Absorption, Metabolism, and Excretion of ACT-1004-1239, a First-In-Class CXCR7 Antagonist: In Vitro, Preclinical, and Clinical Data

Christine Huynh, Swen Seeland, Jerome Segrestaa, Carmela Gnerre, Jens Hogeback, Henriette E Meyer Zu Schwabedissen, Jasper Dingemanse, Patricia N Sidharta, Christine Huynh, Swen Seeland, Jerome Segrestaa, Carmela Gnerre, Jens Hogeback, Henriette E Meyer Zu Schwabedissen, Jasper Dingemanse, Patricia N Sidharta

Abstract

ACT-1004-1239 is a potent, selective, first-in-class CXCR7 antagonist, which shows a favorable preclinical and clinical profile. Here we report the metabolites and the metabolic pathways of ACT-1004-1239 identified using results from in vitro and in vivo studies. Two complementary in vitro studies (incubation with human liver microsomes in the absence/presence of cytochrome P450- [CYP] specific chemical inhibitors and incubation with recombinant CYPs) were conducted to identify CYPs involved in ACT-1004-1239 metabolism. For the in vivo investigations, a microtracer approach was integrated in the first-in-human study to assess mass balance and absorption, distribution, metabolism, and excretion (ADME) characteristics of ACT-1004-1239. Six healthy male subjects received orally 100 mg non-radioactive ACT-1004-1239 together with 1 μCi 14C-ACT-1004-1239. Plasma, urine, and feces samples were collected up to 240 h post-dose and 14C-drug-related material was measured with accelerator mass spectrometry. This technique was also used to construct radiochromatograms of pooled human samples. Metabolite structure elucidation of human-relevant metabolites was performed using high performance liquid chromatography coupled with high resolution mass spectrometry and facilitated by the use of rat samples. CYP3A4 was identified as the major CYP catalyzing the formation of M1 in vitro. In humans, the cumulative recovery from urine and feces was 84.1% of the dose with the majority being eliminated via the feces (69.6%) and the rest via the urine (14.5%). In human plasma, two major circulating metabolites were identified, i.e., M1 and M23. Elimination via M1 was the only elimination pathway that contributed to ≥25% of ACT-1004-1239 elimination. M1 was identified as a secondary amine metabolite following oxidative N-dealkylation of the parent. M23 was identified as a difluorophenyl isoxazole carboxylic acid metabolite following central amide bond hydrolysis of the parent. Other metabolites observed in humans were A1, A2, and A3. Metabolite A1 was identified as an analog of M1 after oxidative defluorination, whereas both, A2 and A3, were identified as a reduced analog of M1 and parent, respectively, after addition of two hydrogen atoms at the isoxazole ring. In conclusion, CYP3A4 contributes to a relevant extent to ACT-1004-1239 disposition and two major circulating metabolites were observed in humans. Clinical Trial Registration: (https://ichgcp.net/clinical-trials-registry/NCT03869320) ClinicalTrials.gov Identifier NCT03869320.

Keywords: 14C-ACT-1004-1239; ADME; CXCR7; CYP3A4; accelerator mass spectrometry; first-in-human; microtracer.

Conflict of interest statement

The authors declare that this study was sponsored by Idorsia Pharmaceuticals Ltd. The sponsor was involved in the study design, collection, analysis, interpretation of data, the writing of this article/the decision to submit it for publication.

Copyright © 2022 Huynh, Seeland, Segrestaa, Gnerre, Hogeback, Meyer zu Schwabedissen, Dingemanse and Sidharta.

Figures

FIGURE 1
FIGURE 1
In vitro studies: metabolic profiles of ACT-1004-1239 following incubation with (A) human liver microsomes (HLM), (B) HLM and 1 μM ketoconazole, or (C) recombinant CYP3A4.
FIGURE 2
FIGURE 2
Arithmetic mean (+SD) concentration vs. time profile of total 14C-radioactivity in plasma on linear and semilog (inset) scale following a single oral administration of 9.2 μg 14C-ACT-1004-1239 on top of 100 mg non-radioactive ACT-1004-1239 (N = 6).
FIGURE 3
FIGURE 3
Arithmetic mean (+SD) cumulative recovery vs. time profile of total 14C-radioactivity in urine and feces (shown as % of administered dose) following a single oral administration of 9.2 μg 14C-ACT-1004-1239 on top of 100 mg non-radioactive ACT-1004-1239 (N = 6).
FIGURE 4
FIGURE 4
Metabolic profiles of ACT-1004-1239 in human plasma (top), urine (middle), and feces (bottom) pools. Squared brackets placed above and below each radiochromatogram indicate metabolites with a relative abundance of

FIGURE 5

Representative metabolic profile of ACT-1004-1239…

FIGURE 5

Representative metabolic profile of ACT-1004-1239 in rat urine after intravenous dosing of 14…

FIGURE 5
Representative metabolic profile of ACT-1004-1239 in rat urine after intravenous dosing of 14C-ACT-1004-1239.

FIGURE 6

Mass spectrometric fragmentation pattern of…

FIGURE 6

Mass spectrometric fragmentation pattern of 14 C-ACT-1004-1239. The asterisk (*) indicates the position…

FIGURE 6
Mass spectrometric fragmentation pattern of 14C-ACT-1004-1239. The asterisk (*) indicates the position of the 14C label, which was taken into consideration for the applicable accurate fragment masses (m/z). The fragment ion with m/z 212 includes in addition to the depicted pattern two hydrogen atoms.

FIGURE 7

Proposed metabolic pathways of ACT-1004-1239…

FIGURE 7

Proposed metabolic pathways of ACT-1004-1239 in humans.

FIGURE 7
Proposed metabolic pathways of ACT-1004-1239 in humans.
All figures (7)
FIGURE 5
FIGURE 5
Representative metabolic profile of ACT-1004-1239 in rat urine after intravenous dosing of 14C-ACT-1004-1239.
FIGURE 6
FIGURE 6
Mass spectrometric fragmentation pattern of 14C-ACT-1004-1239. The asterisk (*) indicates the position of the 14C label, which was taken into consideration for the applicable accurate fragment masses (m/z). The fragment ion with m/z 212 includes in addition to the depicted pattern two hydrogen atoms.
FIGURE 7
FIGURE 7
Proposed metabolic pathways of ACT-1004-1239 in humans.

References

    1. Amaya G. M., Durandis R., Bourgeois D. S., Perkins J. A., Abouda A. A., Wines K. J., et al. (2018). Cytochromes P450 1A2 and 3A4 Catalyze the Metabolic Activation of Sunitinib. Chem. Res. Toxicol. 31, 570–584. 10.1021/acs.chemrestox.8b00005
    1. Arjomand A. (2010). Accelerator Mass Spectrometry-Enabled Studies: Current Status and Future Prospects. Bioanalysis 2, 519–541. 10.4155/bio.09.188
    1. Boddy A. V., Sludden J., Griffin M. J., Garner C., Kendrick J., Mistry P., et al. (2007). Pharmacokinetic Investigation of Imatinib Using Accelerator Mass Spectrometry in Patients with Chronic Myeloid Leukemia. Clin. Cancer Res. 13, 4164–4169. 10.1158/1078-0432.CCR-06-2179
    1. Chen Y., Liu L., Nguyen K., Fretland A. J. (2011). Utility of Intersystem Extrapolation Factors in Early Reaction Phenotyping and the Quantitative Extrapolation of Human Liver Microsomal Intrinsic Clearance Using Recombinant Cytochromes P450. Drug Metab. Dispos. 39, 373–382. 10.1124/dmd.110.035147
    1. Dalvie D. K., Kalgutkar A. S., Khojasteh-Bakht S. C., Obach R. S., O'Donnell J. P. (2002). Biotransformation Reactions of Five-Membered Aromatic Heterocyclic Rings. Chem. Res. Toxicol. 15, 269–299. 10.1021/tx015574b
    1. European Medicines Agency Evaluation of Medicines for Human Use (2018). Guideline on Strategies to Identify and Mitigate Risks for First-In-Human and Early Clinical Trials with Investigational Medicinal Products. Available at: (Accessed September 29, 2021).
    1. Graham L., Garner R. C. (2003). Big Physics, Small Doses: The Use of AMS and PET in Human Microdosing of Development Drugs. Nat. Rev. Drug Discov. 2, 233–240. 10.1038/nrd1037
    1. Hamilton R. A., Garnett W. R., Kline B. J. (1981). Determination of Mean Valproic Acid Serum Level by Assay of a Single Pooled Sample. Clin. Pharmacol. Ther. 29, 408–413. 10.1038/clpt.1981.56
    1. Huynh C., Brussee J. M., Pouzol L., Fonseca M., Meyer zu Schwabedissen H. E., Dingemanse J., et al. (2021a). Target Engagement of the First-In-Class CXCR7 Antagonist ACT-1004-1239 Following Multiple-Dose Administration in Mice and Humans. Biomed. Pharmacother. 144, 112363. 10.1016/j.biopha.2021.112363
    1. Huynh C., Henrich A., Strasser D. S., Boof M. L., Al-Ibrahim M., Meyer zu Schwabedissen H. E., et al. (2021b). A Multipurpose First-In-Human Study with the Novel CXCR7 Antagonist ACT-1004-1239 Using CXCL12 Plasma Concentrations as Target Engagement Biomarker. Clin. Pharmacol. Ther. 109, 1648–1659. 10.1002/cpt.2154
    1. Kalgutkar A. S., Nguyen H. T., Vaz A. D. N., Doan A., Dalvie D. K., McLeod D. G., et al. (2003). In Vitro METABOLISM STUDIES ON THE ISOXAZOLE RING SCISSION IN THE ANTI-INFLAMMATORY AGENT LEFLUNOMIDE TO ITS ACTIVE α-CYANOENOL METABOLITE A771726: MECHANISTIC SIMILARITIES WITH THE CYTOCHROME P450-CATALYZED DEHYDRATION OF ALDOXIMES. Drug Metab. Dispos. 31, 1240–1250. 10.1124/dmd.31.10.1240
    1. Lappin G., Kuhnz W., Jochemsen R., Kneer J., Chaudhary A., Oosterhuis B., et al. (2006). Use of Microdosing to Predict Pharmacokinetics at the Therapeutic Dose: Experience with 5 Drugs. Clin. Pharmacol. Ther. 80, 203–215. 10.1016/j.clpt.2006.05.008
    1. Magdalou J., Fournel-Gigleux S., Testa B., Ouzzine M. (2003). “Biotransformation Reactions,” in The Practice of Medicinal Chemistry (London: Elsevier; ), 517–543. 10.1016/B978-012744481-9/50035-0
    1. Mannens G., Huang M. L., Meuldermans W., Hendrickx J., Woestenborghs R., Heykants J. (1993). Absorption, Metabolism, and Excretion of Risperidone in Humans. Drug Metab. Dispos 21, 1134–1141. Available at: .
    1. Muehlan C., Fischer H., Zimmer D., Aissaoui H., Grimont J., Boss C., et al. (2019). Metabolism of the Dual Orexin Receptor Antagonist ACT-541468, Based on Microtracer/Accelerator Mass Spectrometry. Curr. Drug Metab. 20, 254–265. 10.2174/1389200220666190206141814
    1. Muehlan C., Heuberger J., Juif P. E., Croft M., van Gerven J., Dingemanse J. (2018). Accelerated Development of the Dual Orexin Receptor Antagonist ACT-541468: Integration of a Microtracer in a First-In-Human Study. Clin. Pharmacol. Ther. 104, 1022–1029. 10.1002/cpt.1046
    1. Palmer D. C. (2003). Oxazoles: Synthesis, Reactions, and Spectroscopy, Part A, Vol. 60. Raritan, NJ: John Wiley & Sons.
    1. Pouzol L., Baumlin N., Sassi A., Tunis M., Marrie J., Vezzali E., et al. (2021a). ACT‐1004‐1239, a First‐in‐class CXCR7 Antagonist with Both Immunomodulatory and Promyelinating Effects for the Treatment of Inflammatory Demyelinating Diseases. FASEB j. 35, 1–17. 10.1096/fj.202002465R
    1. Pouzol L., Sassi A., Baumlin N., Tunis M., Strasser D. S., Lehembre F., et al. (2021b). CXCR7 Antagonism Reduces Acute Lung Injury Pathogenesis. Front. Pharmacol. 12, 748740. 10.3389/fphar.2021.748740
    1. Pouzol L., Tunis M., Baumlin N., Sassi A., Marrie J., Vezzali E., et al. (2021c). CXCR7 Antagonism with ACT-1004-1239 Reduces Neuroinflammation and Accelerates Remyelination in Murine Demyelinating Models (2236). Neurology 96, 2236. Available at: .
    1. Richard-Bildstein S., Aissaoui H., Pothier J., Schäfer G., Gnerre C., Lindenberg E., et al. (2020). Discovery of the Potent, Selective, Orally Available CXCR7 Antagonist ACT-1004-1239. J. Med. Chem. 63, 15864–15882. 10.1021/acs.jmedchem.0c01588
    1. Roffel A., Marle S. v., Choi S., Lee H., Dueker S., Tiessen R. (2017). Metabolism and Excretion of GCC-4401C, a Factor Xa Inhibitor, in Humans. Int. J. Pharmacokinet. 2, 93–104. 10.4155/ipk-2016-0015
    1. Roffey S. J., Obach R. S., Gedge J. I., Smith D. A. (2007). What Is the Objective of the Mass Balance Study? A Retrospective Analysis of Data in Animal and Human Excretion Studies Employing Radiolabeled Drugs. Drug Metab. Rev. 39, 17–43. 10.1080/03602530600952172
    1. Spracklin D. K., Chen D., Bergman A. J., Callegari E., Obach R. S. (2020). Mini-Review: Comprehensive Drug Disposition Knowledge Generated in the Modern Human Radiolabeled ADME Study. CPT Pharmacometrics Syst. Pharmacol. 9, 428–434. 10.1002/psp4.12540
    1. Stiborová M., Martínek V., Rýdlová H., Koblas T., Hodek P. (2005). Expression of Cytochrome P450 1A1 and its Contribution to Oxidation of a Potential Human Carcinogen 1-Phenylazo-2-Naphthol (Sudan I) in Human Livers. Cancer Lett. 220, 145–154. 10.1016/j.canlet.2004.07.036
    1. Sugihara K., Kitamura S., Tatsumi K. (1996). Involvement of Mammalian Liver Cytosols and Aldehyde Oxidase in Reductive Metabolism of Zonisamide. Drug Metab. Dispos. 24, 199–202. Available at: .
    1. Ufer M., Juif P. E., Boof M. L., Muehlan C., Dingemanse J. (2017). Metabolite Profiling in Early Clinical Drug Development: Current Status and Future Prospects. Expert Opin. Drug Metab. Toxicol. 13, 803–806. 10.1080/17425255.2017.1351944
    1. United States Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (2020a). In Vitro Drug Interaction Studies - Cytochrome P450 Enzyme and Transporter Mediated Drug Interactions, Guidance for Industry. Available at: (Accessed September 1, 2021).
    1. United States Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (2003). Pharmacokinetics in Patients with Impaired Hepatic Function: Study Design, Data Analysis, and Impact on Dosing and Labeling, Guidance for Industry. Available at: (Accessed August 25, 2021).
    1. United States Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (2020b). Safety Testing of Drug Metabolites, Guidance for Industry. Revision 2. Available at: (Accessed August 16, 2021).
    1. Xie C., Zhou J., Guo Z., Diao X., Gao Z., Zhong D., et al. (2013). Metabolism and Bioactivation of Famitinib, a Novel Inhibitor of Receptor Tyrosine Kinase, in Cancer Patients. Br. J. Pharmacol. 168, 1687–1706. 10.1111/bph.12047
    1. Zhang D., Raghavan N., Chen S. Y., Zhang H., Quan M., Lecureux L., et al. (2008). Reductive Isoxazole Ring Opening of the Anticoagulant Razaxaban Is the Major Metabolic Clearance Pathway in Rats and Dogs. Drug Metab. Dispos. 36, 303–315. 10.1124/dmd.107.018416

Source: PubMed

3
Abonneren