Tolerability in man following inhalation dosing of the selective TLR7 agonist, AZD8848

Stephen Delaney, Mark Biffen, Justine Maltby, John Bell, Sara Asimus, Ajay Aggarwal, Maarten Kraan, David Keeling, Stephen Delaney, Mark Biffen, Justine Maltby, John Bell, Sara Asimus, Ajay Aggarwal, Maarten Kraan, David Keeling

Abstract

Background: Many patients with asthma have a T-helper type 2 (Th2) driven inflammation of the lung, whereas toll-like receptor 7 (TLR7) agonists, by inducing type I interferons, inhibit Th2 responses. In man, oral or parenteral TLR7 agonists can induce influenza-like symptoms through systemic induction of type I interferons. Design of a TLR7 agonist that is only active in the lung could reduce the risk of side effects and offer a new means for treating asthma. We developed a TLR7 agonist antedrug, AZD8848, to determine its local and systemic effects in preclinical models and man.

Methods: In vitro cellular potencies for the TLR7 antedrug agonist, AZD8848, were determined along with pharmacokinetics and efficacy in a rat allergy model. Sputum and blood biomarkers were measured in single ascending and multiple ascending dose clinical studies following inhalation delivery of AZD8848 and tolerability assessed.

Results: AZD8848 was potent in cellular assays and pharmacokinetics confirmed lack of systemic exposure to AZD8848. Weekly lung dosing in an animal model showed efficacy 26 days beyond the final dose. In healthy volunteers, AZD8848 was initially well tolerated with target engagement being demonstrated by induction of CXCL10 in sputum. A second inhaled dose, given 1 week later, amplified the systemic interferon signal in more than half the participants and resulted in significant influenza-like symptoms.

Conclusions: The antedrug design restricted the direct actions of AZD8848 to the lung. However, the type I interferon induced locally by TLR7 spilled over systemically, limiting the utility of this inhaled antedrug approach.

Trial registration number: NCT01560234, NCT01818869.

Keywords: Allergic lung disease; Asthma; Asthma Mechanisms.

Figures

Figure 1
Figure 1
Structure of AZD8848 and potency in recombinant human TLR human embryonic kidney (HEK) reporter cells. (A) The chemical structure of AZD8848 and (B) the activity of AZD8848 and its acid metabolite, AZ12432045, in HEK cells stably expressing human toll-like receptor 7 (TLR7). Activity was calculated from the release of secretory alkaline phosphatase, whose expression was linked to an NF-κB promoter, with the data normalised to the maximal induction produced by the reference TLR7 agonist, R848 (set to 100%). Incubation of cells with compound was for 24 h. Data show the mean±SEM from nine separate experiments (AZD8848) and from six separate experiments (AZ12432045).
Figure 2
Figure 2
Pharmacokinetics and efficacy of AZD8848 in the Brown Norway rat. Brown Norway rats were dosed AZD8848 at 0.3 mg/kg intratracheally in a volume of 150 µL and blood samples taken at the indicated time points. Nine rats were dosed and at each time point samples from at least two animals were taken. Data for the plasma levels of compound (A) are presented as the mean of the determinations±SD or range and are representative of two such determinations. Following intratracheal administration, lung levels of AZD8848 and its acid metabolite were determined (B). At each time point, apart from the first, two animals were sacrificed and data are represented as the mean±range. The limits of detection for AZD8848 and AZ12432045 were 0.2 and 2 nmol/L, respectively. For the purposes of determining a representative value for each group of animals, any sample where the compound was undetectable was arbitrarily assigned a value of half that of the limit of detection. Data points including only such values are shown with a #. OVA-sensitised rats were dosed intratracheally with AZD8848, at the indicated doses (shown in brackets), 24 h prior to and 24 h after OVA challenge (C). Animals were sacrificed 48 h after the OVA challenge and the numbers of eosinophils or level of interleukin 13 in the BAL determined. The following rat numbers, in brackets, were used for each data point: sensitised only (5), intratracheally dosed (9). Data are representative of three studies. In further studies 14 days after OVA-sensitisation, Brown Norway rats were dosed topically with AZD8848 (1 mg/kg) or budesonide (1 mg/kg) via the intratracheal route on eight occasions at weekly intervals. Twenty-six days after the final dose of compound, animals were challenged with aerosolised OVA and 48 h later eosinophil number and IL-13 levels determined in the BAL (D). Ten animals were used for each data point. The effect of acute treatment with compound (24 h before and 24 h after OVA challenge) was also determined in groups of eight animals. The number of eosinophils or level of IL-13 in the BAL per animal was averaged across each treatment group and the result expressed as mean±SEM. All study groups were compared, with their respective OVA-stimulated control dosed with vehicle, using the Mann-Whitney U test; *p

Figure 3

Pharmacokinetics and biomarker induction following…

Figure 3

Pharmacokinetics and biomarker induction following a single inhaled dose of AZD8848 in man.(A)…

Figure 3
Pharmacokinetics and biomarker induction following a single inhaled dose of AZD8848 in man.(A) Plasma concentration-time profiles of AZ12432045, the acid metabolite of AZD8848, following a single predicted lung-deposited dose of AZD8848 as indicated in the key. The predicted lung-deposited dose was assumed to be 63% of the dose delivered from the nebuliser. During sample preparation, any trace amounts of AZD8848 were hydrolysed by esterases in the plasma. All cohorts were administered by nebulisation taken in 10 breaths as described in Methods section. No data are shown for cohort 1 (0.15 µg) as levels of AZ12432045 were below the level of detection. Values shown are geometric means (±SD) from 3 to 5 individuals in each cohort. (B) CXCL10 was determined in induced sputum (black) and in plasma (grey) of healthy volunteers for the indicated predicted lung-deposited dose of AZD8848. The fold change at 24 h postdose from baseline for individual participants is shown. Geometric mean is shown in each cohort and significance relative to placebo (p

Figure 4

Plasma CXCL10 and blood lymphocyte…

Figure 4

Plasma CXCL10 and blood lymphocyte counts following a second inhaled dose of AZD8848.…

Figure 4
Plasma CXCL10 and blood lymphocyte counts following a second inhaled dose of AZD8848. CXCL10 in plasma (A) and lymphocytes in blood (B) were determined in healthy volunteers receiving two 30 μg doses of AZD8848 1 week apart. Data from individual participants are shown as separate lines, treatment as solid lines, and placebo as broken lines. Arrows indicate individual participants who experienced severe (black) or moderate (grey) influenza-like symptoms in the preceding period. Between-treatment and intrasubject comparisons for CXCL10 levels are described in the text.

Figure 5

Monitoring the induction of pro-inflammatory…

Figure 5

Monitoring the induction of pro-inflammatory mediators and toll-like receptors (TLRs) following inhalation of…

Figure 5
Monitoring the induction of pro-inflammatory mediators and toll-like receptors (TLRs) following inhalation of AZD8848. The mediators (A) C reactive protein (CRP), (B) interferon γ (IFNγ), (C) interleukin 8 (IL-8) and (D) tumour necrosis factor α (TNFα) were measured in plasma as described in Methods section in healthy volunteers receiving two 30 μg doses of AZD8848 1 week apart. Data from individual participants are shown as separate lines, treatment as solid lines, and placebo as broken lines. Arrows indicate individual participants who experienced severe (black) or moderate (grey) influenza-like symptoms in the preceding period. Changes in (E) TLR7 and (F) TLR9 RNA levels in blood were determined in healthy volunteers receiving a 30 μg dose of AZD8848 and samples were taken at the time points indicated. Data from individual participants are shown, those receiving placebo as a broken line, those receiving AZD8848 as a solid line. In view of the small numbers of participants, no statistical testing was performed between treatment groups.
Similar articles
Cited by
  • Bacterial vesicles block viral replication in macrophages via TLR4-TRIF-axis.
    Bierwagen J, Wiegand M, Laakmann K, Danov O, Limburg H, Herbel SM, Heimerl T, Dorna J, Jonigk D, Preußer C, Bertrams W, Braun A, Sewald K, Schulte LN, Bauer S, Pogge von Strandmann E, Böttcher-Friebertshäuser E, Schmeck B, Jung AL. Bierwagen J, et al. Cell Commun Signal. 2023 Mar 28;21(1):65. doi: 10.1186/s12964-023-01086-4. Cell Commun Signal. 2023. PMID: 36978183 Free PMC article.
  • Advancing Adjuvants for Mycobacterium tuberculosis Therapeutics.
    Enriquez AB, Izzo A, Miller SM, Stewart EL, Mahon RN, Frank DJ, Evans JT, Rengarajan J, Triccas JA. Enriquez AB, et al. Front Immunol. 2021 Oct 25;12:740117. doi: 10.3389/fimmu.2021.740117. eCollection 2021. Front Immunol. 2021. PMID: 34759923 Free PMC article. Review.
  • TLR2-mediated activation of innate responses in the upper airways confers antiviral protection of the lungs.
    Deliyannis G, Wong CY, McQuilten HA, Bachem A, Clarke M, Jia X, Horrocks K, Zeng W, Girkin J, Scott NE, Londrigan SL, Reading PC, Bartlett NW, Kedzierska K, Brown LE, Mercuri F, Demaison C, Jackson DC, Chua BY. Deliyannis G, et al. JCI Insight. 2021 Mar 8;6(5):e140267. doi: 10.1172/jci.insight.140267. JCI Insight. 2021. PMID: 33561017 Free PMC article.
  • Toll-Like Receptor Agonists as Adjuvants for Allergen Immunotherapy.
    Kirtland ME, Tsitoura DC, Durham SR, Shamji MH. Kirtland ME, et al. Front Immunol. 2020 Nov 12;11:599083. doi: 10.3389/fimmu.2020.599083. eCollection 2020. Front Immunol. 2020. PMID: 33281825 Free PMC article. Review.
  • EAACI Allergen Immunotherapy User's Guide.
    Alvaro-Lozano M, Akdis CA, Akdis M, Alviani C, Angier E, Arasi S, Arzt-Gradwohl L, Barber D, Bazire R, Cavkaytar O, Comberiati P, Dramburg S, Durham SR, Eifan AO, Forchert L, Halken S, Kirtland M, Kucuksezer UC, Layhadi JA, Matricardi PM, Muraro A, Ozdemir C, Pajno GB, Pfaar O, Potapova E, Riggioni C, Roberts G, Rodríguez Del Río P, Shamji MH, Sturm GJ, Vazquez-Ortiz M. Alvaro-Lozano M, et al. Pediatr Allergy Immunol. 2020 May;31 Suppl 25(Suppl 25):1-101. doi: 10.1111/pai.13189. Pediatr Allergy Immunol. 2020. PMID: 32436290 Free PMC article. Review.
References
    1. Pearce N, Ait-Khaled N, Beasley R et al. . Worldwide trends in the prevalence of asthma symptoms: phase III of the International Study of Asthma and Allergies in Childhood (ISAAC). Thorax 2007;62:758–66. doi:10.1136/thx.2006.070169 - DOI - PMC - PubMed
    1. Pawankar R. Allergic diseases and asthma: a global public health concern and a call to action. World Allergy Organ J 2014;7:12 doi:10.1186/1939-4551-7-12 - DOI - PMC - PubMed
    1. Braun-Fahrlander C, Riedler J, Herz U et al. . Environmental exposure to endotoxin and its relation to asthma in school-age children. N Engl J Med 2002;347:869–77. doi:10.1056/NEJMoa020057 - DOI - PubMed
    1. Gern JE. Barnyard microbes and childhood asthma. N Engl J Med 2011;364:769–70. doi:10.1056/NEJMe1013713 - DOI - PubMed
    1. Romagnani S. The increased prevalence of allergy and the hygiene hypothesis: missing immune deviation, reduced immune suppression, or both? Immunology 2004;112:352–63. doi:10.1111/j.1365-2567.2004.01925.x - DOI - PMC - PubMed
Show all 29 references
Associated data
LinkOut - more resources
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3
Figure 3
Pharmacokinetics and biomarker induction following a single inhaled dose of AZD8848 in man.(A) Plasma concentration-time profiles of AZ12432045, the acid metabolite of AZD8848, following a single predicted lung-deposited dose of AZD8848 as indicated in the key. The predicted lung-deposited dose was assumed to be 63% of the dose delivered from the nebuliser. During sample preparation, any trace amounts of AZD8848 were hydrolysed by esterases in the plasma. All cohorts were administered by nebulisation taken in 10 breaths as described in Methods section. No data are shown for cohort 1 (0.15 µg) as levels of AZ12432045 were below the level of detection. Values shown are geometric means (±SD) from 3 to 5 individuals in each cohort. (B) CXCL10 was determined in induced sputum (black) and in plasma (grey) of healthy volunteers for the indicated predicted lung-deposited dose of AZD8848. The fold change at 24 h postdose from baseline for individual participants is shown. Geometric mean is shown in each cohort and significance relative to placebo (p

Figure 4

Plasma CXCL10 and blood lymphocyte…

Figure 4

Plasma CXCL10 and blood lymphocyte counts following a second inhaled dose of AZD8848.…

Figure 4
Plasma CXCL10 and blood lymphocyte counts following a second inhaled dose of AZD8848. CXCL10 in plasma (A) and lymphocytes in blood (B) were determined in healthy volunteers receiving two 30 μg doses of AZD8848 1 week apart. Data from individual participants are shown as separate lines, treatment as solid lines, and placebo as broken lines. Arrows indicate individual participants who experienced severe (black) or moderate (grey) influenza-like symptoms in the preceding period. Between-treatment and intrasubject comparisons for CXCL10 levels are described in the text.

Figure 5

Monitoring the induction of pro-inflammatory…

Figure 5

Monitoring the induction of pro-inflammatory mediators and toll-like receptors (TLRs) following inhalation of…

Figure 5
Monitoring the induction of pro-inflammatory mediators and toll-like receptors (TLRs) following inhalation of AZD8848. The mediators (A) C reactive protein (CRP), (B) interferon γ (IFNγ), (C) interleukin 8 (IL-8) and (D) tumour necrosis factor α (TNFα) were measured in plasma as described in Methods section in healthy volunteers receiving two 30 μg doses of AZD8848 1 week apart. Data from individual participants are shown as separate lines, treatment as solid lines, and placebo as broken lines. Arrows indicate individual participants who experienced severe (black) or moderate (grey) influenza-like symptoms in the preceding period. Changes in (E) TLR7 and (F) TLR9 RNA levels in blood were determined in healthy volunteers receiving a 30 μg dose of AZD8848 and samples were taken at the time points indicated. Data from individual participants are shown, those receiving placebo as a broken line, those receiving AZD8848 as a solid line. In view of the small numbers of participants, no statistical testing was performed between treatment groups.
Figure 4
Figure 4
Plasma CXCL10 and blood lymphocyte counts following a second inhaled dose of AZD8848. CXCL10 in plasma (A) and lymphocytes in blood (B) were determined in healthy volunteers receiving two 30 μg doses of AZD8848 1 week apart. Data from individual participants are shown as separate lines, treatment as solid lines, and placebo as broken lines. Arrows indicate individual participants who experienced severe (black) or moderate (grey) influenza-like symptoms in the preceding period. Between-treatment and intrasubject comparisons for CXCL10 levels are described in the text.
Figure 5
Figure 5
Monitoring the induction of pro-inflammatory mediators and toll-like receptors (TLRs) following inhalation of AZD8848. The mediators (A) C reactive protein (CRP), (B) interferon γ (IFNγ), (C) interleukin 8 (IL-8) and (D) tumour necrosis factor α (TNFα) were measured in plasma as described in Methods section in healthy volunteers receiving two 30 μg doses of AZD8848 1 week apart. Data from individual participants are shown as separate lines, treatment as solid lines, and placebo as broken lines. Arrows indicate individual participants who experienced severe (black) or moderate (grey) influenza-like symptoms in the preceding period. Changes in (E) TLR7 and (F) TLR9 RNA levels in blood were determined in healthy volunteers receiving a 30 μg dose of AZD8848 and samples were taken at the time points indicated. Data from individual participants are shown, those receiving placebo as a broken line, those receiving AZD8848 as a solid line. In view of the small numbers of participants, no statistical testing was performed between treatment groups.

References

    1. Pearce N, Ait-Khaled N, Beasley R et al. . Worldwide trends in the prevalence of asthma symptoms: phase III of the International Study of Asthma and Allergies in Childhood (ISAAC). Thorax 2007;62:758–66.
    1. Pawankar R. Allergic diseases and asthma: a global public health concern and a call to action. World Allergy Organ J 2014;7:12
    1. Braun-Fahrlander C, Riedler J, Herz U et al. . Environmental exposure to endotoxin and its relation to asthma in school-age children. N Engl J Med 2002;347:869–77.
    1. Gern JE. Barnyard microbes and childhood asthma. N Engl J Med 2011;364:769–70.
    1. Romagnani S. The increased prevalence of allergy and the hygiene hypothesis: missing immune deviation, reduced immune suppression, or both? Immunology 2004;112:352–63.
    1. Holgate ST. Pathogenesis of asthma. Clin Exp Allergy 2008;38:872–97.
    1. Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science 2010;327:291–5.
    1. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol 2003;21:335–76.
    1. Camateros P, Tamaoka M, Hassan M et al. . Chronic asthma-induced airway remodeling is prevented by toll-like receptor-7/8 ligand S28463. Am J Respir Crit Care Med 2007;175:1241–9.
    1. Vultaggio A, Nencini F, Fitch PM et al. . Modified adenine (9-benzyl-2-butoxy-8-hydroxyadenine) redirects Th2-mediated murine lung inflammation by triggering TLR7. J Immunol 2009;182:880–9.
    1. Xirakia C, Koltsida O, Stavropoulos A et al. . Toll-like receptor 7-triggered immune response in the lung mediates acute and long-lasting suppression of experimental asthma. Am J Respir Crit Care Med 2010;181:1207–16.
    1. Grela F, Aumeunier A, Bardel E et al. . The TLR7 agonist R848 alleviates allergic inflammation by targeting invariant NKT cells to produce IFN-gamma. J Immunol 2011;186:284–90.
    1. Moisan J, Camateros P, Thuraisingam T et al. . TLR7 ligand prevents allergen-induced airway hyperresponsiveness and eosinophilia in allergic asthma by a MYD88-dependent and MK2-independent pathway. Am J Physiol Lung Cell Mol Physiol 2006;290: L987–95.
    1. Sel S, Wegmann M, Sel S et al. . Immunomodulatory effects of viral TLR ligands on experimental asthma depend on the additive effects of IL-12 and IL-10. J Immunol 2007;178:7805–13.
    1. Edwards S, Jones C, Leishman AJ et al. . TLR7 stimulation of APCs results in inhibition of IL-5 through type I IFN and Notch signaling pathways in human peripheral blood mononuclear cells. J Immunol 2013;190:2585–92.
    1. Matsui H, Tomizawa H, Eiho K et al. . Mechanism of action of inhibition of allergic immune responses by a novel antedrug TLR7 agonist. J Immunol 2012;189:5194–205.
    1. Campbell JD, Kell SA, Kozy HM et al. . A limited CpG-containing oligodeoxynucleotide therapy regimen induces sustained suppression of allergic airway inflammation in mice. Thorax 2014;69:565–73.
    1. Biffen M, Matsui H, Edwards SE et al. . Biological characterisation of a novel class of toll-like receptor 7 (TLR7) agonists designed to have reduced systemic activity. Br J Pharmacol 2012;166:573–86.
    1. Huber JP, Ramos HJ, Gill MA et al. . Cutting edge: type I IFN reverses human Th2 commitment and stability by suppressing GATA3. J Immunol 2010;185:813–17.
    1. Fidock MD, Souberbielle BE, Laxton C et al. . The innate immune response, clinical outcomes, and ex vivo HCV antiviral efficacy of a TLR7 agonist (PF-4878691). Clin Pharmacol Ther 2011;89:821–9.
    1. Dudek AZ, Yunis C, Harrison LI et al. . First in human phase I trial of 852A, a novel systemic toll-like receptor 7 agonist, to activate innate immune responses in patients with advanced cancer. Clin Cancer Res 2007;13:7119–25.
    1. Pockros PJ, Guyader D, Patton H et al. . Oral resiquimod in chronic HCV infection: safety and efficacy in 2 placebo-controlled, double-blind phase IIa studies. J Hepatol 2007;47:174–82.
    1. Kurimoto A, Hashimoto K, Nakamura T et al. . Synthesis and biological evaluation of 8-oxoadenine derivatives as toll-like receptor 7 agonists introducing the antedrug concept. J Med Chem 2010;53:2964–72.
    1. Greiff L, Cervin A, Ahlstrom-Emanuelsson C et al. . Repeated intranasal TLR7 stimulation reduces allergen responsiveness in allergic rhinitis. Respir Res 2012;13:53
    1. Pavord ID, Pizzichini MMM, Pizzichini E et al. . The use of induced sputum to investigate airway inflammation. Thorax 1997;52:498–501.
    1. Padovan E, Spagnoli GC, Ferrantini M et al. . IFN-(alpha)2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8+effector T cells. J Leukoc Biol 2002;71:669–76.
    1. Tovey MG, Lallemand C. Safety, tolerability, and immunogenicity of interferons. Pharmaceuticals 2010;3:1162–86.
    1. Kamphuis E, Junt T, Waibler Z et al. . Type I interferons directly regulate lymphocyte recirculation and cause transient blood lymphopenia. Blood 2006;108:3253–61.
    1. Koga-Yamakawa E, Murata M, Dovedi SJ et al. . TLR7 tolerance is independent of the type I IFN pathway and leads to loss of anti-tumor efficacy in mice. Cancer Immunol Immunother 2015;64:1229–39.

Source: PubMed

3
Se inscrever