Aminobenzosuberone derivatives as PfA-M1 inhibitors: Molecular recognition and antiplasmodial evaluation

https://doi.org/10.1016/j.bioorg.2020.103750Get rights and content

Highlights

Abstract

Aminobenzosuberone-based PfA-M1 inhibitors were explored as novel antimalarial agents against two different Plasmodium falciparum strains. The 4-phenyl derivative 7c exhibited the most encouraging growth inhibitory activity with IC50 values of 6.5–11.2 µM. X-ray crystal structures and early assessment of DMPK/ADME-Tox parameters allowed us to initiate structure-based drug design approach and understand the liabilities (such as potential metabolic and aqueous solubility issues) as well as identify the opportunities for improvement of this aminobenzosuberone series. It also suggested that compound 7c should be regarded as an attractive chemical tool to investigate the different biological roles of this multifunctional PfA-M1 protein.

Introduction

Despite encouraging wide-scale reduction in malaria incidence and mortality rates, malaria is still one of the most severe public health problems worldwide. It is a leading cause of death and disease in many developing countries particularly in young children. According to the WHO world malaria report 2019, there were an estimated 228 million cases of malaria and 405 000 related deaths in 2018 [1]. The regular emergence of antimalarial drugs resistance threatens the global malaria control and elimination effort, especially since artemisinin-resistant strains of Plasmodium falciparum were observed in the Greater Mekong region [2]. There is an urgent need for the discovery of novel Plasmodium molecular targets and original antimalarial agents with new modes of action to combat malaria.

P. falciparum proteases have appeared as promising targets for the development of new classes of therapeutic agents against malaria. They are involved in a number of vital pathways which are essential for parasite survival: red blood cell (RBC) invasion, hemoglobin (Hb) degradation, egress [3]. Among these pathways, Hb degradation provides amino acids necessary to the metabolism and survival of the parasites inside erythrocytes [4], [5], [6], [7], and also reduces the colloid-osmotic pressure within the host RBCs and thus preventing their premature lysis [8], [9].

One of these attractive protease targets is the Plasmodium falciparum M1 aminopeptidase, PfA-M1. This zinc-dependent metallo-aminopeptidase, belonging to the M1 family of aminopeptidases, catalyzes the cleavage of amino-acids at the N-terminus of peptides and plays a major role in the Plasmodium life cycle [10], [11], [12], [13], [14], [15], [16], [17], [18]. It has been mainly involved during asexual blood stage development in the last step of hemoglobin breakdown by hydrolyzing dipeptides into essential amino-acids. Contrary to other endopeptidases also involved in this Hb digestion, PfA-M1 is encoded by a single copy gene (MAL13P1.56 or PF3D7_1311800) [19], [20], [21] and possesses non-redundant function with no overlapping substrate specificity with other hemoglobinases [7], [22], [23]. The disruption of PfA-M1 gene has been unsuccessful until now, suggesting that this protease is important for parasite growth/multiplication [18], [19], [24], [25]. Moreover, in P. berghei most proteases involved in the Hb degradation pathway could be deleted except the orthologues of P. falciparum falcilysin and PfA-M1 [26]. According to omics data (Table S1), PfA-M1 has been reported in other developmental stages of the parasite’s life cycle, as it is expressed in merozoite, gametocyte and insect stages [15], [27], [28], [29], [30], [31], [32], [33], [34], [35]. Besides its involvement in Hb digestion, PfA-M1 might thus also play alternative roles in RBC invasion, egress or during gametogenesis [10], [12], [13], [18]. Currently considered as a promising drug target candidate, PfA-M1 was also recently proposed as a possible diagnostic biomarker for the presence of malaria parasite in young patients, displaying immunologic characteristics similar to those of some vaccine candidates (AMA1, MSP1, CSP) [36].

Given these results, PfA-M1 seems to be an essential protein and hence, a very attractive validated target for the development of novel antiplasmodial drugs [3], [19], [37], [38]. In addition, blocking PfA-M1 catalytic activity with low molecular weight inhibitors is fatal for the parasite without major toxicities for the host [16], [39].

Most of the PfA-M1 inhibitors (Table 1) consist of tetrahedral intermediate mimics, such as the well-known aminopeptidase inhibitor bestatin 1, or as phosphinic acid 2, or zinc-chelating group inserted in a peptide-like scaffold, such as hydroxamic acids 35. Despite demonstrating nanomolar to submicromolar activities in vitro on the enzyme, these compounds were usually 100 to 1000 fold less potent on in cellulo parasite growth inhibition assay (Table 1). Only, a handful of PfA-M1 inhibitors (1, 2 and 4) have been evaluated in vivo and demonstrated a reduction or suppression of the parasitaemia in murine models [24], [40]. However, their frequent lack of selectivity regarding other metallo-aminopeptidase families remains problematic. Indeed, most inhibit more potently PfA-M17, a bimetallic aminopeptidase, which is also considered as a promising antimalarial target [38], [39], [41], [42]. This dual activity is a valuable asset in today’s malaria eradication context, but it questions the relative involvement and importance of each enzyme in the parasite biology.

In this context, our research group has previously described the development of potent and selective aminobenzosuberone-based inhibitors of APN/CD13 [43], [44], [45], a human M1 aminopeptidase involved in angiogenesis and tumor metastasis [46], [47].

Recently, we reported that our scaffold 7c demonstrated potent in vitro inhibitory activity and was selective for rPfA-M1 over rPfA-M17 [48]. This compound was progressed to a 4-day suppressive test of Peters using a non-lethal parasite model P.c.chabaudi and was able to reduce the parasitaemia by 44% with a daily dose of 12 mg/kg, in four daily intraperitoneal injections, further strengthening the potential of PfA-M1 as a target for antimalarial research [49].

We report herein our initial research effort towards the identification and selection of the 4-phenyl aminobenzosuberone derivative 7c for in vivo efficacy assay. We also describe the crystal structure of rPfA-M1 in complex with aminobenzosoberone derivatives 7c and 7h.

Section snippets

Chemistry

Racemic substituted aminobenzosuberone hydrochloride derivatives 7a-j were synthesized according to literature procedure [44].

In vitro Plasmodium growth inhibition and toxicity

The encouraging results, regarding PfA-M1 inhibition [48], prompted us to pursue the evaluation of this series of aminobenzosuberone scaffold as novel antimalarial agent using in vitro parasite growth inhibition assays. The antiplasmodial evaluation was carried out on 3D7 and FcB1 P. falciparum strains. The cytotoxicity of our derivatives was assessed on Vero cells (

Conclusion

A series of aminobenzosuberones has been investigated as novel antiplasmodial agents targeting the Plasmodium falciparum M1 aminopeptidase. This series was found to be highly potent and selective of rPfA-M1 with low micromolar parasite growth inhibitory activities. The co-crystal structures of 7c and 7 h were obtained and resolved allowing us to determine the binding mode of aminobenzosuberones within the rPfA-M1 active site, paving the way for further optimization through structure-based drug

Cloning, expression and purification of recombinant PfA-M1

As described in literature[49], a gene encoding the cleavage site for the tobacco etch virus (TEV) protease fused to residues 192–1085 of native Plasmodium falciparum alanyl aminopeptidase PfA-M1 (PlasmoDB PF3D7_1311800) was synthetized by Genecust (Luxembourg), with the help of algorithms developed for optimizing, DNA sequences to improve protein expression in Escherichia coli. This synthetic gene was cloned into the T7 expression pET45b(+) vector (Novagen), which appended an N-terminal

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

The support of the Université de Haute-Alsace and the Ecole Nationale Supérieure de Chimie de Mulhouse is gratefully acknowledged. We also wish to thank the Agence Nationale de la Recherche for its financial support (ANR-12-BS07-0020-01). The authors acknowledge the support and use of the resources of the French Infrastructure for Integrated Structural Biology (FRISBI) ANR-10-INBS-05 and of Instruct-ERIC. This study was supported by the grant ANR-10-LABX-0030-INRT, a French State fund managed

References (77)

  • F. Silvestrini et al.

    Protein Export Marks the Early Phase of Gametocytogenesis of the Human Malaria Parasite Plasmodium falciparum

    Mol. Cell. Proteomics

    (2010)
  • L. Florens et al.

    Proteomics approach reveals novel proteins on the surface of malaria-infected erythrocytes

    Mol. Biochem. Parasitol.

    (2004)
  • S.E. Lindner et al.

    Total and Putative Surface Proteomics of Malaria Parasite Salivary Gland Sporozoites

    Mol. Cell. Proteomics

    (2013)
  • T.S. Skinner-Adams et al.

    Plasmodium falciparum neutral aminopeptidases: new targets for anti-malarials

    Trends Biochem. Sci.

    (2010)
  • C.M. Stack et al.

    Characterization of the Plasmodium falciparum M17 Leucyl Aminopeptidase a protease involved in amino acid regulation with potential for antimalarial drug development

    J. Biol. Chem.

    (2007)
  • S. Albrecht et al.

    Amino-benzosuberone: A novel warhead for selective inhibition of human aminopeptidase-N/CD13

    Bioorg. Med. Chem.

    (2011)
  • C. Maiereanu et al.

    A novel amino-benzosuberone derivative is a picomolar inhibitor of mammalian aminopeptidase N/CD13

    Bioorg. Med. Chem.

    (2011)
  • C. Schmitt et al.

    Selective aminopeptidase-N (CD13) inhibitors with relevance to cancer chemotherapy

    Bioorg. Med. Chem.

    (2013)
  • P. Mina-Osorio

    The moonlighting enzyme CD13: old and new functions to target

    Trends Mol. Med.

    (2008)
  • N. Drinkwater et al.

    Potent dual inhibitors of Plasmodium falciparum M1 and M17 aminopeptidases through optimization of S1 pocket interactions

    Eur. J. Med. Chem.

    (2016)
  • J. González-Bacerio et al.

    KBE009: An antimalarial bestatin-like inhibitor of the Plasmodium falciparum M1 aminopeptidase discovered in an Ugi multicomponent reaction-derived peptidomimetic library

    Bioorg. Med. Chem.

    (2017)
  • D. Osella et al.

    On the mechanism of the antitumor activity of ferrocenium derivatives

    Inorg. Chim. Acta

    (2000)
  • P.A. Karplus et al.

    Assessing and maximizing data quality in macromolecular crystallography

    Curr. Opin. Struct. Biol.

    (2015)
  • B.W. Matthews

    Solvent content of protein crystals

    J. Mol. Biol.

    (1968)
  • Y. Zhang et al.

    PKSolver: An add-in program for pharmacokinetic and pharmacodynamic data analysis in Microsoft Excel

    Comput. Methods Programs Biomed.

    (2010)
  • WHO | World malaria report 2019, (n.d.). https://www.who.int/publications-detail/world-malaria-report-2019 (accessed...
  • A.M. Dondorp et al.

    Artemisinin Resistance in Plasmodium falciparum Malaria

    N. Engl. J. Med.

    (2009)
  • M. Mishra et al.

    Structural Insights Into Key Plasmodium Proteases as Therapeutic Drug Targets

    Front. Microbiol.

    (2019)
  • J. Liu et al.

    Plasmodium falciparum ensures its amino acid supply with multiple acquisition pathways and redundant proteolytic enzyme systems

    Proc Natl Acad Sci USA

    (2006)
  • D.A. Elliott et al.

    Four distinct pathways of hemoglobin uptake in the malaria parasite Plasmodium falciparum

    Proc. Nat. Acad. Sci. (USA)

    (2008)
  • D.E. Goldberg, Hemoglobin Degradation, in: R.W. Compans, M.D. Cooper, T. Honjo, H. Koprowski, F. Melchers, M.B.A....
  • M.B. Harbut et al.

    Bestatin-based chemical biology strategy reveals distinct roles for malaria M1- and M17-family aminopeptidases

    PNAS

    (2011)
  • O. Azimzadeh et al.

    Plasmodium falciparum PfA-M1 aminopeptidase is trafficked via the parasitophorous vacuole and marginally delivered to the food vacuole

    Malar. J.

    (2010)
  • M. Allary et al.

    Properties, stage-dependent expression and localization of Plasmodium falciparum M1 family zinc-aminopeptidase

    Parasitology

    (2002)
  • M.F. Nankya-Kitaka et al.

    Plasmodium chabaudi chabaudi and P. falciparum : inhibition of aminopeptidase and parasite growth by bestatin and nitrobestatin

    Parasitol Res. 84

    (1998)
  • K.R. Trenholme et al.

    Aminopeptidases of Malaria Parasites: New Targets for Chemotherapy

    IDDT.

    (2010)
  • M.J. Gardner et al.

    Genome sequence of the human malaria parasite Plasmodium falciparum

    Nature

    (2002)
  • C. Aurrecoechea et al.

    PlasmoDB: a functional genomic database for malaria parasites

    Nucleic Acids Res.

    (2009)
  • Cited by (10)

    • Structure-based development of potent Plasmodium falciparum M1 and M17 aminopeptidase selective and dual inhibitors via S1′-region optimisation

      2023, European Journal of Medicinal Chemistry
      Citation Excerpt :

      These findings are in alignment with the previous generation of hydroxamate-based inhibitors, in which it was found that compounds that included the methylene spacer usually outperformed their methylene-lacking counterparts [11]. We have previously shown that the replacement of the tert-butyl group present in 1 and 2 with cyclobutyl or cyclopentyl is reasonably well tolerated in terms of both PfA-M1 and PfA-M17 inhibitory activity [12]. Heteroatomic derivatisation of the cyclobutyl at various positions largely resulted in reduction of inhibitory activity (9j-w).

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    2

    Current address: Laboratoire Vigne, Biotechnologies et Environnement, UPR3991, Université de Haute-Alsace, Université de Strasbourg, Colmar, France.

    View full text