Tolerogenic nanoparticles mitigate the formation of anti-drug antibodies against pegylated uricase in patients with hyperuricemia

Earl Sands, Alan Kivitz, Wesley DeHaan, Sheldon S Leung, Lloyd Johnston, Takashi Kei Kishimoto, Earl Sands, Alan Kivitz, Wesley DeHaan, Sheldon S Leung, Lloyd Johnston, Takashi Kei Kishimoto

Abstract

Biologic drugs have transformed the standard of care for many diseases. However, many biologics induce the formation of anti-drug antibodies (ADAs), which can compromise their safety and efficacy. Preclinical studies demonstrate that biodegradable nanoparticles-encapsulating rapamycin (ImmTOR), but not free rapamycin, mitigate the immunogenicity of co-administered biologic drugs. Here we report the outcomes from two clinical trials for ImmTOR. In the first ascending dose, open-label study (NCT02464605), pegadricase, an immunogenic, pegylated uricase enzyme derived from Candida utilis, is assessed for safety and tolerability (primary endpoint) as well as activity and immunogenicity (secondary endpoint); in the second single ascending dose Phase 1b trial (NCT02648269) composed of both a double-blind and open-label parts, we evaluate the safety of ImmTOR (primary endpoint) and its ability to prevent the formation of anti-drug antibodies against pegadricase and enhance its pharmacodynamic activity (secondary endpoint) in patients with hyperuricemia. The combination of ImmTOR and pegadricase is well tolerated. ImmTOR inhibits the development of uricase-specific ADAs in a dose-dependent manner, thus enabling sustained enzyme activity and reduction in serum uric acid levels. ImmTOR may thus represent a feasible approach for preventing the formation of ADAs to a broad range of immunogenic biologic therapies.

Conflict of interest statement

E.S., W.D., S.S.L., L.J., and T.K.K are current or former employees and shareholders of Selecta Biosciences.

© 2022. The Author(s).

Figures

Fig. 1. Schematic of ImmTOR mechanism of…
Fig. 1. Schematic of ImmTOR mechanism of action. ImmTOR is selectively taken up by antigen-presenting cells, such as dendritic cells (DC) in the spleen and liver.
ImmTOR induces tolerogenic DCs that process and present co-administered antigen in a manner that results in the expansion of antigen-specific regulatory T cells. The Tregs inhibit activation of effector T cells and prevent the formation of anti-drug antibodies. Adapted from Kishimoto.
Fig. 2. Single ascending dose study of…
Fig. 2. Single ascending dose study of pegadricase in patients with hyperuricemia.
A Patients with sUA > 6 mg/dL at screening were assigned to one of five cohorts receiving a single IV infusion of pegadricase (0.1, 0.2, 0.4, 0.8 or 1.2 mg/kg). Each cohort consisted of 5 patients, except for cohort 5 (1.2 mg/kg) which enrolled 2 patients. Blood was drawn at various timepoints indicated and sUA levels were determined. Lines represent the mean for each cohort and error bars represent the standard deviation. Only the mean is depicted for cohort 5. B Serum uric acid, serum pegadricase activity, and anti-uricase IgG antibodies are shown for the five individual patients in cohort 3 (0.4 mg/kg pegadricase). One patient (square symbols) developed only low anti-uricase IgG titers and showed prolonged reduction of sUA and sustained serum pegadricase activity. Source data are provided as a Source Data file.
Fig. 3. Single ascending dose study of…
Fig. 3. Single ascending dose study of ImmTOR and SEL-212 in patients with hyperuricemia.
A Schematic of cohorts dosed with pegadricase alone (Cohort A), single escalating doses of ImmTOR alone (Cohorts B–E), or single escalating doses of SEL-212 (Cohorts F–J, 0.03, 0.1, 0.15, or 0.3 mg/kg ImmTOR combined with a fixed dose of 0.4 mg/kg pegadricase). Cohorts B–E were double blinded and randomized to either placebo or study drug in a 2:5 rato, while Cohorts A and F–J were open-label. All cohorts consisted of 5 drug-treated patients, except for Cohort G, that contained 10 patients and Cohort J which contained 6 patients. In addition, Cohorts B–F each contained two patients that received placebo (vehicle). Patients in Cohorts F–I received an IV infusion of ImmTOR followed immediately by an IV infusion of pegadricase. Patients in Cohort J received an IV infusion of pegadricase followed 48 h later by an IV infusion of ImmTOR. B Serum uric acid levels over time. Each symbol represents sUA levels for an individual patient at various timepoints indicated and the lines indicate mean sUA levels for the cohort. The dotted black line represents 6 mg/dL sUA. Source data are provided as a Source Data file.
Fig. 4. Correlation of sUA with anti-uricase…
Fig. 4. Correlation of sUA with anti-uricase IgG and serum pegadricase activity.
Anti-uricase IgG titers, serum pegadricase activity, and sUA are plotted against time for individual patients in cohorts A, G, H, and I. Each line represents an individual patient.
Fig. 5. Serum uric acid and anti-uricase…
Fig. 5. Serum uric acid and anti-uricase IgG for selected patients followed past day 30.
Patients in cohorts G, H, and I that maintained sUA levels below 6 mg/dL for 30 days were selected on a voluntary basis to participate in additional monitoring. Serum uric acid and anti-uricase IgG titers are plotted for individual patients at various timepoints indicated. Patients in cohorts G, H, and I are represented by circle, square, and triangle symbols, respectively. All patients showed a rebound in sUA levels by day 50, indicating clearance of enzyme. Ten of the twelve patients showed no increase in anti-uricase IgG titers after day 30 and the remaining two patients showed only modest elevations in anti-uricase IgG titers. Source data are provided as a Source Data file.

References

    1. Sriranganathan, M. K., Vinik, O., Pardo Pardo, J., Bombardier, C. & Edwards, C. J. Interventions for tophi in gout. Cochrane. Database. Syst. Rev.8, CD010069 (2021).
    1. Sundy JS, et al. Efficacy and tolerability of pegloticase for the treatment of chronic gout in patients refractory to conventional treatment: two randomized controlled trials. JAMA. 2011;306:711–720. doi: 10.1001/jama.2011.1617.
    1. Baraf HS, et al. Tophus burden reduction with pegloticase: results from phase 3 randomized trials and open-label extension in patients with chronic gout refractory to conventional therapy. Arthritis. Res. Ther. 2013;15:R137. doi: 10.1186/ar4318.
    1. Mandell BF, Yeo AE, Lipsky PE. Tophus resolution in patients with chronic refractory gout who have persistent urate-lowering responses to pegloticase. Arthritis. Res. Ther. 2018;20:286. doi: 10.1186/s13075-018-1782-x.
    1. Araujo EG, et al. Tophus resolution with pegloticase: a prospective dual-energy CT study. RMD. Open. 2015;1:e000075. doi: 10.1136/rmdopen-2015-000075.
    1. Richette P, et al. 2016 updated EULAR evidence-based recommendations for the management of gout. Ann. Rheum. Dis. 2017;76:29–42. doi: 10.1136/annrheumdis-2016-209707.
    1. Lipsky PE, et al. Pegloticase immunogenicity: The relationship between efficacy and antibody development in patients treated for refractory chronic gout. Arthritis. Res. Ther. 2014;16:R60. doi: 10.1186/ar4497.
    1. Kishimoto TK. Development of ImmTOR tolerogenic nanoparticles for the mitigation of anti-drug antibodies. Front. Immunol. 2020;11:969. doi: 10.3389/fimmu.2020.00969.
    1. Kishimoto TK, et al. Improving the efficacy and safety of biologic drugs with tolerogenic nanoparticles. Nat. Nanotechnol. 2016;11:890–899. doi: 10.1038/nnano.2016.135.
    1. Saunders RN, Metcalfe MS, Nicholson ML. Rapamycin in transplantation: A review of the evidence. Kidney. Int. 2001;59:3–16. doi: 10.1046/j.1523-1755.2001.00460.x.
    1. Fischer, R., Turnquist, H. R., Taner, T. & Thomson, A. W. Use of rapamycin in the induction of tolerogenic dendritic cells. Handb. Exp. Pharmacol.188, 215–232 (2009).
    1. Ilyinskii PO, Roy CJ, LePrevost J, Rizzo GL, Kishimoto TK. Enhancement of the tolerogenic phenotype in the liver by ImmTOR nanoparticles. Front. Immunol. 2021;12:637469. doi: 10.3389/fimmu.2021.637469.
    1. Maldonado RA, et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc. Natl. Acad. Sci. U S A. 2015;112:E156–165. doi: 10.1073/pnas.1408686111.
    1. Mazor R, et al. Tolerogenic nanoparticles restore the antitumor activity of recombinant immunotoxins by mitigating immunogenicity. Proc. Natl. Acad. Sci. U S A. 2018;115:E733–E742. doi: 10.1073/pnas.1717063115.
    1. LaMothe RA, et al. Tolerogenic nanoparticles induce antigen-specific regulatory T Cells and provide therapeutic efficacy and transferrable tolerance against experimental autoimmune encephalomyelitis. Front. Immunol. 2018;9:281. doi: 10.3389/fimmu.2018.00281.
    1. Meliani A, et al. Antigen-selective modulation of AAV immunogenicity with tolerogenic rapamycin nanoparticles enables successful vector re-administration. Nat. Commun. 2018;9:4098. doi: 10.1038/s41467-018-06621-3.
    1. Boers-Doets CB, et al. Mammalian target of rapamycin inhibitor-associated stomatitis. Future. Oncol. 2013;9:1883–1892. doi: 10.2217/fon.13.141.
    1. Guttmann A, Krasnokutsky S, Pillinger MH, Berhanu A. Pegloticase in gout treatment - safety issues, latest evidence and clinical considerations. Ther. Adv. Drug. Saf. 2017;8:379–388. doi: 10.1177/2042098617727714.
    1. Sundy JS, et al. Pharmacokinetics and pharmacodynamics of intravenous PEGylated recombinant mammalian urate oxidase in patients with refractory gout. Arthritis. Rheum. 2007;56:1021–1028. doi: 10.1002/art.22403.
    1. Mahlangu JN, et al. Changes in the amino acid sequence of the recombinant human factor VIIa analog, vatreptacog alfa, are associated with clinical immunogenicity. J. Thromb. Haemost. 2015;13:1989–1998. doi: 10.1111/jth.13141.
    1. Ridker PM, et al. Lipid-reduction variability and antidrug-antibody formation with Bococizumab. N. Engl. J. Med. 2017;376:1517–1526. doi: 10.1056/NEJMoa1614062.
    1. Liao K, et al. Characterization of the robust humoral immune response to GSK2618960, a humanized anti-IL-7 receptor monoclonal antibody, observed in healthy subjects in a Phase 1 study. PLoS. One. 2021;16:e0249049. doi: 10.1371/journal.pone.0249049.
    1. Herold, K. C. et al. Immunomodulatory activity of humanized anti-IL-7R monoclonal antibody RN168 in subjects with type 1 diabetes. JCI. Insight.4, e126054 (2019).
    1. Li J, et al. Thrombocytopenia caused by the development of antibodies to thrombopoietin. Blood. 2001;98:3241–3248. doi: 10.1182/blood.V98.12.3241.
    1. Parenky A, et al. New FDA draft guidance on immunogenicity. AAPS. J. 2014;16:499–503. doi: 10.1208/s12248-014-9587-6.
    1. Pegvaliase-pqpz BLA approval letter .
    1. Zhang AH, Rossi RJ, Yoon J, Wang H, Scott DW. Tolerogenic nanoparticles to induce immunologic tolerance: Prevention and reversal of FVIII inhibitor formation. Cell. Immunol. 2016;301:74–81. doi: 10.1016/j.cellimm.2015.11.004.
    1. Lim HH, et al. A pilot study on using rapamycin-carrying synthetic vaccine particles (SVP) in conjunction with enzyme replacement therapy to induce immune tolerance in Pompe disease. Mol .Genet. Metab. Rep. 2017;13:18–22. doi: 10.1016/j.ymgmr.2017.03.005.
    1. Ilyinskii, P. O. et al. Enhancement of liver-directed transgene expression at initial and repeat doses of AAV vectors admixed with ImmTOR nanoparticles. Sci. Adv.7, eabd0321 (2021).
    1. Bomalaski JS, Holtsberg FW, Ensor CM, Clark MA. Uricase formulated with polyethylene glycol (uricase-PEG 20): Biochemical rationale and preclinical studies. J. Rheumatol. 2002;29:1942–1949.

Source: PubMed

3
Se inscrever