Safety of the use of gold nanoparticles conjugated with proinsulin peptide and administered by hollow microneedles as an immunotherapy in type 1 diabetes

D Tatovic, M A McAteer, J Barry, A Barrientos, K Rodríguez Terradillos, I Perera, E Kochba, Y Levin, M Dul, S A Coulman, J C Birchall, C von Ruhland, A Howell, R Stenson, M Alhadj Ali, S D Luzio, G Dunseath, W Y Cheung, G Holland, K May, J R Ingram, M M U Chowdhury, F S Wong, R Casas, C Dayan, J Ludvigsson, D Tatovic, M A McAteer, J Barry, A Barrientos, K Rodríguez Terradillos, I Perera, E Kochba, Y Levin, M Dul, S A Coulman, J C Birchall, C von Ruhland, A Howell, R Stenson, M Alhadj Ali, S D Luzio, G Dunseath, W Y Cheung, G Holland, K May, J R Ingram, M M U Chowdhury, F S Wong, R Casas, C Dayan, J Ludvigsson

Abstract

Antigen-specific immunotherapy is an immunomodulatory strategy for autoimmune diseases, such as type 1 diabetes, in which patients are treated with autoantigens to promote immune tolerance, stop autoimmune β-cell destruction and prevent permanent dependence on exogenous insulin. In this study, human proinsulin peptide C19-A3 (known for its positive safety profile) was conjugated to ultrasmall gold nanoparticles (GNPs), an attractive drug delivery platform due to the potential anti-inflammatory properties of gold. We hypothesised that microneedle intradermal delivery of C19-A3 GNP may improve peptide pharmacokinetics and induce tolerogenic immunomodulation and proceeded to evaluate its safety and feasibility in a first-in-human trial. Allowing for the limitation of the small number of participants, intradermal administration of C19-A3 GNP appears safe and well tolerated in participants with type 1 diabetes. The associated prolonged skin retention of C19-A3 GNP after intradermal administration offers a number of possibilities to enhance its tolerogenic potential, which should be explored in future studies.

Keywords: gold nanoparticle; microneedle; peptide immunotherapy; proinsulin; type 1 diabetes.

© The Author(s) 2022. Published by Oxford University Press on behalf of the British Society for Immunology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Study recruitment and visit schedule. (a) Consort diagram and (b) schedule of the study visits. D, dose; V, visit; W, week.
Figure 2.
Figure 2.
Effect of C19-A3 GNP on β-cell function and metabolic parameters. (a) Serum C-peptide expressed as area under the curve (AUC) over 120 min after mixed-meal challenge; (b) urine C-peptide to creatinine ratio (UCPCR) 2 h after mixed meal; (c) HbA1c; (d) median total daily insulin dose recorded over 3 days before the study visit and normalised for body weight; (e) insulin dose-adjusted A1c (IDAA1c) calculated according to the formula: HbA1c (%) + [4× insulin dose (units per kg per 24 h)]. Each line represents an individual participant with an assigned study number as shown in the legend.
Figure 3.
Figure 3.
Effect of C19-A3 GNP on glucose variability. (a) Time in range (the % of time participants recorded a blood glucose level of 4–10 mmol/l); (b) time above range (the % of time participants recorded a blood glucose level of >10 mmol/l); (c) time below range (the % of time participants recorded a blood glucose level of

Figure 4.

Serum and urine gold concentration…

Figure 4.

Serum and urine gold concentration following administration of C19-A3 GNP. (a) Serum gold…

Figure 4.
Serum and urine gold concentration following administration of C19-A3 GNP. (a) Serum gold concentration; (b) urine gold concentration. Blood and urine samples for measurement of gold concentration were taken at baseline (week 0), one day after 1st dose (week 0 + 1 day) and one day after 3rd dose (week 8 + 1 day). Detection limit was 0.2 ng/ml and quantitation limit 0.6 ng/ml. Levels below detection and quantitation limit are presented as zero on the graph. Each line represents an individual participant with an assigned study number as shown in the legend.

Figure 5.

Skin changes after intradermal injection…

Figure 5.

Skin changes after intradermal injection of C19-A3 GNP. (a) Immediately after injection; (b)…

Figure 5.
Skin changes after intradermal injection of C19-A3 GNP. (a) Immediately after injection; (b) 5 min after injection; (c) 24 h after injection; (d) 30 days after injection; (e) 2 months after injection (upper left), 1 month after injection (right) and 7 days after injection (lower left); (f) 20 months after injection; (g) close-up of the injection site showing central area of hyperpigmentation and surrounding induration and redness.

Figure 6.

Histopathology and immunohistochemistry staining of…

Figure 6.

Histopathology and immunohistochemistry staining of the punch biopsies of the injection sites. (a)…

Figure 6.
Histopathology and immunohistochemistry staining of the punch biopsies of the injection sites. (a) Anti-CD3 staining; (b) anti-CD20 staining; (c) anti-CD4 staining; (d) anti-CD8 staining; (e) and (f) gold staining.
All figures (7)
Figure 4.
Figure 4.
Serum and urine gold concentration following administration of C19-A3 GNP. (a) Serum gold concentration; (b) urine gold concentration. Blood and urine samples for measurement of gold concentration were taken at baseline (week 0), one day after 1st dose (week 0 + 1 day) and one day after 3rd dose (week 8 + 1 day). Detection limit was 0.2 ng/ml and quantitation limit 0.6 ng/ml. Levels below detection and quantitation limit are presented as zero on the graph. Each line represents an individual participant with an assigned study number as shown in the legend.
Figure 5.
Figure 5.
Skin changes after intradermal injection of C19-A3 GNP. (a) Immediately after injection; (b) 5 min after injection; (c) 24 h after injection; (d) 30 days after injection; (e) 2 months after injection (upper left), 1 month after injection (right) and 7 days after injection (lower left); (f) 20 months after injection; (g) close-up of the injection site showing central area of hyperpigmentation and surrounding induration and redness.
Figure 6.
Figure 6.
Histopathology and immunohistochemistry staining of the punch biopsies of the injection sites. (a) Anti-CD3 staining; (b) anti-CD20 staining; (c) anti-CD4 staining; (d) anti-CD8 staining; (e) and (f) gold staining.

References

    1. Foster NC, Beck RW, Miller al. . State of type 1 diabetes management and outcomes from the T1D exchange in 2016-2018. Diabetes Technol Ther 2019;21:66–72.
    1. Skyler JS. Primary and secondary prevention of Type 1 diabetes. Diabet Med. 2013;30(2):161–9.
    1. Dayan CM, Korah M, Tatovic al. . Changing the landscape for type 1 diabetes: the first step to prevention. Lancet 2019;394:1286–96.
    1. Mallone R, Brezar V, Boitard C.. T cell recognition of autoantigens in human type 1 diabetes: clinical perspectives. Clin Dev Immunol 2011;2011:513210.
    1. Tian J, Kaufman DL.. Antigen-based therapy for the treatment of type 1 diabetes. Diabetes 2009;58:1939–46.
    1. von Herrath M, Peakman M, Roep B.. Progress in immune-based therapies for type 1 diabetes. Clin Exp Immunol 2013;172:186–202.
    1. Atkinson MA, Roep BO, Posgai al. . The challenge of modulating β-cell autoimmunity in type 1 diabetes. Lancet Diabetes Endocrinol 2019;7:52–64.
    1. Roep BO, Wheeler DCS, Peakman M.. Antigen-based immune modulation therapy for type 1 diabetes: the era of precision medicine. Lancet Diabetes Endocrinol 2019;7:65–74.
    1. Ludvigsson J. Autoantigen treatment in type 1 diabetes: unsolved questions on how to select autoantigen and administration route. Int J Mol Sci. 2020;21(5):1958.
    1. Diabetes Prevention Trial—Type 1 Diabetes Study G. Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med. 2002;346(22):1685–91.
    1. Orban T, Farkas K, Jalahej al. . Autoantigen-specific regulatory T cells induced in patients with type 1 diabetes mellitus by insulin B-chain immunotherapy. J Autoimmun 2010;34:408–15.
    1. Ludvigsson J, Faresjö M, Hjorth al. . GAD treatment and insulin secretion in recent-onset type 1 diabetes. N Engl J Med 2008;359:1909–20.
    1. Wherrett DK, Bundy B, Becker al. .; Type 1 Diabetes TrialNet GAD Study Group. Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial. Lancet 2011;378:319–27.
    1. Ludvigsson J, Krisky D, Casas al. . GAD65 antigen therapy in recently diagnosed type 1 diabetes mellitus. N Engl J Med 2012;366:433–42.
    1. Chaillous L, Lefèvre H, Thivolet al. . Oral insulin administration and residual beta-cell function in recent-onset type 1 diabetes: a multicentre randomised controlled trial. Diabète Insuline Orale group. Lancet 2000;356:545–9.
    1. Skyler JS, Krischer JP, Wolfsdorf al. . Effects of oral insulin in relatives of patients with type 1 diabetes: the diabetes prevention trial—type 1. Diabetes Care 2005;28:1068–76.
    1. Näntö-Salonen K, Kupila A, Simell al. . Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a double-blind, randomised controlled trial. Lancet 2008;372:1746–55.
    1. Fourlanos S, Perry C, Gellert al. . Evidence that nasal insulin induces immune tolerance to insulin in adults with autoimmune diabetes. Diabetes 2011;60:1237–45.
    1. Weaver DJ Jr, Liu B, Tisch R.. Plasmid DNAs encoding insulin and glutamic acid decarboxylase 65 have distinct effects on the progression of autoimmune diabetes in nonobese diabetic mice. J Immunol 2001;167:586–92.
    1. Johnson MC, Wang B, Tisch R.. Genetic vaccination for re-establishing T-cell tolerance in type 1 diabetes. Hum Vaccin 2011;7:27–36.
    1. Walter M, Philotheou A, Bonnici al. .; NBI-6024 Study Group. No effect of the altered peptide ligand NBI-6024 on beta-cell residual function and insulin needs in new-onset type 1 diabetes. Diabetes Care 2009;32:2036–40.
    1. Mathieu C, Gillard P.. Arresting type 1 diabetes after diagnosis: GAD is not enough. Lancet 2011;378:291–2.
    1. Adorini L, Penna G.. Induction of tolerogenic dendritic cells by vitamin D receptor agonists. Handb Exp Pharmacol 2009;188:251–73.
    1. Ali MA, Thrower SL, Hanna al. . Topical steroid therapy induces pro-tolerogenic changes in Langerhans cells in human skin. Immunology 2015;146:411–22.
    1. Boks MA, Kager-Groenland JR, Haasjes al. . IL-10-generated tolerogenic dendritic cells are optimal for functional regulatory T cell induction – a comparative study of human clinical-applicable DC. Clin Immunol 2012;142:332–42.
    1. Hannelius U, Beam CA, Ludvigsson J.. Efficacy of GAD-alum immunotherapy associated with HLA-DR3-DQ2 in recently diagnosed type 1 diabetes. Diabetologia 2020;63:2177–81.
    1. Thrower SL, James L, Hall al. . Proinsulin peptide immunotherapy in type 1 diabetes: report of a first-in-man Phase I safety study. Clin Exp Immunol 2009;155:156–65.
    1. Alhadj Ali M, Liu YF, Arif al. . Metabolic and immune effects of immunotherapy with proinsulin peptide in human new-onset type 1 diabetes. Sci Transl Med. 2017;9(402):eaaf7779.
    1. de Araújo RF Júnior, de Araújo AA, Pessoa al. . Anti-inflammatory, analgesic and anti-tumor properties of gold nanoparticles. Pharmacol Rep 2017;69:119–29.
    1. Dul M, Nikolic T, Stefanidou al. .; EE-ASI Consortium. Conjugation of a peptide autoantigen to gold nanoparticles for intradermally administered antigen specific immunotherapy. Int J Pharm 2019;562:303–12.
    1. Merad M, Ginhoux F, Collin M.. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol 2008;8:935–47.
    1. Steinman RM, Hawiger D, Nussenzweig MC.. Tolerogenic dendritic cells. Annu Rev Immunol 2003;21:685–711.
    1. Klechevsky E, Banchereau J.. Human dendritic cells subsets as targets and vectors for therapy. Ann N Y Acad Sci 2013;1284:24–30.
    1. Chu CC, Ali N, Karagiannis al. . Resident CD141 (BDCA3)+ dendritic cells in human skin produce IL-10 and induce regulatory T cells that suppress skin inflammation. J Exp Med 2012;209:935–45.
    1. Singh RK, Malosse C, Davies al. . Using gold nanoparticles for enhanced intradermal delivery of poorly soluble auto-antigenic peptides. Nanomedicine 2021;32:102321.
    1. Levin Y, Kochba E, Hung al. . Intradermal vaccination using the novel microneedle device MicronJet600: Past, present, and future. Hum Vaccin Immunother 2015;11:991–7.
    1. Levin Y, Kochba E, Shukarev al. . A phase 1, open-label, randomized study to compare the immunogenicity and safety of different administration routes and doses of virosomal influenza vaccine in elderly. Vaccine 2016;34:5262–72.
    1. Carter D, van Hoeven N, Baldwin al. . The adjuvant GLA-AF enhances human intradermal vaccine responses. Sci Adv 2018;4:eaas9930.
    1. Beals CR, Railkar RA, Schaeffer al. . Immune response and reactogenicity of intradermal administration versus subcutaneous administration of varicella-zoster virus vaccine: an exploratory, randomised, partly blinded trial. Lancet Infect Dis 2016;16:915–22.
    1. Hung IF, Yap DY, Yip al. . A double-blind randomized phase 2 controlled trial of intradermal hepatitis B vaccination with a topical Toll-like receptor 7 agonist imiquimod, in patients on dialysis. Clin Infect Dis. 2021;73:e304–11.
    1. Greenbaum CJ, Mandrup-Poulsen T, McGee al. .; Type 1 Diabetes Trial Net Research Group; European C-Peptide Trial Study Group. Mixed-meal tolerance test versus glucagon stimulation test for the assessment of beta-cell function in therapeutic trials in type 1 diabetes. Diabetes Care 2008;31:1966–71.
    1. Tatovic D, Luzio S, Dunseath al. .; MonoPepT1De Study Group. Stimulated urine C-peptide creatinine ratio vs serum C-peptide level for monitoring of β-cell function in the first year after diagnosis of Type 1 diabetes. Diabet Med 2016;33:1564–8.
    1. Newman GR, Jasani B.. Silver development in microscopy and bioanalysis: a new versatile formulation for modern needs. Histochem J 1998;30:635–45.
    1. Dhanjal TS, Lellouche N, von Ruhland al. . Massive accumulation of myofibroblasts in the critical isthmus is associated with ventricular tachycardia inducibility in post-infarct swine heart. Jacc Clin Electrophysiol 2017;3:703–14.
    1. Mortensen HB, Hougaard P, Swift al. .; Hvidoere Study Group on Childhood Diabetes. New definition for the partial remission period in children and adolescents with type 1 diabetes. Diabetes Care 2009;32:1384–90.
    1. Greenbaum CJ, Beam CA, Boulware al. .; Type 1 Diabetes TrialNet Study Group. Fall in C-peptide during first 2 years from diagnosis: evidence of at least two distinct phases from composite Type 1 Diabetes TrialNet data. Diabetes 2012;61:2066–73.
    1. Bruze M, Edman B, Björkner al. . Clinical relevance of contact allergy to gold sodium thiosulfate. J Am Acad Dermatol 1994;31:579–83.
    1. Chen JK, Lampel HP.. Gold contact allergy: clues and controversies. Dermatitis 2015;26:69–77.

Source: PubMed

3
Předplatit