The neuronal calcium ion channel activity of constrained analogues of MONIRO-1

https://doi.org/10.1016/j.bmc.2020.115655Get rights and content

Abstract

Structural modifications of the neuronal calcium channel blocker MONIRO-1, including constraining the phenoxyaniline portion of the molecule and replacing the guanidinium functionality with tertiary amines, led to compounds with significantly improved affinities for the endogenously expressed CaV2.2 channel in the SH-SY5Y neuroblastoma cell line. These analogues also showed promising activity towards the CaV3.2 channel, recombinantly expressed in HEK293T cells. Both of these ion channels have received attention as likely targets for the treatment of neuropathic pain. The dibenzoazepine and dihydrobenzodiazepine derivatives prepared in this study show an encouraging combination of neuronal calcium ion channel inhibitory potency, plasma stability and potential to cross the blood–brain-barrier.

Introduction

Neuropathic pain is a type of chronic pain that results from nerve damage caused by trauma, surgery or diseases, including diabetes, shingles (herpes zoster) and cancer, and is often characterized by a complex array of unpleasant sensory experiences. The estimated prevalence within the general population ranges from 1 to 10%, and this number is likely to increase with the aging of the population.1, 2 Unlike nociceptive pain, neuropathic pain can occur without stimuli, and although non-prescription pain medications such as NSAIDs are often widely used by patients suffering from this condition, there is a lack of strong evidence supporting this use.3 Instead, patients must often rely on the off-label prescription of antidepressants, such as amitriptyline and nortriptyline,4 anticonvulsants like carbamazepine, and in more severe cases, opioids.1, 2 The CaV2.2 (N-Type) voltage-gated calcium channel has been shown to be a promising target for the treatment of neuropathic pain.5, 6 The gabapentinoids, gabapentin and pregabalin, are also widely prescribed pain medications, and have been shown to interact with the auxiliary α2δ subunit of the CaV2.2 channel.7 Many neuropathic pain patients, however, obtain only limited relief from these available therapies.

The selective peptidic CaV2.2 inhibitor, ziconotide, a synthetic version of ω-conotoxin MVIIA, can be used as a last-resort treatment for different types of neuropathic pain in the US and Europe, with some guidelines now beginning to recommended it as a first-line agent.8 However, its side effects are often severe at therapeutic doses (therapeutic window 1.5–2.1) and needs to be delivered by injection into the spinal canal via an intrathecal catheter. Importantly, ziconotide is about 10-times more potent than intrathecally administered morphine, and unlike with morphine, its administration does not appear to induce tolerance, addiction or respiratory depression.6, 8, 9, 10, 11, 12 The clinical results obtained with ziconotide provide some of the strongest validation of the N-type neuronal calcium channel as target for the treatment of intractable neuropathic pain. The limitations associated with ziconotide treatments, however, mean that there is still a strong need to develop small molecule drugs that selectively inhibit CaV2.2 channels and can cross the blood–brain barrier following oral or intravenous delivery. Despite concerted efforts, both academic and commercial, no drugs of this type have so far successfully passed through Phase III clinical trials.13

One approach is to engineer low molecular weight compounds that mimic key features of pain-blocking bioactive peptides.14 Following this strategy, efforts initially aimed to develop type III peptide mimetics of ω-conotoxin GVIA,15, 1617 a 27-residue peptide with greater selectivity and inhibitory potency for CaV2.2 than ω-conotoxin MVIIA. In an attempt to engender more drug-like characteristics into these early, relatively high molecular weight and polar compounds, one program of work led to a series of phenoxyanilides, which retained good levels of CaV2.2 inhibition.18, 19 It was later discovered that these compounds are also antagonists of the T-type CaV3.2 calcium channel, inhibitors of which have also been shown to have analgesic effects in animal models.20, 21 While these compounds are distant from ideal drugs, particularly because they are readily metabolised to toxic phenoxyanilines,22 they are of interest because one of the first to be investigated, MONIRO-1 (1, Fig. 1), was found in whole-cell patch clamp electrophysiology experiments to inhibit CaV2.2 channels with an IC50 = 34 µM and CaV3 channels at 2–7 µM, while showing selectivity for these channels over other neuronal calcium channels.23 More interestingly, MONIRO-1 exhibited state- and use-dependent inhibition of CaV2.2 channels and stronger inhibition at high stimulation frequencies with CaV3.1 channels, properties which, if reproduced in more drug-like compounds, are likely to provide clinically-relevant functional selectivity.

In the absence of a crystal structure or a well-developed in silico model for these neuronal calcium channels, improved knowledge of an optimum pharmacophore continues to be attained from bioassay-guided development of compound libraries that embody iterative structural modifications around promising leads. With the phenoxyaniline series, it was of interest to understand if constraining the relatively free rotation of the two phenyl rings about the bonds attached to the bridging oxygen in MONIRO-1 would lead to improved inhibitory potency for the target N-type and T-type neuronal calcium ion channels. In addition, it was hoped that activity would be retained if the guanidinium group was replaced with less hydrophilic tertiary amine functionalities, as this would provide more drug-like compounds with greater potential to cross the blood–brain barrier.

Section snippets

Chemistry

Two approaches to restricting the rotational freedom in the phenoxylaniline fragment of 1 were investigated. The incorporation of ortho-methyl substituents would favour a twisted conformation whereas a planar arrangement could be simply obtained by replacing the phenoxyaniline fragment with a phenoxazine. For synthetic accessibility, the effect of these structural changes on biological activity was assessed with the corresponding non-fluorinated analogues. It was thus necessary to prepare 6b,

Biology

The bioactivities of guanidinium (6), dimethylamine (7), pyrrolidine (8) and morpholine (9) analogues were evaluated for inhibition of the human CaV2.2 channel endogenously expressed in the neuroblastoma cell line SH-SY5Y,27, 28 and CaV3.2 channels, recombinantly expressed in HEK293T cells.24 The CaV2.2 assays were performed in the presence of the CaV1 blocker nifedipine. These calcium flux imaging assays employed the FLIPRTETRA platform and were undertaken at the natural membrane potential of

Discussion

The guanidium compounds (1, 6b and 6d) displayed relatively weak inhibition of calcium influx in FLIPR imaging assays for the CaV2.2 channel, compared with that observed for MONIRO-1(1) in the previously reported whole-cell patch clamp studies.23 In addition, the lack of the fluorine substituent in 6b also to led to a significant drop in activity relative to the fluorinated analogue (1). It was therefore pleasing to observe an approximately 10-fold improvement in potency when the guanidium

Conclusions

This investigation initially set out to determine the effect of constraining the phenoxyaniline portion of MONIRO-1 on the compound’s inhibitory potency for CaV2.2 and CaV3.2 channels, targets for neuropathic pain treatments. For the CaV2.2 channel, neither enforcing a twist (7c) nor locking the phenoxyaniline segment into planarity, in the form of a phenoxazine motif (7d), had a marked effect on inhibitory potency, whereas for the CaV3.2 channel, there was a slight preference for blocking by

General experimental

General experimental methods, reagents and instrumentation used to prepare the tested compounds were the same as those previously reported.24 4-(3-Chloropropoxy)benzoic acid was prepared as previously described.19 The synthesis of compounds 2c is reported in Supplementary material.

Column chromatography was performed using silica gel (100–200 mesh size) with indicated solvents. Thin-layer chromatography (TLC) was conducted with silica gel 60 F254 precoated plates (0.25 mm) and visualized with

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

Monash University and CSIRO are acknowledged for funding. This work was also supported by a NHMRC Program Grant (APP1072113, RJL) and NHMRC Fellowship (1119056, RJL).

References (36)

  • I. Vetter et al.

    Biochem Pharmacol

    (2010)
  • T. Yamamoto et al.

    Bioorg Med Chem Lett

    (2011)
  • R. Aslanian et al.

    Bioorg Med Chem Lett

    (2008)
  • G. Cruccu et al.

    Pain Ther

    (2017)
  • K.E. Galluzzi

    J Am Osteopath Assoc

    (2005)
  • C.-C. Chi et al.

    The Cochrane Library

    (2015)
  • J. Wong et al.

    BMJ

    (2017)
  • W.G. Brose et al.

    Clin J Pain

    (1997)
  • View full text