Inhibition of the cellular function of perforin by 1-amino-2,4-dicyanopyrido[1,2-a]benzimidazoles

https://doi.org/10.1016/j.bmc.2011.05.013Get rights and content

Abstract

A high throughput screen showed the ability of a 1-amino-2,4-dicyanopyrido[1,2-a]benzimidazole analogue to directly inhibit the lytic activity of the pore-forming protein perforin. A series of analogues were prepared to study structure–activity relationships (SAR) for the this activity, either directly added to cells or released in situ by KHYG-1 NK cells, at non-toxic concentrations. These studies showed that the pyridobenzimidazole moiety was required for effective activity, with strongly basic centres disfavoured. This class of compounds was relatively unaffected by the addition of serum, which was not the case for a previous class of direct inhibitors.

Introduction

Perforin (PRF) is a pore-forming member of the membrane-attack-complex/PRF (MACPF) protein family1, and is secreted, along with a range of serine proteases (granzymes), by both cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells.2 Together, perforin and granzymes comprise a principal mechanism of CTL/NK cytotoxicity by which the immune systems of higher organisms protect themselves against pathogens and transformed cells. The critical role of perforin in this process is its ability, upon binding calcium, to assemble into aggregates of 12–18 molecules that form trans-membrane pores of 10–20 nm in diameter in the plasma membrane of the target cell, facilitating the entry of granzymes, which can trigger apoptosis.3, 4 Initially, the calcium-dependent membrane binding of perforin monomers is mediated by their C terminal C2 domains.5 Successive perforin monomers then assemble into a cylindrical pore via a series of salt bridges linking Arg213 on the ‘front’ of one monomer with Glu343 on the ‘back’ of the next.6 The structure of mouse perforin monomer at a resolution of 2.75 Å has recently been reported as a bent and twisted four-stranded β-sheet MACPF domain flanked by two clusters of α-helices that insert into membranes as β-strands upon pore formation.7 In the same report, cryo-electron microscopy also confirmed the distribution of individual perforin sub-domains within an assembled pore structure.

Apart from well-recognised disease syndromes due to mutation and/or down-regulation of perforin expression, there is also much evidence linking inappropriate perforin activity to several human pathologies, including cerebral malaria, type 1 diabetes, juvenile idiopathic arthritis and postviral myocarditis,7 as well as therapy-induced conditions such as allograft rejection and graft-versus-host disease.8, 9 Perforin is therefore a potentially druggable target, especially as it is encoded by a single-copy gene in both mice and humans.

However, there have been few reports of small-molecule inhibitors of perforin function, as distinguished from compounds that affect it indirectly by inhibiting cell respiration or perforin processing.10 Enzastaurin (1) (Fig. 1), a bisindolylmaleimide structurally related to staurosporine and currently in Phase II trial for cancer, has recently been reported11 to inhibit perforin release from NK cells, but is thought to act via the PKC/PI3K/AKT pathway. We recently reported10 a series of dihydrofuro[3,4-c]pyridinones (e.g., 2) (Fig. 1) as direct inhibitors of the cytolytic effects of perforin in cells, and demonstrated limited structure–activity effects across the series. The most active compound in the series (2) had a IC50 of 1 μM for inhibition of the lysis of Jurkat cells by added perforin, and at a concentration of 20 μM showed a 60% inhibition in the killing of K562 cells by perforin released from KHYG-1 NK cells, with little toxicity to the latter. However, the cellular activity of these compounds was later shown to be adversely affected by increasing concentrations of serum, limiting their further development. In the present paper we discuss the discovery of this new class of direct perforin inhibitors, the 1-amino-2,4-dicyanopyrido[1,2-a]benzimidazoles, and initial structure–activity relationship (SAR) studies on this new class.

Section snippets

Chemistry

The lead compound (5) was selected from a high throughput screen12 of 100,000 compounds sourced from commercial libraries, using a 384-well plate format. Compounds were screened (at 20 μM) by incubation with sheep red blood cells, which in the absence of an inhibitor are lysed by perforin, generating turbidity that can be measured by the change in absorbance at 650 nm. Compound 5 was one of a few hits that were confirmed in an assay measuring its potency for inhibiting the lysis of Jurkat human

Conclusions

Molecules with the ability to directly inhibit the function of perforin are of potential use in the treatment of some autoimmune diseases and therapy-induced conditions characterised by undesired perforin secretion, but to date only one class, the dihydrofuro[3,4-c]pyridinones have been reported.10 Compound 5 in the present paper was one of a small number of compounds from a high throughput screen to show confirmatory inhibition of the lytic activity of perforin in a cell-based assay, and as a

Chemistry

Analyses were performed by the Microchemical Laboratory, University of Otago, Dunedin, NZ. Many samples tenaciously held water or DMF (verified by NMR). Melting points were determined using an Electrothermal Model 9200 and are as read. NMR spectra were measured in CD3SOCD3 (unless otherwise specified) on a Bruker Advance 400 spectrometer and referenced to Me4Si. Mass spectra were recorded either on a Varian VG 7070 spectrometer at nominal 5000 resolution or a Finnigan MAT 900Q spectrometer. All

Acknowledgments

The authors thank the Wellcome Trust for partial support. K.M.H. thanks the Academy of Finland (Grant no. 135439), the Finnish Cultural Foundation, and Jenny and Antti Wihuri Foundation for financial support.

References and notes (17)

  • M.G. Lichtenheld et al.

    Nature

    (1988)
  • P.J. Peters et al.

    J. Exp. Med.

    (1991)
  • C.J. Rosado et al.

    Science

    (2007)
  • D. Kagi et al.

    Nature

    (1994)
  • I. Voskoboinik et al.

    J. Biol. Chem.

    (2005)
  • K. Baran et al.

    Immunity

    (2009)
  • R.H.P. Law et al.

    Nature

    (2010)
  • J.C. Choy

    Cell Death Differ.

    (2010)
There are more references available in the full text version of this article.

Cited by (28)

  • Pore-forming proteins and their role in cancer and inflammation: Mechanistic insights and plausible druggable targets

    2022, Chemico-Biological Interactions
    Citation Excerpt :

    The discovery of a novel class of 1-amino-2,4-dicyanopyrido [1,2-a]benzimidazoles as direct inhibitors of perforin overcame these pitfalls later. Analogs with O-linked side chains 3 and amide-linked side chains including its NMe analogs 4,and 5 exhibited an optimized solubility considering steric factors also in the frame [121]. Compared to dihydro[3,4-c]pyridinone (IC50 = 0.97 μM), aryl substituted isobenzofuran-1(3H)-one produced consistent inhibitory activity (C; IC50 = 6.20 μM).

  • Substituted arylsulphonamides as inhibitors of perforin-mediated lysis

    2017, European Journal of Medicinal Chemistry
    Citation Excerpt :

    The one unexpected outlier was the 3-cyano compound 49 which, although it had poor activity against isolated recombinant perforin, had excellent activity against perforin produced by whole NK cells. The NK cells also retained excellent viability across all examples, consistent with our previous findings for this series [32] and in contrast to earlier reported classes [26–28]. Preliminary physicochemical data was collected on the same (active) subset of compounds in order to assess their potential for progression to in vivo pharmacokinetic (PK) studies (Table 5).

View all citing articles on Scopus
View full text