Elsevier

Pharmacology & Therapeutics

Volume 93, Issues 2–3, February–March 2002, Pages 125-133
Pharmacology & Therapeutics

Associate editor: D. Shugar
Structure-based design of cyclin-dependent kinase inhibitors

https://doi.org/10.1016/S0163-7258(02)00182-1Get rights and content

Abstract

The eukaryotic cell cycle is tightly regulated by the sequential activation and deactivation of the cyclin-dependent kinases (CDKs). Aberrant CDK activity is a common defect in human tumours, and clinically, it often confers a poor prognosis. The strong genetic link between CDKs and the molecular pathology of cancer has provided the rationale for developing small-molecule inhibitors of these kinases. X-ray crystallography recently has revealed the molecular details of CDK regulation by cyclin binding and phosphorylation, and by complex formation with endogenous inhibitors. Knowledge of the structure of CDK2 has been key in driving the design and development of a large number of ATP competitive inhibitors. Crystallography has revealed that the ATP-binding site of CDK2 can accommodate a number of diverse molecular frameworks, exploiting various sites of interaction. In addition, residues outside the main ATP-binding cleft have been identified that could be targeted to increase specificity and potency. These results suggest that it may be possible to design pharmacologically relevant ligands that act as specific and potent inhibitors of CDK activity. We provide a review of the current state of the field, along with an overview of our current inhibitor design studies.

Introduction

Attention is increasingly being focussed on the cell cycle as a potential target for therapeutic intervention in several proliferative diseases, including cancer. Timely and coordinated progression through the cell cycle is controlled by the sequential activation and deactivation of members of the cyclin-dependent kinase (CDK) family (Morgan, 1997). Disrupted cell-cycle control due to aberrant CDK activity has been linked directly to the molecular pathology of cancer Kamb, 1995, Vogt & Reed, 1998, Pavletich, 1999. For example, loss of function of endogenous inhibitors such as p16INK4A, over-expression of cyclin D1 and CDK4, and alterations to CDK substrates such as retinoblastoma gene product (pRb) are frequently observed in human tumours Shapiro & Harper, 1999, Senderowicz & Sausville, 2000.

Members of the CDK family are active at distinct points in the cell cycle (Morgan, 1997). The G0/G1 transition is characterised by increased transcription of cyclin D, giving rise to activation of CDK4 and CDK6, whose primary target is pRb. In its hypophosphorylated state, pRb binds to the E2F transcription factor, repressing its activity at promoter sites. pRb hyperphosphorylation disrupts these complexes, permitting the transcription of several genes whose products are associated with S-phase progression. General targets include cyclin E, dihydrofolate reductase, and DNA polymerases Dyson, 1998, Zhang et al., 1999, Harbour & Dean, 2000. Alterations in the level of CDK4 or CDK6 activity in malignant cells have suggested these kinases as important targets for therapeutic intervention. However, inhibition of CDK4/6 by p16INK4A in cells lacking pRb does not lead to cell-cycle arrest (Shapiro & Harper, 1999).

CDK2 is essential for G1 progression and S-phase entry. Complexed with cyclin E, it sustains pRb hyperphosphorylation to support progression through G1 into S-phase. Early in S-phase, CDK2 associates with cyclin A, where it plays a role in inactivating E2F, and it is required for the completion of S-phase. Persistence of E2F activity during S-phase leads to apoptosis Krek et al., 1995, Shapiro & Harper, 1999, and, therefore, selective inhibition of CDK2-cyclin A may achieve cytotoxicity instead of cell-cycle arrest. Inhibition in S-phase may also be a strategy to which transformed cells are particularly sensitive due to their higher levels of E2F (Chen, 1999).

These studies have provided the rationale for developing small-molecule CDK inhibitors as anticancer agents (Hall & Peters, 1996). Interfering with CDK catalytic activity through ATP-competitive ligands has proved a successful strategy. A disadvantage of this approach is the large number of protein kinases found within the cell Consortium, 2001, Venter, 2001, most of which share a high degree of sequence similarity within the active site. Nevertheless, highly potent and selective ATP-competitive inhibitors have been identified for a number of other kinases. The epidermal growth factor receptor tyrosine kinase inhibitor PD 153035 (Fry et al., 1994) and the p38 mitogen-activated protein kinase (MAPK) inhibitor SB 203580 (Lee et al., 1999) are examples.

Section snippets

Structural overview of cyclin-dependent kinase 2

Structure-based CDK inhibitor design has been driven forward predominantly by studies on monomeric CDK2. Human CDK2 encodes little more than the conserved catalytic core domain, which contains the classic bi-lobal kinase fold De Bondt et al., 1993, Morgan, 1997. The N-terminal domain is composed mainly of β-sheet, containing five anti-parallel β-strands, and one helix (the C-helix). The larger C-terminal domain is predominantly α-helical, and is linked to the N-terminal domain by a flexible

Cyclin-dependent kinase 2 inhibitor design

The CDK2 ATP-binding site may be conceptually divided into four main subsites that might be targeted for the development of potent and specific inhibitors (Fig. 2). The interactions formed in each subsite for a selection of inhibitors are discussed in the following sections.

Overview

The majority of the structural studies to date have focused on exploiting interactions formed between inhibitors and monomeric CDK2 to increase potency and specificity. However, monomeric CDK2 is inactive, association with cyclin A and phosphorylation on Thr160 being prerequisites for its full activity Jeffrey et al., 1995, Brown et al., 1999, Endicott et al., 1999. Activation of CDK2 involves various conformational changes, including the reorientation of key catalytic residues involved in

Conclusions

CDK2 is now recognised as an important target for the design of novel antitumour therapies. Structural feedback in the rational design of CDK inhibitors has been derived in the main from studies of monomeric CDK2/inhibitor complexes. These structures have shown that a large number of diverse compounds have the necessary ability to satisfy the hydrogen-bonding potential of the kinase hinge region in addition to complementing the shape and chemistry of the cleft. These diverse backbones provide

Acknowledgements

We would like to acknowledge the seminal contributions made by D.R. Newell, R. Griffin, H. Calvert, B. Golding, N. Curtin, and members of the Anticancer Drug Discovery Initiative at Newcastle University, UK and by T. Boyle and P. Jewsbury at AstraZeneca Pharmaceuticals, UK. We would also like to thank colleagues at the LMB for their support and encouragement with this work, in particular N. Brown, E. Garman, A. Lawrie, J. Tucker, P. Tunnah, and R. Bryan. We acknowledge with gratitude the help

References (44)

  • M.E.M. Noble et al.

    Chemical inhibitors of cyclin-dependent kinases: insights into design from X-ray crystallographic studies

    Pharmacol. Ther

    (1999)
  • N. Pavletich

    Mechanisms of cyclin-dependent kinase regulation: structure of cdks, their cyclin activators, and Cip and INK4 inhibitors

    J Mol Biol

    (1999)
  • H.S. Zhang et al.

    Active transcriptional repression by the Rb-E2F complex mediates G1 arrest triggered by p16INK4a, TGFβ and contact inhibition

    Cell

    (1999)
  • C.E. Arris et al.

    Identification of novel purine and pyrimidine cyclin-dependent kinase inhibitors with distinct molecular interactions and tumor cell growth inhibition profiles

    J. Med. Chem.

    (2000)
  • H.-J. Böhm

    Prediction of binding constants of protein ligands: a fast method for the prioritization of hits obtained from de novo design or 3D database search programs

    J. Comput-Aided Mol Des

    (1998)
  • N.R. Brown et al.

    The structural basis for specificity of substrate and recruitment peptides for cyclin-dependent kinases

    Nat Cell Biol

    (1999)
  • C.J. Bruns et al.

    Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma

    Cancer Res

    (2000)
  • Y. Chen

    Selective killing of transformed cells by cyclin/cyclin-dependent kinase 2 antagonists

    Proc. Natl. Acad. Sci. USA

    (1999)
  • I.H.G. Consortium

    Initial sequencing and analysis of the human genome

    Nature

    (2001)
  • S. Davis et al.

    Prevention of chemotherapy-induced alopecia in rats by CDK inhibitors

    Science

    (2001)
  • W.F. De Azevedo et al.

    Inhibition of cyclin-dependent kinases by purine analogues. Crystal structure of human cdk2 complexed with roscovitine

    Eur. J. Biochem.

    (1997)
  • W.F. De Azevedo et al.

    Structural basis for specificity and potency of a flavonoid inhibitor of human CDK2, a cell cycle kinase

    Proc. Natl. Acad. Sci. USA

    (1996)
  • Cited by (96)

    • Imidazo[1,2-c]pyrimidin-5(6H)-one inhibitors of CDK2: Synthesis, kinase inhibition and co-crystal structure

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      However, comparative structural analyses have revealed minor differences, which offer some space to be exploited to successfully differentiate CDK1 from other CDKs in inhibitor design [12,13]. Compounds possessing selectivity include NU6102 originally described as a CDK1/2 inhibitor [14], but following studies confirmed its preference for CDK2 [15,16], or compound 73 (CAS 2079895-42-2), currently the most selective CDK2 inhibitor available [12]. Interestingly, these nanomolar CDK2 inhibitors display no or only weak antiproliferative activity [12].

    • Identification of novel inhibitors against Cyclin Dependent Kinase 9/Cyclin T1 complex as: Anti cancer agent

      2017, Saudi Journal of Biological Sciences
      Citation Excerpt :

      To date only 11 kinase inhibitors received US Food and Drug Administration approval as Cancer treatments (Zhang et al., 2009). Structure Based Drug Design for development of Cell cycle inhibitor is the right approach for the Cancer therapy (Davies et al., 2002; Dickson and Schwartz, 2009). Hence, it is very important to develop and discovery some novel inhibitors, which can bind to the ATP binding site of the CDK9 kinase and reduce some of the life-threatening diseases like Cancer, AIDS, Cardiac Hypertrophy and several Proliferative diseases.

    View all citing articles on Scopus
    View full text