Selective Inhibition of c‐Myc/Max Dimerization by a Pyrazolo[1,5‐a]pyrimidine

The c-Myc proto-oncogene is involved in many human tumors, and needs to bind to its activation partner Max for all of its known biological activities. Inhibition of the protein–protein interactions between c-Myc and Max by cell-permeable molecules is therefore an attractive goal. Dimerization between c-Myc and Max occurs via a-helical domains comprising leucine zipper motifs, which display no obvious binding sites for inhibitory ligands. We recently identified two pyrazolo ACHTUNGTRENNUNG[1,5a]pyrimidines which inhibit c-Myc/Max dimerization from a diverse collection of chemicals (Figure 1a). These compounds, dubbed Mycro1 and Mycro2, inhibited c-Myc/Max dimerization and DNA binding with preference over other structurally related transcription factors in vitro, and exhibited c-Myc dependent effects in cellular assays. To explore the chemical space around the pyrazoloACHTUNGTRENNUNG[1,5-a]pyrimidine core structure for substitution patterns which are associated with activity against cMyc/Max dimerization, and to possibly identify a Myc/Max dimerization inhibitor with improved properties, we screened a 1438-membered pyrazoloACHTUNGTRENNUNG[1,5-a]pyrimidine library based on the structures of Mycro1 and Mycro2 (Figure 1b and Figure S1 in the Supporting Information). As c-Myc can bind DNA only as dimer with Max, we tested the compounds for their abilities to inhibit DNA binding of cMyc in a fluorescence polarization assay. Five test compounds (1–5) inhibited DNA binding of c-Myc/Max with preference over Max/Max DNA binding by more than 50% at a concentration of 100 mm (Table 1). DNA binding of Max/Max dimers is the most stringent specificity control possible, as cMyc and Max are 59% similar at the protein level in the dimerization domains, and the overall structure of the DNA-bound dimers are very similar to each other. As an additional specificity control, we analyzed the effect of compounds 1–5 on the function of the Src-homology 2 (SH2) domain of the structurally unrelated transcription factor STAT3. None of the compounds affected the interactions between STAT3 and a phosphotyrosine-containing peptide comprising the STAT3 binding motif to a major extent (Table 1). Confirmation of compound activities in well-controlled cellular systems would provide a strong argument against the notion that any in vitro data could be influenced by factors which are irrelevant under cellular conditions. In addition, cellular assays allow the analysis of compound specificities against all relevant proteins; therefore, their scope is incomparably larger than any in vitro analysis. We chose a cell proliferation assay to further analyze the effects of inhibitor candidates 1–5. Cell cycle progression and proliferation of almost all cell types, including U-2OS osteosarcoma cells, requires c-Myc function, and hence c-Myc’s ability to bind to its activation partner Max. However, for reasons not yet fully understood, PC-12 pheochromocytoma cells proliferate independent of cMyc/Max dimerization, as they express a truncated Max protein which is unable to interact with c-Myc. Therefore, a selective inhibitor of c-Myc/Max dimerization can be expected to inhibit proliferation of the c-Myc/Max-dependent U-2OS cells, without inhibiting the growth of the c-Myc/Max-independent PC-12 cells, provided it is cell permeable and stable in the cellular environment. In contrast, compounds which act unspecifically in the cellular context will either cause a reduction in the proliferative rate of both U-2OS and PC-12 cells, or display toxic effects. Whereas compound 5 appeared to be toxic in both cell lines, compound 4 inhibited proliferation of the cell lines only to a minor extent at 20 mm (Figure S2 in the Supporting Information), possibly for reasons related to cellular uptake or intracellular stability. In contrast, compounds 2 and 3 inhibited proliferation of U-2OS cells with good selectivity over proliferation of PC-12 cells at 10 mm (Figure S2 in the Supporting Information), even though compound 3 was toxic at 20 mm. The best selectivity was observed with compound 1, which strongly inhibited proliferation of U-2OS cells (70% reduction of cell number after 5 days, Figure 2a), but had no significant effect on the proliferation of PC-12 cells (Figure 2b). These data sugFigure 1. a) Structures of the c-Myc/Max dimerization inhibitors Mycro1 and Mycro2. b) Structural diversity elements of the pyrazolopyrimidine library, and classification of substitution patterns present in the library. All compounds carrying furane or thiophene as R contain CF3 or CF2Cl as substituent X.

[1]  I. D'Agnano,et al.  Oligopeptides impairing the Myc‐Max heterodimerization inhibit lung cancer cell proliferation by reducing Myc transcriptional activity , 2007, Journal of cellular physiology.

[2]  M. Henriksson,et al.  Myc Overexpression Enhances Apoptosis Induced by Small Molecules , 2006, Cell cycle.

[3]  H. Arndt Kleine Moleküle als Transkriptionsmodulatoren , 2006 .

[4]  Hans‐Dieter Arndt Small molecule modulators of transcription. , 2006, Angewandte Chemie.

[5]  Dirk Eick,et al.  Selective inhibition of c-Myc/Max dimerization and DNA binding by small molecules. , 2006, Chemistry & biology.

[6]  M. Henriksson,et al.  Identification of small molecules that induce apoptosis in a Myc-dependent manner and inhibit Myc-driven transformation. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[7]  P. Vogt,et al.  A credit-card library approach for disrupting protein-protein interactions. , 2006, Bioorganic & medicinal chemistry.

[8]  M. Eilers,et al.  Transcriptional regulation and transformation by Myc proteins , 2005, Nature Reviews Molecular Cell Biology.

[9]  T. Berg,et al.  A high-throughput fluorescence polarization assay for signal transducer and activator of transcription 3. , 2004, Analytical biochemistry.

[10]  S. McMahon,et al.  Analysis of genomic targets reveals complex functions of MYC , 2004, Nature Reviews Cancer.

[11]  John S Lazo,et al.  Low molecular weight inhibitors of Myc–Max interaction and function , 2003, Oncogene.

[12]  Stephen K. Burley,et al.  X-Ray Structures of Myc-Max and Mad-Max Recognizing DNA Molecular Bases of Regulation by Proto-Oncogenic Transcription Factors , 2003, Cell.

[13]  Stella Pelengaris,et al.  c-MYC: more than just a matter of life and death , 2002, Nature Reviews Cancer.

[14]  P. Vogt Fortuitous convergences: the beginnings of JUN , 2002, Nature Reviews Cancer.

[15]  D. Boger,et al.  Small-molecule antagonists of Myc/Max dimerization inhibit Myc-induced transformation of chicken embryo fibroblasts , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[16]  R. Eisenman,et al.  The Myc/Max/Mad network and the transcriptional control of cell behavior. , 2000, Annual review of cell and developmental biology.

[17]  Chi V. Dang,et al.  c-Myc Target Genes Involved in Cell Growth, Apoptosis, and Metabolism , 1999, Molecular and Cellular Biology.

[18]  B. Amati,et al.  Myc and the cell cycle. , 1998, Frontiers in bioscience : a journal and virtual library.

[19]  E. Ziff,et al.  The nerve growth factor-responsive PC12 cell line does not express the Myc dimerization partner Max , 1995, Molecular and cellular biology.

[20]  Stephen K. Burley,et al.  Recognition by Max of its cognate DNA through a dimeric b/HLH/Z domain , 1993, Nature.