Control of hyperuricemia in subjects with refractory gout, and induction of antibody against poly(ethylene glycol) (PEG), in a phase I trial of subcutaneous PEGylated urate oxidase

Nancy J Ganson, Susan J Kelly, Edna Scarlett, John S Sundy, Michael S Hershfield, Nancy J Ganson, Susan J Kelly, Edna Scarlett, John S Sundy, Michael S Hershfield

Abstract

PEG-modified recombinant mammalian urate oxidase (PEG-uricase) is being developed as a treatment for patients with chronic gout who are intolerant of, or refractory to, available therapy for controlling hyperuricemia. In an open-label phase I trial, single subcutaneous injections of PEG-uricase (4 to 24 mg) were administered to 13 such subjects (11 had tophaceous gout), whose plasma uric acid concentration (pUAc) was 11.3 +/- 2.1 mg/dl (mean +/- SD). By day seven after injection of PEG-uricase, pUAc had declined by an average of 7.9 mg/dl and had normalized in 11 subjects, whose mean pUAc decreased to 2.8 +/- 2.2 mg/dl. At doses of 8, 12, and 24 mg, the mean pUAc at 21 days after injection remained no more than 6 mg/dl. In eight subjects, plasma uricase activity was still measurable at 21 days after injection (half-life 10.5 to 19.9 days). In the other five subjects, plasma uricase activity could not be detected beyond ten days after injection; this was associated with the appearance of relatively low-titer IgM and IgG antibodies against PEG-uricase. Unexpectedly, these antibodies were directed against PEG itself rather than the uricase protein. Three PEG antibody-positive subjects had injection-site reactions at 8 to 9 days after injection. Gout flares in six subjects were the only other significant adverse reactions, and PEG-uricase was otherwise well tolerated. A prolonged circulating life and the ability to normalize plasma uric acid in markedly hyperuricemic subjects suggest that PEG-uricase could be effective in depleting expanded tissue stores of uric acid in subjects with chronic or tophaceous gout. The development of anti-PEG antibodies, which may limit efficacy in some patients, is contrary to the general assumption that PEG is non-immunogenic. PEG immunogenicity deserves further investigation, because it has potential implications for other PEGylated therapeutic agents in clinical use.

Figures

Figure 1
Figure 1
Plasma uricase activity and plasma uric acid concentration after subcutaneous injection of PEG-uricase. A single 8 mg injection of PEG-modified recombinant mammalian urate oxidase (PEG-uricase) was administered. The horizontal axis indicates days after dosing.
Figure 2
Figure 2
Relationship of dose of PEG-uricase and serum or plasma uric acid concentration. (a) Data for each dose cohort. (b) Data for two groups of subjects with distinct pharmacokinetic patterns: 'long-circulating' (plasma uricase activity (pUox) present three weeks after injection (n = 8)) and 'early elimination' (pUox undetectable beyond 10 days after injection (n = 5)). The horizontal axis indicates days after injection of PEG-modified recombinant mammalian urate oxidase (PEG-uricase); 'pre*', serum obtained at screening, just before the two-week allopurinol washout period; 'd 0', plasma obtained immediately before PEG-uricase injection.
Figure 3
Figure 3
Two pharmacokinetic patterns after single subcutaneous injections of PEG-uricase. (a) 'Long-circulating' group: eight subjects with uricase activity present in plasma at 21 days after injection. (b) 'Early elimination' group: five subjects with undetectable plasma uricase activity beyond 10 days after injection. The keys indicate the dose of PEG-modified recombinant mammalian urate oxidase (PEG-uricase).
Figure 4
Figure 4
Time course of appearance of IgM and IgG antibodies against PEG-uricase, and of plasma uricase activity, in 'early elimination' subjects 002, 011, and 013. Subject 002 received 4 mg, and subjects 011 and 013 each received 12 mg, of PEG-modified recombinant mammalian urate oxidase (PEG-uricase).
Figure 5
Figure 5
Titer of ELISA for IgG antibody against PEG-uricase. The data shown are for the five subjects who showed early elimination of plasma uricase activity. PEG-uricase, PEG-modified recombinant mammalian urate oxidase.
Figure 6
Figure 6
Competition ELISA to determine the specificity of IgG antibody against PEG-uricase. Increasing amounts of the indicated materials were added to aliquots of plasma obtained from subject 013 on day 14 after subcutaneous injection of 12 mg of PEG-modified recombinant mammalian urate oxidase (PEG-uricase). After incubation overnight, these mixtures were tested at a 1:60 dilution in the ELISA for IgG antibody against PEG-uricase. (a) 'Uricase' is the unmodified recombinant protein used in PEG-uricase, and 'PEG 10000' is 10 K monomethoxyPEG (mPEG) conjugated with glycine instead of uricase protein. 'PEG-PNP' is bacterial purine nucleoside phosphorylase conjugated with mPEG of molecular mass 5 kDa, prepared as described [18]. (b) 'PEG 350' and 'PEG 2000' are unconjugated, nonactivated mPEGs of molecular masses 350 and 2,000 Da. 'PEG 5000' and 'PEG 10000' are p-nitrophenyl carbonate-activated mPEGs of the indicated molecular masses conjugated with glycine.
Figure 7
Figure 7
Comparison of ELISAs performed with mPEGs and PEG-uricase. ELISA plates were coated with 2.5 μg of PEG-modified recombinant mammalian urate oxidase (PEG-uricase) or with 25 μg of p-nitrophenyl carbonate-activated monomethoxyPEGs (mPEGs) of molecular mass 5 kDa (mPEG-5K) or 10 kDa (mPEG-10K). After washing, the plates were blocked with 1% BSA, 1% glycine in PBS. The indicated plasma samples obtained on day 0 or day 21 after subcutaneous injection of PEG-uricase were then assayed with each substrate at a dilution of 1:50 as described in the Materials and methods section for the PEG-uricase ELISA.

References

    1. Becker MA. Hyperuricemia and gout. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editor. The Metabolic and Molecular Bases of Inherited Disease. 8. New York: McGraw-Hill; 2001. pp. 2513–2535.
    1. Wortmann RL, Kelley WN. Gout and hyperuricemia. In: Ruddy S, Harris ED Jr, Sledge CB, editor. Kelley's Textbook of Rheumatology. 6. St Louis: WB Saunders; 2001. pp. 1339–1371.
    1. Terkeltaub RA. Clinical practice. Gout. N Engl J Med. 2003;349:1647–1655. doi: 10.1056/NEJMcp030733.
    1. Riedel AA, Nelson M, Joseph-Ridge N, Wallace K, MacDonald P, Becker M. Compliance with allopurinol therapy among managed care enrollees with gout: a retrospective analysis of administrative claims. J Rheumatol. 2004;31:1575–1581.
    1. London M, Hudson PM. Uricolytic activity of purified uricase in two human beings. Science. 1957;125:937–938.
    1. Kissel P, Lamarche M, Royer R. Modification of uricaemia and the excretion of uric acid nitrogen by an enzyme of fungal origin. Nature. 1968;217:72–74.
    1. Goldman SC, Holcenberg JS, Finklestein JZ, Hutchinson R, Kreissman S, Johnson FL, Tou C, Harvey E, Morris E, Cairo MS. A randomized comparison between rasburicase and allopurinol in children with lymphoma or leukemia at high risk for tumor lysis. Blood. 2001;97:2998–3003. doi: 10.1182/blood.V97.10.2998.
    1. Coiffier B, Mounier N, Bologna S, Ferme C, Tilly H, Sonet A, Christian B, Casasnovas O, Jourdan E, Belhadj K, et al. Efficacy and safety of rasburicase (recombinant urate oxidase) for the prevention and treatment of hyperuricemia during induction chemotherapy of aggressive non-Hodgkin's lymphoma: results of the GRAAL1 (Groupe d'Étude des Lymphomes de l'Adulte Trial on Rasburicase Activity in Adult Lymphoma) study. J Clin Oncol. 2003;21:4402–4406. doi: 10.1200/JCO.2003.04.115.
    1. Abuchowski A, McCoy JR, Palczuk NC, van Es T, Davis FF. Effect of attachment of polyethylene glycol on immunogenicity and circulating life of bovine liver catalase. J Biol Chem. 1977;252:3582–3586.
    1. Veronese FM, Harris JM. Introduction and overview of peptide and protein pegylation. Adv Drug Deliv Rev. 2002;54:453–456. doi: 10.1016/S0169-409X(02)00029-7.
    1. Harris JM, Chess RB. Effect of pegylation on pharmaceuticals. Nat Rev Drug Discov. 2003;2:214–221. doi: 10.1038/nrd1033.
    1. Chua CC, Greenberg ML, Viau AT, Nucci M, Brenckman WD, Jr, Hershfield MS. Use of polyethylene glycol-modified uricase (PEG-uricase) to treat hyperuricemia in a patient with non-Hodgkin lymphoma. Ann Int Med. 1988;109:114–117.
    1. Kelly SJ, Delnomdedieu M, Oliverio MI, Williams LD, Saifer MGP, Sherman MR, Coffman TM, Johnson GA, Hershfield MS. Diabetes insipidus in uricase-deficient mice: a model for evaluating therapy with poly(ethylene glycol)-modified uricase. J Am Soc Nephrol. 2001;12:1001–1009.
    1. Ganson N, Kelly SJ, Scarlett E, Sundy JS, Hershfield MS. Antibodies to polyethylene glycol (PEG) during phase I investigation of PEG-urate oxidase (PEG-uricase; Puricase®) for refractory gout [abstract] Arthritis Rheum. 2004;50:s338.
    1. Greenberg ML, Hershfield MS. A radiochemical-high-performance liquid chromatographic assay for urate oxidase in human plasma. Anal Biochem. 1989;176:290–293. doi: 10.1016/0003-2697(89)90311-4.
    1. Guidance for Industry/Bioanalytic Method Validation
    1. Kelley WN, Rosenbloom FM, Henderson JF, Seegmiller JE. A specific enzyme defect in gout associated with overproduction of uric acid. Proc Natl Acad Sci USA. 1967;57:1735–1739. doi: 10.1073/pnas.57.6.1735.
    1. Hershfield MS, Chaffee S, Koro-Johnson L, Mary A, Smith AA, Short SA. Use of site-directed mutagenesis to enhance the epitope-shielding effect of covalent modification of proteins with polyethylene glycol. Proc Natl Acad Sci USA. 1991;88:7185–7189. doi: 10.1073/pnas.88.16.7185.
    1. Shoji A, Yamanaka H, Kamatani N. A retrospective study of the relationship between serum urate level and recurrent attacks of gouty arthritis: evidence for reduction of recurrent gouty arthritis with antihyperuricemic therapy. Arthritis Rheum. 2004;51:321–325. doi: 10.1002/art.20405.
    1. Ackerman AL, Kyritsis C, Tampe R, Cresswell P. Access of soluble antigens to the endoplasmic reticulum can explain cross-presentation by dendritic cells. Nat Immunol. 2005;6:107–113. doi: 10.1038/ni1147.
    1. Ackerman AL, Cresswell P. Cellular mechanisms governing cross-presentation of exogenous antigens. Nat Immunol. 2004;5:678–684. doi: 10.1038/ni1082.
    1. Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425:516–521. doi: 10.1038/nature01991.
    1. Gu Y, Hershfield MS, Cohen A. The danger within. N Engl J Med. 2004;350:2721–2722. doi: 10.1056/NEJM200406243502622.
    1. Agar NS, Sadrzadeh SMH, Hallaway PE, Eaton JW. Erythrocyte catalase. A somatic oxidant defense? J Clin Invest. 1986;77:319–321.
    1. Hershfield MS, Buckley RH, Greenberg ML, Melton AL, Schiff R, Hatem C, Kurtzberg J, Markert ML, Kobayashi RH, Kobayashi AL, et al. Treatment of adenosine deaminase deficiency with polyethylene glycol-modified adenosine deaminase. N Engl J Med. 1987;316:589–596.
    1. Chaffee S, Mary A, Stiehm ER, Girault D, Fischer A, Hershfield MS. IgG antibody response to polyethylene glycol-modified adenosine deaminase in patients with adenosine deaminase deficiency. J Clin Invest. 1992;89:1643–1651.
    1. Hershfield MS. Biochemistry and immunology of poly(ethylene glycol)-modified adenosine deaminase (PEG-ADA) In: Harris JM, Zalipsky S, editor. Poly(ethylene glycol) Chemistry and Biological Applications. Washington DC: American Chemical Society; 1997. pp. 145–154.
    1. Avramis VI, Sencer S, Periclou AP, Sather H, Bostrom BC, Cohen LJ, Ettinger AG, Ettinger LJ, Franklin J, Gaynon PS, et al. A randomized comparison of native Escherichia coli asparaginase and polyethylene glycol conjugated asparaginase for treatment of children with newly diagnosed standard-risk acute lymphoblastic leukemia: a Children's Cancer Group study. Blood. 2002;99:1986–1994. doi: 10.1182/blood.V99.6.1986.
    1. Hawkins DS, Park JR, Thomson BG, Felgenhauer JL, Holcenberg JS, Panosyan EH, Avramis VI. Asparaginase pharmacokinetics after intensive polyethylene glycol-conjugated L-asparaginase therapy for children with relapsed acute lymphoblastic leukemia. Clin Cancer Res. 2004;10:5335–5341. doi: 10.1158/1078-0432.CCR-04-0222.
    1. Caliceti P, Schiavon O, Veronese FM. Immunological properties of uricase conjugated to neutral soluble polymers. Bioconjug Chem. 2001;12:515–522. doi: 10.1021/bc000119x.
    1. Richter AW, Akerblom E. Antibodies against polyethylene glycol produced in animals by immunization with monomethoxy polyethylene glycol modified proteins. Int Arch Allergy Appl Immunol. 1983;70:124–131.
    1. Cheng TL, Wu PY, Wu MF, Chern JW, Roffler SR. Accelerated clearance of polyethylene glycol-modified proteins by anti-polyethylene glycol IgM. Bioconjug Chem. 1999;10:520–528. doi: 10.1021/bc980143z.
    1. Richter AW, Akerblom E. Polyethylene glycol reactive antibodies in man: titer distribution in allergic patients treated with monomethoxy polyethylene glycol modified allergens or placebo, and in healthy blood donors. Int Arch Allergy Appl Immunol. 1984;74:36–39.
    1. Wilkinson I, Jackson CJ, Lang GM, Holford-Strevens V, Sehon AH. Tolerance induction in mice by conjugates of monoclonal immunoglobulins and monomethoxypolyethylene glycol. Transfer of tolerance by T cells and by T cell extracts. J Immunol. 1987;139:326–331.
    1. Chen Y, Takata M, Maiti PK, Rector ES, Sehon AH. Characterization of suppressor T cell clones derived from a mouse tolerized with conjugates of ovalbumin and monomethoxypolyethylene glycol. Cell Immunol. 1992;142:16–27. doi: 10.1016/0008-8749(92)90265-Q.
    1. Lee WY, Sehon AH. Suppression of reaginic antibodies with modified allergens. I. Reduction in allergenicity of protein allergens by conjugation to polyethylene glycol. Int Arch Allergy Appl Immunol. 1978;56:159–170.

Source: PubMed

3
Abonner