Elsevier

European Journal of Medicinal Chemistry

Volume 164, 15 February 2019, Pages 423-439
European Journal of Medicinal Chemistry

Research paper
Exploration of novel macrocyclic dipeptide N-benzyl amides as proteasome inhibitors

https://doi.org/10.1016/j.ejmech.2018.12.072Get rights and content

Highlights

  • A series of macrocyclic dipeptide N-benzyl amides were designed.

  • Most compounds exhibited potent proteasome inhibition.

  • They also showed excellent anti-proliferative activity against blood cancer cells.

  • Macrocyclic dipeptides were more stable than their corresponding linear analogues.

  • Docking study was performed to analyze the binding mode of 23h within proteasome.

Abstract

As proteasome inhibitors, a series of novel macrocyclic dipeptide N-benzyl amides were designed, synthesized and evaluated. Most of them exhibited potent proteasome inhibition and excellent anti-proliferative activity against RPMI 8226, MM1S, and MV-4-11 cell lines. As the most distinguished one among this series, compound 23h displayed potent and selective proteasome inhibitory potency (IC50: β5c = 29 nM, β5i = 35 nM, β1c, β2c, β1i, β2i > 10 μM), excellent anti-proliferative activity against RPMI 8226, MM1S, and MV-4-11 cell lines with IC50 values of 18 nM, 15 nM, and 21 nM, respectively, as well as favorable metabolic stability in human liver microsomes (HLMs), highlighting that it is a promising lead compound for further development of proteasome inhibitors.

Introduction

The eukaryotic 26S proteasome is a large (1.6–2.4 MDa) ATP-dependent proteolytic complex, composed of a cylindrical 20S core particle (CP) capped by two 19S regulatory complexes [1]. The 20S proteasome is the proteolytically active key element of the ubiquitin proteasome system (UPS) that directs the majority of intracellular protein degradation in eukaryotic cells. It is composed by four heptameric rings stacked in a α7β7β7α7 arrangement and contains three proteolytic subunits, β1, β2, and β5 [2], respectively. There are two main proteasome subtypes: the constitutive proteasome (cCP) which contains three catalytic subunits denoted as β1c, β2c, and β5c and the immunoproteasome (iCP) which contains three catalytic subunits denoted as β1i, β2i, and β5i [3]. Inhibition of the 20S proteasome leads to the accumulation of substrate proteins involved in signal transduction, antigen presentation, cell-cycle progression and apoptosis, and is preferentially cytotoxic to cancer cells [[4], [5], [6], [7], [8]]. Indeed, proteasome inhibitors are recognized as clinically effective anti-cancer agents, primarily for hematological malignancies [9]. To date, three proteasome inhibitors Bortezomib, Carfilzomib, and Ixazomib (Fig. 1) are FDA approved for the treatment of multiple myeloma (MM) and mantle cell lymphoma [[10], [11], [12]]. All three are covalent inhibitors with an electrophilic warhead at the C-terminal end of a peptidyl backbone for covalent attachment to the catalytic Thr1 residues of the proteasome [13]. However, the electrophilic warhead is often related to excessive reactivity, lack of specificity and instability, which is believed to be the major cause of side effects during therapy [12,14]. These inhibitors have also been unsuccessful in the treatment of solid cancers [15,16], with the lack of efficacy most likely due to their covalent binding to the proteasome, limiting their widespread tissue distribution. In contrast, non-covalent proteasome inhibitors do not possess an electrophilic warhead with less reactivity. Non-covalent and reversible binding mode ensure them with rapid binding and dissociation kinetics. These features may allow non-covalent inhibitors to overcome drawbacks arising in therapeutics of covalent ones.

Representative peptidyl non-covalent proteasome inhibitors have been reported. TMC-95A (4, Fig. 2) is a macrocyclic natural product isolated from Apiospora montagnei Sacc TC 1093, which potently inhibits all the three proteasome catalytic activities with preference for β5c activity (IC50 = 5.4 nM) [17,18]. However, the structural complexity of TMC-95A maybe one of important obstacle to its further development. Additionally, a series of 5-methoxy-1-indanone di-peptide benzyl amides have been reported, including CVT-659 (5, Fig. 2) which selectively inhibits β5c site with submicromolar potency (IC50 = 0.14 μM), but this compound displayed poor cellular activity (IC50 = 8 μM) [19]. Researchers from Millennium Pharmaceuticals, Inc. identified a series of di- and tripeptides (e. g. 6 and 7, Fig. 2) with potency and selectivity for both constitutive proteasome and immunoproteasome β5 sites [20]. Moreover, the X-ray structures of the inhibitors in complex with proteasome suggest that the occupancy of S1 and S3 pockets was crucial to inhibitor potency. Of note, the size of the hydrophobic benzyl group was well suited to S1 pocket, which may be a reason for the wide use of various substituted benzyl groups in peptidyl non-covalent proteasome inhibitors. Meanwhile, the 2-(neopentylamino)-2-oxoethyl group (as present in compound 6) has been reported to provide a near-optimal fit for the S3 binding pocket [[20], [21], [22]]. Other reported peptidyl non-covalent proteasome inhibitors include 2-aminobenzylstatine derivatives [23,24] and linear TMC-95A analogues [25,26]. Overall, linear peptide-based proteasome inhibitors remain to be the mainstream of this field [13,21,[27], [28], [29], [30]].

However, linear peptides are always thought to be unstable with unsatisfied pharmacokinetic profiles. In contrast, macrocyclic peptides often show distinct and (in comparison to linear peptides) superior pharmacokinetic and pharmacological properties [31,32]. Desirable properties introduced by the macrocycle include increased metabolic stability, cellular penetration and selectivity [[33], [34], [35]]. We have identified a series of oral and potent macrocyclic dipeptide epoxyketones as covalent proteasome inhibitors in our previous work [36]. Due to the disadvantages of the electrophilic warhead discussed, in this manuscript, we explored novel proteasome inhibitors based on the peptidyl macrocyclic skeleton of macrocyclic dipeptide epoxyketones, replacing the electrophilic warhead with substituted benzyl groups (Fig. 3). During further optimization, a series of macrocyclic dipeptide N-benzyl amides were designed and synthesized. Moreover, the anti-proliferative activity against various cancer cells of selected compounds were evaluated. In addition, we explored the differences in metabolic stability between our macrocyclic dipeptides and the corresponding linear analogues.

Section snippets

Chemistry

All the target compounds were synthesized by following routes as described in Scheme 1, Scheme 2, Scheme 3, Scheme 4, Scheme 5. Scheme 1, Scheme 2 show the synthetic routes of the intermediates which would be used in Scheme 3, Scheme 4, Scheme 5. Scheme 3, Scheme 4 show the synthetic routs of macrocyclic dipeptides 23a-23j and 31. Finally, Scheme 5 shows the synthetic rout of linear analogue 33.

The synthetic routes of terminal alkene-containing carboxylic intermediates 10a - 10e are displayed

Conclusions

In this study, a series of novel macrocyclic dipeptide N-benzyl amides were rationally designed, synthesized, and biologically evaluated. Most of them exhibited potent proteasomal inhibition, as well as excellent anti-proliferative activity against RPMI 8226, MM1S, and MV-4-11 cell lines. Compound 23h and 23i displayed potent and selective inhibition against β5c and β5i. Further studies found that compound 23h was more potent and more stable than its corresponding linear analogues, proving that

Experimental section

1H and 13C NMR spectra were recorded on Brüker 500 MHz spectrometer (Brüker Bioscience, Billerica, MA, USA) with CDCl3 or DMSO‑d6 as solvent. Chemical shifts (d) were reported in parts per million (ppm) relative to internal TMS, and coupling constants (J) were reported in Hertz (Hz). Splitting patterns were designated as singlet (s), broad singlet (brs), doublet (d), double doublet (dd), triplet (t), quartet (q) and multiplet (m). Melting points were determined using a Buchi B-540 capillary

Acknowledgments

We thank Jianyang Pan (Research and Service Center, College of Pharmaceutical Sciences, Zhejiang University) for performing NMR spectrometry for structure elucidation. This work was supported by grants from the key project of Zhejiang Provincial Natural Science Foundation of China (LZ15H300001), and the Science and Technology Commission of Shanghai Municipality (17431903000).

References (48)

  • F. Parlati et al.

    Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome

    Blood

    (2009)
  • R. Ettari et al.

    Immunoproteasome-selective and non-selective inhibitors: a promising approach for the treatment of multiple myeloma

    Pharmacol. Therapeut.

    (2018)
  • J. Yu et al.

    Design, synthesis, and biological evaluation of novel phenol ether derivatives as non-covalent proteasome inhibitors

    Eur. J. Med. Chem.

    (2019)
  • J. Adams

    The proteasome: a suitable antineoplastic target

    Nat. Rev. Canc.

    (2004)
  • J.D. Etlinger et al.

    Soluble atp-dependent proteolytic system responsible for degradation of abnormal proteins in reticulocytes

    Proc. Natl. Acad. Sci. U. S. A

    (1977)
  • A. Ciechanover

    The ubiquitin–proteasome pathway: on protein death and cell life

    EMBO J.

    (1998)
  • M. Muratani et al.

    How the ubiquitin–proteasome system controls transcription

    Nat. Rev. Mol. Cell. Biol.

    (2003)
  • W. Lee et al.

    The immunoproteasome: an emerging therapeutic target

    Curr. Top. Med. Chem.

    (2011)
  • E.E. Manasanch et al.

    Proteasome inhibitors in cancer therapy

    Nat. Rev. Clin. Oncol.

    (2017)
  • P. Moreau et al.

    Oral Ixazomib, lenalidomide, and dexamethasone for multiple myeloma

    N. Engl. J. Med.

    (2016)
  • K. Redic

    Carfilzomib: a novel agent for multiple myeloma

    J. Pharm. Pharmacol.

    (2013)
  • D. Chen et al.

    Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives

    Curr. Cancer Drug Targets

    (2011)
  • N. Micale et al.

    Peptide-based proteasome inhibitors in anticancer drug design

    Med. Res. Rev.

    (2014)
  • H. Wang et al.

    An analysis of the safety profile of proteasome inhibitors for treating various cancers

    Expert Opin. Drug Saf.

    (2014)
  • Cited by (4)

    • Pseudopeptides with aldehyde or vinylsulfone warheads: Synthesis and antiproteasomal activity

      2021, Bioorganic Chemistry
      Citation Excerpt :

      Unlike bortezomib, ixazomib has improved pharmacokinetics and pharmacodynamics that make it advantageous for therapy [16]. Numerous recent studies have analysed how the incorporation of non-proteinogenic amino acids and a variety of terminal headgroups affect cell permeability, potency to different proteasomal subunits and preference for specific types of proteasomes [9,12,17–20]. On the other hand, only a small number of comparative studies on various functional reactive groups have been published to date.

    1

    Equal contribution.

    View full text