Population pharmacokinetics of sifalimumab, an investigational anti-interferon-α monoclonal antibody, in systemic lupus erythematosus

Rajesh Narwal, Lorin K Roskos, Gabriel J Robbie, Rajesh Narwal, Lorin K Roskos, Gabriel J Robbie

Abstract

Background and objectives: Sifalimumab is a fully human immunoglobulin G1κ monoclonal antibody that binds to and neutralizes a majority of the subtypes of human interferon-α. Sifalimumab is being evaluated as a treatment for systemic lupus erythematosus (SLE). The primary objectives of this analysis were (a) to develop a population pharmacokinetic model for sifalimumab in SLE; (b) to identify and quantitate the impact of patient/disease characteristics on pharmacokinetic variability; and (c) to evaluate fixed versus body weight (WT)-based dosing regimens.

Methods: Sifalimumab serum concentration-time data were collected from a phase Ib study (MI-CP152) designed to evaluate the safety and tolerability of sifalimumab in adult patients with SLE. Sifalimumab was administered every 14 days as a 30- to 60-minute intravenous infusion with escalating doses of 0.3, 1.0, 3.0, and 10 mg/kg and serum concentrations were collected over 350 days. A total of 120 patients provided evaluable pharmacokinetic data with a total of 2,370 serum concentrations. Sifalimumab serum concentrations were determined using a validated colorimetric enzyme-linked immunosorbent assay (ELISA) with a lower limit of quantitation of 1.25 μg/mL. Population pharmacokinetic modeling of sifalimumab was performed using a non-linear mixed effects modeling approach with NONMEM VII software. Impact of patient demographics, clinical indices, and biomarkers on pharmacokinetic parameters were explored using a stepwise forward selection and backward elimination approach. The appropriateness of the final model was tested using visual predictive check (VPC). The impact of body WT-based and fixed dosing of sifalimumab was evaluated using a simulation approach. The final population model was utilized for phase IIb dosing projections.

Results: Sifalimumab pharmacokinetics were best described using a two-compartment linear model with first order elimination. Following intravenous dosing, the typical clearance (CL) and central volume of distribution (V 1) were estimated to be 176 mL/day and 2.9 L, respectively. The estimates (coefficient of variation) of between-subject variability for CL and V 1 were 28 and 31 %, respectively. Patient baseline body WT, interferon gene signature from 21 genes, steroid use, and sifalimumab dose were identified as significant covariates for CL, whereas only baseline body WT was a significant covariate for V 1 and peripheral volume of distribution (V 2). Although the above-mentioned covariates were statistically significant, they did not explain variability in pharmacokinetic parameters to any relevant extent (<7 %). Thus, no dosing adjustments are necessary. VPC confirmed good predictability of the final population pharmacokinetic model. Simulation results demonstrate that both fixed and body WT-based dosing regimens yield similar median steady state concentrations and overall variability. Fixed sifalimumab doses of 200, 600, and 1,200 mg monthly (with a loading dose at Day 14) were selected for a phase IIb clinical trial.

Conclusion: A two-compartment population pharmacokinetic model adequately described sifalimumab pharmacokinetics. The estimated typical pharmacokinetic parameters were similar to other monoclonal antibodies without target mediated elimination. Although the population pharmacokinetic analysis identified some statistically significant covariates, they explained <7 % between-subject variability in pharmacokinetic parameters indicating that these covariates are not clinically relevant. The population pharmacokinetic analysis also demonstrated the feasibility of switching to fixed doses in phase IIb clinical trials of sifalimumab.

Trial registration: ClinicalTrials.gov NCT00482989.

Figures

Fig. 1
Fig. 1
Final model goodness-of-fit plots for sifalimumab serum concentrations. The thin black line (diagonal and horizontal) and thick black line represent line of unity and loess fit, respectively
Fig. 2
Fig. 2
Visual predictive check for sifalimumab serum concentrations. Observed median (solid red line), 5th and 9th percentiles (dashed red lines) and corresponding simulation-based prediction intervals (solid and dashed black lines) and 95 % confidence intervals (green and beige shaded areas)
Fig. 3
Fig. 3
Similarity of predicted pharmacokinetic profiles (median, 5th and 95th percentiles) following fixed (200 mg every 14 days) and body weight-based (3 mg/kg every 14 days) dosing of sifalimumab. Q14D every 14 days, WT body weight
Fig. 4
Fig. 4
Predicted serum concentrations following 200, 600, and 1,200 mg monthly intravenous dosing of sifalimumab (with single loading dose at Day 14)

References

    1. American College of Rheumatology Ad Hoc Committee on Systemic Lupus Erythematosus Guidelines. Guidelines for referral and management of systemic lupus erythematosus in adults. Arthritis Rheum. 1999;42:1785–96.
    1. Petri M. Long-term outcomes in lupus. Am J Managed Care. 2001;7:S480-5.
    1. Lawrence RC, Helmick CG, Arnett FC, et al. Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States. Arthritis Rheum. 1998;41(5):778–799. doi: 10.1002/1529-0131(199805)41:5<778::AID-ART4>;2-V.
    1. Cooper GS, Dooley MA, Treadwell EL, et al. Hormonal, environmental, and infectious risk factors for developing systemic lupus erythematosus. Arthritis Rheum. 1998;41(10):1714–1724. doi: 10.1002/1529-0131(199810)41:10<1714::AID-ART3>;2-U.
    1. Lahita RG. The role of sex hormones in systemic lupus erythematosus. Curr Opin Rheumatol. 1999;11(5):352–356. doi: 10.1097/00002281-199909000-00005.
    1. Wallace D. Advances in the management of systemic lupus erythematosus. Bull Rheum Dis. 2004;51(11):1–7.
    1. Urowitz MB, Bookman AA, Koehler BE, et al. The bimodal mortality pattern of systemic lupus erythematosus. Am J Med. 1976;60(2):221–225. doi: 10.1016/0002-9343(76)90431-9.
    1. Theofilopoulos AN, Baccala R, Beutler B, et al. Type I interferons (α/β) in immunity and autoimmunity. Immunol Rev. 2005;204:9–26. doi: 10.1111/j.0105-2896.2005.00252.x.
    1. Crow M. Interferon-α: a new target for therapy in systemic lupus erythematosus? Arthritis Rheum. 2003;48:2396–2401. doi: 10.1002/art.11226.
    1. Kirou KA, Lee C, George S, et al. Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum. 2005;52(5):1491–1503. doi: 10.1002/art.21031.
    1. Dall’era MC, Cardarelli PM, Preston BT, et al. Type I interferon correlates with clinical and serologic manifestations of systemic lupus erythematosus. Ann Rheum Dis. 2005;64:1692–1697. doi: 10.1136/ard.2004.033753.
    1. Baechler EC, Batliwalla FM, Karypis G, et al. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci. 2003;100(5):2610–2615. doi: 10.1073/pnas.0337679100.
    1. Baechler EC, Gregersen PK, Behrens TW. The emerging role of interferon systemic lupus erythematosus. Curr Opin Immunol. 2004;16(6):801–807. doi: 10.1016/j.coi.2004.09.014.
    1. Bengtsson AA, Sturfelt G, Truedsson L, et al. Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies. Lupus. 2000;9:664–671. doi: 10.1191/096120300674499064.
    1. Santiago-Raber M-L, Baccala R, Haraldsson KM, et al. Type I interferon receptor deficiency reduces lupus-like disease in NZB mice. J Exp Med. 2003;197:777–788. doi: 10.1084/jem.20021996.
    1. Niewold TB, Swedler WI. Systemic lupus erythematosus arising during interferon-alpha therapy for cryoglobulinemic vasculitis associated with hepatitis C. Clin Rheumatol. 2005;24(2):178–181. doi: 10.1007/s10067-004-1024-2.
    1. Ioannou Y, Isenberg DA. Current evidence for the induction of autoimmune rheumatic manifestations by cytokine therapy. Arthritis Rheum. 2000;43(7):1431–1442. doi: 10.1002/1529-0131(200007)43:7<1431::AID-ANR3>;2-E.
    1. Bennett L, Palucka AK, Arce E, et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003;197:711–723. doi: 10.1084/jem.20021553.
    1. Kirou KA, Lee C, George S, et al. Coordinate overexpression of interferon-alpha-induced genes in systemic lupus erythematosus. Arthritis Rheum. 2004;50(12):3958–3967. doi: 10.1002/art.20798.
    1. Feng X, Wu H, Grossman JM, et al. Association of increased interferon-inducible gene expression with disease activity and lupus nephritis in patients with systemic lupus erythematosus. Arthritis Rheum. 2006;54(9):2951–2962. doi: 10.1002/art.22044.
    1. Blomberg S, Eloranta ML, Cederblad B, et al. Presence of cutaneous interferon-alpha producing cells in patients with systemic lupus erythematosus. Lupus. 2001;10(7):484–490. doi: 10.1191/096120301678416042.
    1. Farkas L, Beiske K, Lund-Johansen F, et al. Plasmacytoid dendritic cells (natural interferon-alpha/beta-producing cells) accumulate in cutaneous lupus erythematosus lesions. Am J Pathol. 2001;159(1):237–243. doi: 10.1016/S0002-9440(10)61689-6.
    1. Wang Y. Derivation of various NONMEM estimation methods. J Pharmacokinet Pharmacodyn. 2007;34(5):575–593. doi: 10.1007/s10928-007-9060-6.
    1. Ette EI, Williams PJ. Population pharmacokinetics I: background, concepts, and models. Ann Pharmacother. 2004;38(10):1702–1706. doi: 10.1345/aph.1D374.
    1. Bonate PL. Pharmacokinetic-pharmacodynamic modeling and simulation. New York: Springer; 2006.
    1. Boeckmann AJ, Scheiner LB, Beal SL. NONMEM user guide: part V. San Francisco: NONMEM Project Group; 1994.
    1. Mandema JW, Verotta D, Sheiner LB. Building population pharmacokinetic–pharmacodynamic models. I. Models for covariate effects. J Pharmacokinet Biopharm. 1992;20(5):511–528. doi: 10.1007/BF01061469.
    1. Sheiner LB, Beal SL. Evaluation of methods for estimating population pharmacokinetic parameters. III. Monoexponential model: routine clinical pharmacokinetic data. J Pharmacokinet Biopharm. 1983;11(3):303–319. doi: 10.1007/BF01061870.
    1. Lindbom L, Pihlgren P, Jonsson EN. PsN-Toolkit–a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed. 2005;79(3):241–257. doi: 10.1016/j.cmpb.2005.04.005.
    1. Lindbom L, Ribbing J, Jonsson EN. Perl-speaks-NONMEM (PsN)—a Perl module for NONMEM related programming. Comput Methods Programs Biomed. 2004;75(2):85–94. doi: 10.1016/j.cmpb.2003.11.003.
    1. Jonsson EN, Karlsson MO. Xpose–an S-PLUS based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput Methods Programs Biomed. 1999;58(1):51–64. doi: 10.1016/S0169-2607(98)00067-4.
    1. Holford N. The visual predictive check: superiority to standard diagnostic (Rorschach) plots. PAGE. 2005;14:738.
    1. Tabrizi MA, Tseng CM, Roskos LK. Elimination mechanisms of therapeutic monoclonal antibodies. Drug Discov Today. 2006;11(1–2):81–88. doi: 10.1016/S1359-6446(05)03638-X.
    1. Wang W, Wang EQ, Balthasar JP. Monoclonal antibody pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther. 2008;84(5):548–558. doi: 10.1038/clpt.2008.170.
    1. Mould DR, Green B. Pharmacokinetics and pharmacodynamics of monoclonal antibodies: concepts and lessons for drug development. BioDrugs. 2010;24(1):23–39. doi: 10.2165/11530560-000000000-00000.
    1. Tabrizi M, Wang B, Lu H, et al. Population pharmacokinetic evaluation of a fully human IgG monoclonal antibody in patients with inflammatory diseases. Inflamm Allergy Drug Targets. 2010;9(4):229–237. doi: 10.2174/187152810793358796.
    1. Singh D, Kane B, Molfino NA, et al. A phase 1 study evaluating the pharmacokinetics, safety and tolerability of repeat dosing with a human IL-13 antibody (CAT-354) in subjects with asthma. BMC Pulm Med. 2010;10(1):3. doi: 10.1186/1471-2466-10-3.
    1. Oh CK, Faggioni R, Jin F, et al. An open-label, single-dose bioavailability study of the pharmacokinetics of CAT-354 after subcutaneous and intravenous administration in healthy males. Br J Clin Pharmacol. 2010;69(6):645–655. doi: 10.1111/j.1365-2125.2010.03647.x.
    1. Lu JF, Bruno R, Eppler S, et al. Clinical pharmacokinetics of bevacizumab in patients with solid tumors. Cancer Chemother Pharmacol. 2008;62(5):779–786. doi: 10.1007/s00280-007-0664-8.
    1. Ng CM, Bruno R, Combs D, et al. Population pharmacokinetics of rituximab (anti-CD20 monoclonal antibody) in rheumatoid arthritis patients during a phase II clinical trial. J Clin Pharmacol. 2005;45(7):792–801. doi: 10.1177/0091270005277075.
    1. Dirks NL, Nolting A, Kovar A, et al. Population pharmacokinetics of cetuximab in patients with squamous cell carcinoma of the head and neck. J Clin Pharmacol. 2008;48(3):267–278. doi: 10.1177/0091270007313393.
    1. Mould DR, Holford NH, Schellens JH, et al. Population pharmacokinetic and adverse event analysis of topotecan in patients with solid tumors. Clin Pharmacol Ther. 2002;71(5):334–348. doi: 10.1067/mcp.2002.123553.
    1. White B, Leon F, White W, et al. Two first-in-human, open-label, phase I dose-escalation safety trials of MEDI-528, a monoclonal antibody against interleukin-9, in healthy adult volunteers. Clin Therap. 2009;31(4):728–740. doi: 10.1016/j.clinthera.2009.04.019.
    1. Narwal R, Akhlaghi F, Asberg A, et al. Development of a population pharmacokinetic model for atorvastatin acid and its lactone metabolite. Clin Pharmacokinet. 2010;49(10):693–702. doi: 10.2165/11535980-000000000-00000.
    1. Holford NH. A size standard for pharmacokinetics. Clin Pharmacokinet. 1996;30(5):329–332. doi: 10.2165/00003088-199630050-00001.
    1. Petri M, Wallace DJ, Spindler A, et al. Sifalimumab, a human anti-interferon-α monoclonal antibody, in systemic lupus erythematosus. Arthritis Rheum. 2013;65(4):1011–1021. doi: 10.1002/art.37824.
    1. Ng CM, Lum BL, Gimenez V, et al. Rationale for fixed dosing of pertuzumab in cancer patients based on population pharmacokinetic analysis. Pharm Res. 2006;23(6):1275–1284. doi: 10.1007/s11095-006-0205-x.
    1. Wang DD, Zhang S, Zhao H, et al. Fixed dosing versus body size-based dosing of monoclonal antibodies in adult clinical trials. J Clin Pharmacol. 2009;49(9):1012–1024. doi: 10.1177/0091270009337512.
    1. Zhang S, Shi R, Li C, et al. Fixed dosing versus body size-based dosing of therapeutic peptides and proteins in adults. J Clin Pharmacol. 2012;52(1):18–28. doi: 10.1177/0091270010388648.

Source: PubMed

3
購読する