermal (ID) injection of Nexvax2, a peptide composition with dominant epitopes for gluten-reactive CD4 + T cells, activates T cells and tiggers acute gastrointestinal symptoms in HLA-DQ2.5 + people with celiac disease (CeD) Gastroenterology. 2016;150:S304.
    1. Goel G., King T., Daveson A.J., Andrews J.M., Krishnarajah J., Krause R., Brown G.J.E., Fogel R., Barish C.F., Epstein R., Kinney T.P., Miner P.B., Jr., Tye-Din J.A., Girardin A., Taavela J., Popp A., Sidney J., Mäki M., Goldstein K.E., Griffin P.H., Wang S., Dzuris J.L., Williams L.J., Sette A., Xavier R.J., Sollid L.M., Jabri B., Anderson R.P. Epitope-specific immunotherapy targeting CD4-positive T cells in coeliac disease: two randomised, double-blind, placebo-controlled phase 1 studies. Lancet Gastroenterol. Hepatol. 2017;2:479–493.
    1. Haselden B.M., Kay A.B., Larché M. Immunoglobulin E-independent major histocompatibility complex-restricted T cell peptide epitope-induced late asthmatic reactions. J. Exp. Med. 1999;189:1885–1894.
    1. Hoppe C., Gobel R., Kristensen M., Lind M.V., Matthiessen J., Christensen T., Trolle E., Fagt S., Madsen M.L., Husby S. Intake and sources of gluten in 20- to 75-year-old Danish adults: a national dietary survey. Eur. J. Nutr. 2017;56:107–117.
    1. Karell K., Louka A.S., Moodie S.J., Ascher H., Clot F., Greco L., Ciclitira P.J., Sollid L.M., Partanen J., European Genetics Cluster on Celiac Disease HLA types in celiac disease patients not carrying the DQA1*05-DQB1*02 (DQ2) heterodimer: results from the European Genetics Cluster on Celiac Disease. Hum. Immunol. 2003;64:469–477.
    1. Kasarda D.D. Can an increase in celiac disease be attributed to an increase in the gluten content of wheat as a consequence of wheat breeding? J. Agric. Food Chem. 2013;61:1155–1159.
    1. Kim H.S., Patel K.G., Orosz E., Kothari N., Demyen M.F., Pyrsopoulos N., Ahlawat S.K. Time trends in the prevalence of celiac disease and gluten-free diet in the US population: results from the National Health and Nutrition Examination Surveys 2009-2014. JAMA Intern. Med. 2016;176:1716–1717.
    1. Kurada S., Yadav A., Leffler D.A. Current and novel therapeutic strategies in celiac disease. Expert. Rev. Clin. Pharmacol. 2016;9:1211–1223.
    1. Larche M., Wraith D.C. Peptide-based therapeutic vaccines for allergic and autoimmune diseases. Nat. Med. 2005;11:S69–76.
    1. Leffler D.A., Dennis M., Edwards George J.B., Jamma S., Magge S., Cook E.F., Schuppan D., Kelly C.P. A simple validated gluten-free diet adherence survey for adults with celiac disease. Clin. Gastroenterol. Hepatol. 2009;7:530–536.
    1. Ludvigsson J.F., Bai J.C., Biagi F., Card T.R., Ciacci C., Ciclitira P.J., Green P.H., Hadjivassiliou M., Holdoway A., van Heel D.A., Kaukinen K., Leffler D.A., Leonard J.N., Lundin K.E., McGough N., Davidson M., Murray J.A., Swift G.L., Walker M.M., Zingone F., Sanders D.S., BSG Coeliac Disease Guidelines Development Group, British Society of Gastroenterology Diagnosis and management of adult coeliac disease: guidelines from the British Society of Gastroenterology. Gut. 2014;63:1210–1228.
    1. McPherson R.C., Konkel J.E., Prendergast C.T., Thomson J.P., Ottaviano R., Leech M.D., Kay O., Zandee S.E., Sweenie C.H., Wraith D.C., Meehan R.R., Drake A.J., Anderton S.M. Epigenetic modification of the PD-1 (Pdcd1) promoter in effector CD4(+) T cells tolerized by peptide immunotherapy. elife. 2014;29:3.
    1. Medawar P.B. Immunological tolerance. Science. 1961;133:303–306.
    1. Milewski M., Manser K., Nissley B.P., Mitra A. Analysis of the absorption kinetics of macromolecules following intradermal and subcutaneous administration. Eur. J. Pharm. Biopharm. 2015;89:134–144.
    1. Murray J.A., Moore S.B., Van Dyke C.T., Lahr B.D., Dierkhising R.A., Zinsmeister A.R., Melton L.J., 3rd, Kroning C.M., El-Yousseff M., Czaja A.J. HLA DQ gene dosage and risk and severity of celiac disease. Clin. Gastroenterol. Hepatol. 2007;5(12):1406.
    1. Pape K.A., Merica R., Mondino A., Khoruts A., Jenkins M.K. Direct evidence that functionally impaired CD4 + T cells persist in vivo following induction of peripheral tolerance. J. Immunol. 1998;160:4719–4729.
    1. Ráki M., Fallang L.E., Brottveit M., Bergseng E., Quarsten H., Lundin K.E., Sollid L.M. Tetramer visualization of gut-homing gluten-specific T cells in the peripheral blood of celiac disease patients. Proc. Natl. Acad. Sci. U. S. A. 2007;104:2831–2836.
    1. Sabatos-Peyton C.A., Verhagen J., Wraith D.C. Antigen-specific immunotherapy of autoimmune and allergic diseases. Curr. Opin. Immunol. 2010;22:609–615.
    1. See J.A., Kaukinen K., Makharia G.K., Gibson P.R., Murray J.A. Practical insights into gluten-free diets. Nat. Rev. Gastroenterol. Hepatol. 2015;12:580–591.
    1. Sollid L.M., Jabri B. Triggers and drivers of autoimmunity: lessons from coeliac disease. Nat. Rev. Immunol. 2013;13:294–302.
    1. Streeter H.B., Rigden R., Martin K.F., Scolding N.J., Wraith D.C. Preclinical development and first-in-human study of ATX-MS-1467 for immunotherapy of MS. Neurol. Neuroimmunol. Neuroinflammation. 2015;2
    1. Svedlund J., Sjodin I., Dotevall G. GSRS – a clinical rating scale for gastrointestinal symptoms in patients with irritable bowel syndrome and peptic ulcer disease. Dig. Dis. Sci. 1988;33:129–134.
    1. Tye-Din J.A., Stewart J.A., Dromey J.A., Beissbarth T., van Heel D.A., Tatham A., Henderson K., Mannering S.I., Gianfrani C., Jewell D.P., Hill A.V., McCluskey J., Rossjohn J., Anderson R.P. Comprehensive, quantitative mapping of T cell epitopes in gluten in celiac disease. Sci. Transl. Med. 2010;2 (41ra51)
    1. Tye-Din J.A., Dzuris J.L., Russell A.K., Wang S., Goldstein K.E., Williams L.J., Anderson R.P. Gluten ingestion and intradermal injection of peptides that activate gluten-specific CD4 + T cells elicit a cytokine signature dominated by interleukin-2 in celiac disease. United European Gastroenterol. J. 2017;5:A26–27.

Source: PubMed

3
Abonner