Clinical Implications and Translation of an Off-Target Pharmacology Profiling Hit: Adenosine Uptake Inhibition In Vitro

Hamid R Amouzadeh, Isaiah Dimery, Jonathan Werner, Gataree Ngarmchamnanrith, Michael J Engwall, Hugo M Vargas, Deborah Arrindell, Hamid R Amouzadeh, Isaiah Dimery, Jonathan Werner, Gataree Ngarmchamnanrith, Michael J Engwall, Hugo M Vargas, Deborah Arrindell

Abstract

Off-target activities of drug candidates observed during in vitro pharmacological profiling frequently do not translate to adverse events (AEs) in human. This could be because off-target activities do not have functional consequences, are not observed at exposures achieved during clinical testing, or may not translate into clinical outcomes. We report clinical consequences of an off-target activity observed during profiling of AMG 337, a selective inhibitor of the mesenchymal-epithelial transition factor being evaluated for treatment of solid tumors. In our screen of 151 potential off-targets, AMG 337 inhibited only adenosine transporter (AT). During clinical trials, headache emerged as the dose-limiting AE in the first-in-human trial. It was thought that headache was caused by extracellular accumulation of adenosine from inhibition of AT by AMG 337 and subsequent adenosine-mediated vasodilation through adenosine receptors (ARs). Further nonclinical studies were performed to evaluate this hypothesis. AMG 337 inhibited AT function in dog and human cells in vitro and dog and human arteries ex vivo. In a dog telemetry study, AMG 337 caused hypotension, which was reduced by pretreatment with theophylline, an AR antagonist. Overall, nonclinical and clinical data suggested that headache was due to cerebral vasorelaxation caused by AMG 337-mediated inhibition of AT. When subjects were advised to drink coffee, an AR antagonist, prior to AMG 337, the severity of headaches was reduced, allowing them to continue treatment. These findings demonstrate the importance of carefully evaluating clinical observations during early drug development and the value of translational nonclinical studies to investigate the mechanism of action driving clinical observations.

Copyright © 2019. Published by Elsevier Inc.

Figures

Figure 1
Figure 1
Dose-dependent inhibition of AT by AMG 337 in guinea-pig cerebral cortex (binding, blue) and human HeLa cells (functional, black) and in vivo exposure in humans and dogs. The maximum concentration of free AMG 337 at the 400-mg QD dose in human is estimated to be 3.4 μM. This level of exposure is approximately 12 times higher than the IC50 of the AT in the functional assay with human cells (0.28 μM) and 20 times higher than the IC50 of the AT in the functional assay with dog cells (0.17 μM). In the dog study with theophylline, the free concentration of AMG 337 at 8 hours postdose (30 mg/kg) was 5.4 μM, which is similar to the free Cmax of 5.8 μM on day 1 in a previous 1-month study in dogs with AMG 337 at 30 mg/kg/day.
Figure 2
Figure 2
Relaxation responses of AMG 337 in precontracted dog and human vessels (A) dog subcutaneous artery and (B) human subcutaneous artery. Black = vehicle and blue = AMG 337.
Figure 3
Figure 3
Mean arterial pressure in dogs administered AMG 337.
Figure 4
Figure 4
Mean heart rate in dogs administered AMG 337.
Figure 5
Figure 5
Mean arterial pressure in dogs treated with empty capsules, theophylline, AMG 337, or a combination of theophylline and AMG 337.
Figure 6
Figure 6
Heart rate in dogs treated with empty capsules, theophylline, AMG 337, or a combination of theophylline and AMG 337.

References

    1. Bowes J, Brown AJ, Hamon J, Jarolimek W, Sridhar A, Waldron G, Whitebread S. Reducing safety-related drug attrition: the use of in vitro pharmacological profiling. Nat Rev Drug Discov. 2012;11:909–922.
    1. Lynch JJ, III, Nan Vleet TR, Mittelstadt SW, Bloome AG. Potential functional and pathological side effects related to off-target pharmacological activity. J Pharmacol Toxicol Meth. 2017;87:108–126.
    1. Valentin JP, Guillon JM, Jenkinsn S, Kadambi V, Ravikumar P, Roberts S, Rosenbrier-Ribeiro L, Schmidt F, Armstrong D. In vitro secondary pharmacological profiling: An IQ-DruSafe industry survey on current practices. J Pharmacol Toxicol Meth. 2018;93:7–14.
    1. Rosenbrier-Ribeiro L, Storer RI. A semi-quantitative translational pharmacology analysis to understand the relationship between in vitro ENT1 inhibition and the clinical incidence of dyspnoea and bronchospasm. Toxicol Appl Pharmacol. 2017;317:41–50.
    1. Papoian T, Chiu HJ, Ikram E, Jagadeesh G, Khan I, Laniyonu AA, Li CX, Saulnier M, Simpson N, Yang B. Secondary pharmacology data to assess potential off-target activity of new drugs: a regulatory perspective. Nat Rev Drug Discov. 2015;14:294.
    1. Hughes PE, Rex K, Caenepeel S, Yang Y, Zhang Y, Broome MA, Kha HT, Burgess TL, Amore B, Kaplan-Lefko PJ. In vitro and in vivo activity of AMG 337, a potent and selective MET kinase inhibitor in MET-dependent cancer models. Mol Cancer Ther. 2016;15(7):1568–1579.
    1. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–925.
    1. Fabian MA, Biggs WH, III, Treiber DK, Atteridge CE, Azimioara MD, Benedetti MG, Carter TA, Ciceri P, Edeen PT, Floyd M. A small molecule-kinase interaction map for clinical kinase inhibitors. Nat Biotechnol. 2005;23:329–336.
    1. Karaman MW, Herrgard S, Treibert DK, Gallant P, Atteridge CE, Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT. A quantitative analysis of kinase inhibitor selectivity. Nature Biotech. 2008;26(1):127–132.
    1. Eurofins CEREP 2019.
    1. Verma A, Marangos PJ. Nitrobenzylthioinosine binding in brain: an interspecies study. Life Sci. 1985;36:286–290.
    1. Harley ER, Patterson AR, Cass CE. Initial rate kinetics of the transport of adenosine and 4-amino-7-(β-D-ribofuranosyl) pyrrolo[2,3-d]pyrimidine (tubercidin) in cultured cells. Cancer Res. 1982;42:1289–1295.
    1. Mulvany MJ, Halpern W. Contractile properties of small arterial resistance vessels in spontaneously hypertensive and normotensive rats. Circ Res. 1977;41:19–26.
    1. Afonso S. Inhibition of coronary vasodilating action of dipyridamole and adenosine by aminophylline in the dog. Circ Res. 1970;26:743–752.
    1. Munsiff IJ, Mckiernan BC, Neff-Davis CA, Koritz GD. Determination of the acute oral toxicity of theophylline in conscious dogs. J Vet Pharmacol Thera. 1988;11:381–389.
    1. Noji T, Karasawa A, Kusada H. Adenosine uptake inhibitors. Eur J Pharmacol. 2004;495:1–16.
    1. Sheth S, Brito R, Mukhejea D, Rybak LP, Ramkumar V. Adenosine receptors: expression, function and regulation. Int J Mol Sci. 2014;15:2024–2052.
    1. Layland J, Carrick D, Lee M, Oldroyd K, Berry C. Adenosine. physiology, pharmacology, and clinical applications J Am Coll Cardiol Intv. 2014;7:581–591.
    1. Young JD, Yao SYM, Baldwin JM, Cass CE, Baldwin SA. The human concentrative and equilibrative nucleoside transporter families, SLC28 and SLC29. Mol Aspects Med. 2013;34:529–547.
    1. Borea AP, Gessi S, Merighi S, Vincenzi F, Varani K. Pharmacology of adenosine receptors: the state of the art. Physiol Rev. 2018;98:1591–1625.
    1. Feuerstein TJ, Zeides A. theophylline relieves headache following lumbar puncture — placebo-controlled, double-blind pilot study. Klin Wochenschr. 1986;64:216–218.
    1. Lexiscan (regadenoson), prescribing information (2014). Drugs@FDA () accessed August 2018.
    1. Kruuse C, Jacobsen TB, Lassen LH, Thomsen LL, Hasselbalch SG, Dige-Petersen H, Olesen J. Dipyridamole dilates large cerebral arteries concomitant to headache induction in healthy subjects. J of Cereb Bld Flow and Metab. 2000;20:1372–1379.
    1. Meyers AM, Topham L, Ballow J, Totah D, Wilke R. Adverse reactions to dipyridamole in patients undergoing stress/rest cardiac perfusion testing. J Nucl Med Technol. 2002;30:21–24.
    1. Pennycooke M, Chaudary N, Shuralyova I, Zhang Y, Coe IR. Differential expression of human nucleoside transporters in normal and tumor tissue. Biochem Biophys Res Comm. 2001;280:951–959.
    1. Farre X, Guillen-Gomez E, Sanchez L, Hardisson D, Plaza Y, Lloberas J, Casado J, Palacios J, Al Pastor-Anglada M. Expression of the nucleoside-derived drug transporters hCNT1, hENT1 and hENT2 in gynecologic tumors. Int J Cancer. 2004;112:959–966.

Source: PubMed

3
订阅