Preclinical pharmacokinetics, interspecies scaling, and tissue distribution of humanized monoclonal anti-IL-13 antibodies with different IL-13 neutralization mechanisms

https://doi.org/10.1016/j.intimp.2007.12.004Get rights and content

Abstract

Numerous animal and in vitro studies suggest that neutralization of IL-13 is an attractive approach for therapeutic intervention in asthma. In this paper we describe preclinical pharmacokinetics (PK), interspecies scaling, and biodistribution of two humanized anti-IL-13 IgG1 monoclonal antibodies, Ab-01 and Ab-02, with different IL-13 neutralization mechanisms. PK parameters of Ab-01 and Ab-02 following IV or SC dosage to mouse, rat, cynomolgus monkey, and sheep, were similar. After IV administration, the elimination of anti-IL-13 antibodies was slow in all species tested and the serum clearance ranged from 0.13 mL/h/kg in monkeys to 0.81 mL/h/kg in mice. Both anti-IL-13 antibodies appeared to be confined primarily to the vascular space, as volume of distribution was relatively small (< 120 mL/kg) in all species and tissue-to-serum concentration ratios (in mice and rats) were low (< 0.5) in the tissues examined. The elimination half-life ranged from 3–6 days in mice to 14–17 days in monkey and sheep. In monkeys, PK parameters appeared to be approximately linear in the 1–100 mg/kg dose range. Following SC administration, the bioavailability of anti-IL-13 antibodies was 60–90% in all species tested. PK profile of Ab-02 in the model of acute airway inflammation (induced by Ascaris challenge) was, in general, similar to that in unchallenged monkeys; however, volume of distribution and clearance tended to decrease in Ascaris-challenged animals. Allometric scaling suggested that anti-IL-13 antibodies would likely to have a favorable PK profile, such as slow clearance and long terminal half-life, following IV or SC administration to humans.

Introduction

Numerous animal and in vitro studies support a major role for Th2 cytokine, interleukin-13 (IL-13), in asthma pathogenesis [1], [2], [3], [4]. In addition, there are consistent correlations between polymorphism in the IL-13 gene and asthma susceptibility in humans [5]. Neutralization of IL-13 with anti-IL-13 antibodies or with IL-13 receptor α2/Fc fusion protein (IL-13Rα2-Fc) prevents airway hyperresponsiveness and other asthmatic changes in mice [3], [6], [7], [8], [9], [10], sheep [11], and cynomolgus monkeys [12], [13]. Thus, neutralization of IL-13 is an attractive approach for therapeutic intervention in asthma.

IL-13 interacts with cells through a receptor complex consisting of IL-13 receptor α1 (IL-13αR1) and interleukin-4 receptor alpha (IL-4Rα) subunits [1], [2]. IL-13 first undergoes a low affinity interaction with IL-13Rα1, which recruits IL-4Rα to form an active signaling complex with high affinity for IL-13, leading to phosphorylation of STAT6 and downstream cellular activation events.

Ab-01 and Ab-02 are the humanized anti-IL-13 monoclonal antibodies that have been shown to be efficacious in a cynomolgus monkey model of acute airway inflammation induced by lung segmental challenge with Ascaris antigen [12], [13]. Although Ab-01 and Ab-02 have comparable inhibitory activity in vitro and comparable in vivo efficacy, these antibodies neutralize IL-13 bioactivity by different mechanisms [12], [13]. Ab-01 blocks the interaction of IL-13 with the IL-4Rα chain (but not the IL-13Rα1/IL-13Rα2 chains), while Ab-02 blocks binding of IL-13Rα1/IL-13Rα2 and IL-13. Since anti-IL-13 antibodies bind to a soluble ligand, the potential for a direct Fc-mediated activity is low. However, by virtue of its epitope Ab-01 has the potential for an indirect Fc-mediated effector activity on IL-13Rα1/IL-4Rα-expressing cells, while Ab-02 does not. Therefore, two point mutations that reduce potential effector activity were introduced into the lower hinge region of the IgG1Fc tail of both antibodies [13]. Neither Ab-01 nor Ab-02 binds to rodent IL-13. Ab-01 cross-reacts with monkey IL-13 (KD of 1.4 × 10 11 M) and with sheep IL-13, albeit with significantly lower affinity compared to human IL-13, while Ab-02 cross-reacts only with monkey IL-13 (KD of 9.2 × 10 11 M) [13].

To support development of anti-IL-13 antibodies, pharmacokinetic studies were conducted in mice, rats, sheep, and cynomolgus monkeys after IV and SC administration. Tissue distribution of [125I]Ab-01 and [125I]Ab-02 was studied in mice and rats, respectively. The pharmacokinetic data in animals were used to predict the disposition of anti-IL13 in humans using allometric scaling. In addition, we obtained PK profiles in Ascaris-challenged monkeys and compared to those in normal unchallenged animals.

Section snippets

Test articles

Mouse Ab-01 was cloned from BALB/c mice immunized with human IL-13. The humanized version of this antibody, Ab-01 (IgG1, k), was generated at Wyeth Research (Cambridge, MA) [11], [12]. Mouse Ab-02 was cloned from BALB/c mice immunized with the N-terminal 19 amino acids of nonhuman primate IL-13. The humanized version of this antibody, Ab-02 (IgG1, k), was generated at Wyeth Research (Cambridge, MA) [13]. Carimune NH immune globulin intravenous (human IVIG; ZLB Bioplasma Inc., Berne,

PK summary of Ab-01 and Ab-02 anti-IL-13 antibodies

The pharmacokinetic profiles of Ab-01 were determined in mice, rats, monkeys and sheep after single 1 or 2 mg/kg IV administration (Table 1). In general, the elimination of Ab-01 was slow after IV administration and the serum clearance (CL) ranged from 0.13 mL/h/kg in sheep to 0.81 mL/h/kg in mice. The steady-state volume of distribution (Vdss) was also small (< 120 mL/kg) in all species tested, suggesting that the protein was mainly confined in the vascular circulation. The elimination

Discussion

In this report we describe preclinical PK and biodistribution of two anti-IL-13 humanized IgG1 antibodies that have different IL-13 neutralization mechanisms. In general, PK parameters of Ab-01 and Ab-02 following IV or SC dosage were similar in all species tested.

Both anti-IL-13 antibodies (Ab-01 and Ab-02) were eliminated slowly following IV administration to rodents, which are non-binding species, as Ab-01 and Ab-02 do not bind to mouse or rat IL-13. PK of anti-IL-13 antibodies was also

Acknowledgements

We thank Tamera Ashworth, Nicole Duriga, and David Defranco for technical help with BioAnalytical aspects of the studies; Macy Jin, Xiang-Yang Tan, and Khetemenee Lam for technical help with generation and purification of the test articles; Andrea Bree, Franklin Schlerman, and Michael Wadanoli for help with Ascaris challenge studies in monkeys; Vikram Patel, Aqel Abu-Qare, Mauricio Leal, Stan Spence, Garvin Warner, and Tom Brown for scientific discussions on data interpretation.

References (17)

  • A.L. Andrews et al.

    Kinetic analysis of the interleukin-13 receptor complex

    J Biol Chem

    (2002)
  • A. Bree et al.

    IL-13 blockade reduces lung inflammation after Ascaris suum challenge in cynomolgus monkeys

    J Allergy Clin Immunol

    (2007)
  • D.B. Corry et al.

    Biology and therapeutic potential of the interleukin-4/interleukin-13 signaling pathway in asthma

    Am J Respir Medicine

    (2002)
  • G. Grunig et al.

    Requirement for IL-13 independently of IL-4 in experimental asthma

    Science

    (1998)
  • M. Wills-Karp

    Interleukin-13 in asthma pathogenesis

    Curr Allergy Asthma Rep

    (2004)
  • D. Vercelli

    Genetics of IL-13 and functional relevance of IL-13 variants

    Curr Opin Allergy Clin Immunol

    (2002)
  • R.K. Kumar et al.

    Effects of anticytokine therapy in a mouse model of chronic asthma

    Am J Respir Crit Care Med

    (2004)
  • G. Yang et al.

    Therapeutic dosing with anti-interleukin-13 monoclonal antibody inhibits asthma progression in mice

    J Pharmacol Exp Ther

    (2005)
There are more references available in the full text version of this article.

Cited by (40)

  • Whole-Body Pharmacokinetics of Antibody in Mice Determined using Enzyme-Linked Immunosorbent Assay and Derivation of Tissue Interstitial Concentrations

    2021, Journal of Pharmaceutical Sciences
    Citation Excerpt :

    Since most mAbs act in the tissue interstitial space, it is important to accurately measure/predict the concentration of these molecules in tissues, to better understand their exposure-response relationships. Tissue distribution of mAbs is typically measured in preclinical species, and predicted in humans using allometric scaling.2-7 or physiologically-based pharmacokinetic (PBPK) models.8,9

  • Pharmacokinetics of anti-IL17A and anti-IL22 peptide-antibody bispecific genetic fusions in mice

    2014, International Immunopharmacology
    Citation Excerpt :

    The serum concentration profiles after a single IV dose of peptide/Ab genetic fusions were reminiscent of that for a human or mouse IgG, with a relatively rapid distribution phase, followed by a slow elimination phase. However, elimination of genetic fusions in the terminal phase appeared somewhat faster than that of a typical human IgG1 in mice, which in general have a t1/2 between 5 days to 3 weeks [11–13]. For comparison, the t1/2 of an anti-IL-22 human IgG1 was 2–3 weeks in mice, based on various studies utilizing different dosing regimens (data not shown).

  • Pharmacokinetics, biocompatibility and bioavailability of a controlled release monoclonal antibody formulation

    2013, Journal of Controlled Release
    Citation Excerpt :

    The human IgG1 does not bind to its corresponding target in rat, and, therefore, antigen-binding does not impact the pharmacokinetic behavior of the antibody. The obtained pharmacokinetic data was comparable to that of previous studies of subcutaneously delivered humanized monoclonal antibodies in rodents [27–29]. It is known that the used antibody displays linear and dose proportional kinetic behavior, which is typical for non-binding, intravenously administered human IgGs in rodent.

  • Airway disease: The use of large animal models for drug discovery

    2011, Pulmonary Pharmacology and Therapeutics
    Citation Excerpt :

    There are a number of lines of evidence that support that the Th2 cytokine IL-13 plays an important role in asthma pathophysiology and therefore the neutralisation of IL-13 is an attractive approach for intervention in asthma [14]. Preclinical testing of the pharmacokinetics of two humanised monoclonal antibodies against IL-13 have been performed in a number of species including sheep [15,16]. Allometric scaling suggested that anti-IL-13 antibodies would likely have a favourable pharmacokinetic profile in humans.

  • Emerging Role of Interleukins for the Assessment and Treatment of Liver Diseases

    2022, Endocrine, Metabolic and Immune Disorders - Drug Targets
View all citing articles on Scopus

Disclosures and conflict of interest statement: All authors are employed by or are consultants for Wyeth, Inc.

View full text