Elsevier

Biochemical Pharmacology

Volume 107, 1 May 2016, Pages 1-13
Biochemical Pharmacology

Research update
Targeting transcription factors by small compounds—Current strategies and future implications

https://doi.org/10.1016/j.bcp.2015.12.006Get rights and content

Abstract

Transcription factors are central regulators of gene expression and critically steer development, differentiation and death. Except for ligand-activated nuclear receptors, direct modulation of transcription factor function by small molecules is still widely regarded as “impossible”.

This “un-druggability” of non-ligand transcription factors is due to the fact that the interacting surface between transcription factor and DNA is huge and subject to significant changes during DNA-binding. Besides some “success studies” with compounds that directly interfere with DNA binding, drug targeting approaches mostly address protein–protein interfaces with essential co-factors, transcription factor dimerization partners, chaperone proteins or proteins that regulate subcellular shuttling. An alternative strategy represent DNA-intercalating, alkylating or DNA-groove-binding compounds that either block transcription factor-binding or change the 3D-conformation of the consensus DNA-strand. Recently, much interest has been focused on chromatin reader proteins that steer the recruitment and activity of transcription factors to a gene transcription start site. Several small compounds demonstrate that these epigenetic reader proteins are exciting new drug targets for inhibiting lineage-specific transcription in cancer therapy.

In this research update we will discuss recent advances in targeting transcription factors with small compounds, the challenges that are related to the complex function and regulation of these proteins and also the possible future directions and applications of transcription factor drug targeting.

Introduction

For a long time the classical targets for improving cancer treatment have been receptors, kinases or other proteins involved in signal transduction, whereas transcription factors have long been considered as un-druggable targets. However, transcription factors are the central regulators of gene transcription and a large number of diseases, such as neurodegenerative disorders, diabetes and also cancer are associated with the deregulation of transcriptional networks. In fact it has been estimated that transcription factors account for 20% of oncogenes in cancer [1]. The understanding of these complex networks and pharmacological strategies to modulate the activity of distinct transcription factors will therefore be essential for the development of novel therapeutic approaches.

Most current strategies to modulate gene expression during e.g., cancer treatment indirectly affect transcription factors activity, since the inhibition of upstream kinases by specific small molecules results in modulation of multiple downstream pathways and therefore usually does not affect one single transcription factor. To further improve specific therapeutic intervention, minimize side effects and develop a “patient-specific therapy” the interest in directly targeting transcription factors has increased, since effective manipulation of these regulators may allow a “transcriptome-specific” therapy.

A proof for the relevance of directly modulating transcription factors in various therapies, most prominent cancer therapy, has been provided since many years/decades by targeting nuclear receptors that contain a clearly defined ligand-binding pocket and are activated by natural ligands, such as retinoic acid-, glucocorticoid-, estrogen-, or androgen-receptors. Activation or inhibition of these transcription factors is a central aspect of many standard cancer therapies as it leads to tumor cell type specific cell death and growth inhibition and the primary response to these therapeutics is of significant prognostic value. In fact e.g., the use of glucocorticoids in childhood leukemia represented a real therapeutic success story since its introduction 50 years ago. The use of these hormones/synthetic ligands is nowadays indispensable in cancer therapy as exemplified by the pronounced effects of glucocorticoids in leukemia, anti-estrogens in breast cancer or anti-androgens in prostate cancer therapy. However, almost all other transcription factors that lack pockets for activating ligands were ignored for drug discovery strategies and considered as “un-druggable”—a frequent comment from reviewers when grant proposals on this topic were rejected. One reason for neglecting these essential central regulators of life and death lies in the lack of a typical small binding pocket and the fact that usually the only clearly defined ligand is the cognate DNA-consensus sequence. In contrast to a steroid- or ATP-binding pocket this “ligand” requires a huge protein surface as interaction site on the transcription factor that is difficult to target with small compounds. Fortunately, our understanding of the transcription factor nucleosome complex has led to several “success stories” which have changed prevalent attitudes about targeting non-ligand transcription factors in drug discovery.

Section snippets

Current strategies for transcription factor targeting

Gene transcription results not from the activation of a single protein, but requires a complex system of protein–protein interaction and, in part, chromatin remodeling that finally leads to the assembly of a transcriptome complex. In principal, besides the inhibition of transcription factor expression there are four major strategies to modulate the activity of transcription factors with small compounds or peptide-mimetics:

  • The first strategy focuses on inhibition of protein/protein interactions

Small compounds for the modulation of transcription factor activity—challenges

Compared to e.g., highly specific, humanized antibodies that are mostly restricted to surface antigens, small molecules have the tremendous advantage that they enter the cell and modulate the activity of intracellular proteins. However, the main problem connected with small size and sterical flexibility is that many small molecules can bind in different conformations also into different binding pockets and thereby modulate the activity of unrelated proteins, which leads to unwanted pleotropic

Targeting transcription factors for the treatment of polygenic disorders

Complex disorders depend on a joint contribution of various polymorphic genes so that not the mutation of one single gene is responsible for the disease phenotype but the sum effect of different gene variants. Such polygenic diseases have a much higher social impact than monogenic diseases as they include almost any complex disorder ranging from diabetes, immune disorders and coronary heart disease to cancer. Transcription factors or chromatin reader proteins like BET-proteins that contribute

Outlook and future perspective

Different strategies to inhibit the transcription of specific genes involved in cancer development, viral infection and other diseases have evolved during the last years and several “success stories” have provided evidence that the former dogma “transcription factors are un-druggable” is not valid any longer. Transcription factors need co-factors for proper functioning and the design of small drugs that interfere with transcription factor/co-factor interaction or with homo-/heterodimerization

Acknowledgements

We would like to thank the “Provita Leukaemie Stiftung”, “Austrian Wirtschaftsservice (AWS)” and “Medizinischer Forschungsfond Tirol” for the financial support of our research.

References (83)

  • L. Bella et al.

    FOXM1: a key oncofoetal transcription factor in health and disease

    Semin. Cancer Biol.

    (2014)
  • E.A. Griffiths et al.

    DNA methyltransferase and histone deacetylase inhibitors in the treatment of myelodysplastic syndromes

    Semin. Hematol.

    (2008)
  • P. Filippakopoulos et al.

    Histone recognition and large-scale structural analysis of the human bromodomain family

    Cell

    (2012)
  • B. Barneda-Zahonero et al.

    Histone deacetylases and cancer

    Mol. Oncol.

    (2012)
  • M.K. Jang et al.

    The bromodomain protein Brd4 Is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription

    Mol. Cell

    (2005)
  • G. LeRoy et al.

    The double bromodomain proteins Brd2 and Brd3 couple histone acetylation to transcription

    Mol. Cell

    (2008)
  • J.E. Delmore et al.

    BET bromodomain inhibition as a therapeutic strategy to target c-Myc

    Cell

    (2011)
  • M. Kundu et al.

    Interaction between cell cycle regulator, E2F-1, and NF-kappaB mediates repression of HIV-1 gene transcription

    J. Biol. Chem.

    (1997)
  • D. Sun et al.

    TBP binding to the TATA box induces a specific downstream unwinding site that is targeted by pluramycin

    Chem. Biol.

    (1995)
  • N.S. Raja et al.

    Chromium(III) complexes inhibit transcription factors binding to DNA and associated gene expression

    Toxicology

    (2008)
  • C. Zimmer et al.

    Interaction of the oligopeptide antibiotics netropsin and distamycin a with nucleic acids

    J. Mol. Biol.

    (1971)
  • A. Ciucci et al.

    Distamycin analogues with improved sequence-specific DNA binding activities

    Biochem. Pharmacol.

    (1994)
  • G. Feriotto et al.

    Binding of Epstein–Barr virus nuclear antigen 1 to DNA: inhibition by distamycin and two novel distamycin analogues

    Eur. J. Pharmacol.

    (1994)
  • L. Zhang et al.

    Antibiotic susceptibility of mammalian mitochondrial translation

    FEBS Lett.

    (2005)
  • G. LeRoy et al.

    The double bromodomain proteins Brd2 and Brd3 couple histone acetylation to transcription

    Mol. Cell

    (2008)
  • S.Y. Wu et al.

    Phospho switch triggers Brd4 chromatin binding and activator recruitment for gene-specific targeting

    Mol. Cell

    (2013)
  • J.M. Vaquerizas et al.

    A census of human transcription factors: function, expression and evolution

    Nat. Rev. Genet.

    (2009)
  • P.A.J. Muller et al.

    p53 mutations in cancer

    Nat. Cell Biol.

    (2013)
  • N. Issaeva et al.

    Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors

    Nat. Med.

    (2004)
  • P. Secchiero et al.

    Recent advances in the therapeutic perspectives of Nutlin-3

    Curr. Pharm. Des.

    (2011)
  • C. Zhan et al.

    An ultrahigh affinity d-peptide antagonist of MDM2

    J. Med. Chem.

    (2012)
  • H. Jiang et al.

    Stabilizers of the Max homodimer identified in virtual ligand screening inhibit Myc function

    Mol. Pharmacol.

    (2009)
  • A. Xiong et al.

    Transcription factor STAT3 as a novel molecular target for cancer prevention

    Cancers

    (2014)
  • S. Fletcher et al.

    Molecular approaches towards the inhibition of the signal transducer and activator of transcription 3 (Stat3) protein

    ChemMedChem

    (2008)
  • M. Avadisian et al.

    Artificially induced protein membrane anchorage with cholesterol-based recognition agents as a new therapeutic concept

    Angew. Chem. Int. Ed.

    (2011)
  • H.S. Ban et al.

    Identification of HSP60 as a primary target of o-carboranylphenoxyacetanilide, an HIF-1alpha inhibitor

    J. Am. Chem. Soc.

    (2010)
  • E. Miranda et al.

    A cyclic peptide inhibitor of HIF-1 heterodimerization that inhibits hypoxia signaling in cancer cells

    J. Am. Chem. Soc.

    (2013)
  • T.H. Scheuermann et al.

    Allosteric inhibition of hypoxia inducible factor-2 with small molecules

    Nat. Chem. Biol.

    (2013)
  • K. Lee et al.

    Acriflavine inhibits HIF-1 dimerization, tumor growth, and vascularization

    Proc. Natl. Acad. Sci.

    (2009)
  • W. Huang et al.

    Small-molecule inhibitors targeting the DNA-binding domain of STAT3 suppress tumor growth, metastasis and STAT3 target gene expression in vivo

    Oncogene

    (2015)
  • S.A. Khedkar et al.

    Discovery of small molecule inhibitors to Kruppel-like factor 10 (KLF10): implications for modulation of T regulatory cell differentiation

    J. Med. Chem.

    (2015)
  • Cited by (0)

    View full text