Elsevier

Tetrahedron

Volume 63, Issue 35, 27 August 2007, Pages 8537-8562
Tetrahedron

Synthesis and biological evaluation of (−)-dictyostatin and stereoisomers

Dedicated to Professor Hisashi Yamamoto on his receipt of the Tetrahedron Prize
https://doi.org/10.1016/j.tet.2007.05.033Get rights and content

Abstract

Total syntheses of (−)-dictyostatin, 6,16-bis-epi-dictyostatin, 6,14,19-tris-epi-dictyostatin, and a number of other isomers and analogs are reported. Three main fragments—top, middle, and bottom—were first assembled and then joined by olefination or anionic addition reactions. After appending the two dienes at either end of the molecule, macrolactonization and deprotection completed the syntheses. The work proves both the relative and absolute configurations of (−)-dictyostatin. The compounds were evaluated by cell-based measurements of increased microtubule mass and antiproliferative activity, and in vitro tubulin polymerization assays as well as competitive assays with paclitaxel for its binding site on microtubules. These assays showed dictyostatin to be the most potent of the agents and further showed that the structural alterations caused from 20- to >1000-fold decreases in activity.

Introduction

Microtubule stabilization by small molecule natural products and analogs1 is a clinically proven chemotherapeutic approach for the treatment of solid tumors, and microtubule stabilizers exhibit a diverse assortment of molecular scaffolds. Dictyostatin and discodermolide (Fig. 1), the taxanes paclitaxel and docetaxel, the epothilones, the sarcodictyins and eleutherobin, and the ketosteroids 2-ethoxy-7-keto-17β-estradiol and the taccalonolides all bind with varying affinities to the paclitaxel binding site on β-tubulin within microtubules.2, 2(a), 2(b), 3, 4, 5, 6, 7, 7(a), 7(b) The tau neuronal protein binds onto microtubules in the vicinity of the paclitaxel site, and this site is well-described due to high resolution cryoelectron microscopic analyses of zinc-induced sheets of tubulin polymer stabilized by taxanes or epothilones.8, 9 Drugs binding to the site interact with amino acid residues on the M-loop (for example, Phe270, Thr274, and Arg276) and the H7 alpha helix (for example, Ala231 and His227) of β-tubulin.

Interest in the dictyostatin family of microtubule-stabilizing agents has increased significantly over the last several years. Pettit initially isolated dictyostatin 1 from a marine sponge, suggested the correct two-dimensional structure (constitution), and showed that the compound had potent activity against cancer cells.10 Subsequently, Wright and co-workers isolated 1 from a different sponge species and showed that the compound displays potent microtubule-stabilizing actions.11, 11(a), 11(b)

Dictyostatin is a structural cousin of the important microtubule-stabilizing agent discodermolide 2.12, 12(a), 12(b) Dictyostatin's 26-carbon backbone is two carbons longer than discodermolide's, and it is joined by a 22-membered macrolactone formed between the carboxyl group on C1 and the alcohol on C21. (The longer chain length means that carbon numbers of dictyostatin are two higher than the comparable carbons of discodermolide.) Dictyostatin also differs from discodermolide by possessing a Z/E diene (C2–C5) instead of a γ-lactone, and it lacks a double bond at C15,16 and a methyl group at C18. Nonetheless, 10 of dictyostatin's 11 stereocenters are also present in the discodermolide structure.

The three-dimensional structure (configuration) of dictyostatin was uncertain for almost a decade. As an outgrowth of our interest in discodermolide analogs,13, 13(a), 13(b), 13(c) we made ‘dictyostatin/discodermolide’ hybrids like 3 in 2002,14, 14(a), 14(b), 14(c) and these compounds exhibited high biological activities in both tubulin and cell assays. An underlying tenet of this work was that a hypothetical cyclization of the carbamate nitrogen of discodermolide 2 onto its lactone carbonyl (C1) would provide a 22-membered ring, the same size as the macrocycle of dictyostatin 1. At this juncture, the lack of knowledge of the complete structure of dictyostatin and the tiny quantities available from isolation were serious impediments to further medicinal chemistry research. With a solid foundation in place from the synthesis of compound 3 and related molecules, we decided to address the dictyostatin structure and supply problems by total synthesis.

In a 1995 patent, Pettit and co-workers suggested a partial configuration for dictyostatin,15 and we selected the absolute configuration depicted for compound 4 because this enantiomer is more closely related to discodermolide. In 4, seven stereocenters have the same configurations as discodermolide, two are different (C6 and C14), and two are not assigned (C16 and C19). In 2004, Paterson and co-workers suggested structure 1 for dictyostatin based on detailed NMR studies;16 in addition to assigning the two missing stereocenters, the configurations of two other centers were inverted. Structure 1 was promptly proved by a pair of total syntheses that appeared in simultaneous communications from Paterson's group17 and ours,18 and recently Phillips and Ramachandran have also reported the total syntheses of 1.19, 19(a), 19(b), 20, 20(a), 20(b), 20(c), 20(d), 20(e)

In this paper, we provide details of our total synthesis of dictyostatin 1. Along the way, we made several stereoisomers of the natural product as we drew gradually closer to the correct structure. All of these compounds have been characterized by a battery of biological assays. These results, combined with the additional results for new analogs described elsewhere21, 22, 23 and with the known SAR for discodermolide, provide for the first time a good outline of the SAR of dictyostatin. Over 5 mg of synthetic dictyostatin was provided by this work, and this was used for the detailed biological characterization of this molecule. The results of these studies have fully validated the high level of interest in dictyostatin.2, 2(a), 2(b)

Section snippets

Synthesis of 6,16-bis-epi-dictyostatin 5

Though differing in relative configuration from Pettit's structure 4 in the middle fragment, we initially decided to make compound 5 because a number of key early intermediates were already in hand. We selected the (R) configuration at C16 arbitrarily, and though this proved to be wrong, the molecule—6,16-bis-epi-dictyostatin—ultimately turned out to be structurally much closer to dictyostatin than we had initially thought.

The strategy for the synthesis of this molecule is summarized at a high

Conclusions

In summary, we have provided here the full details of synthesis of dictyostatin 1, its open-chain methyl ester analog, assorted C6, C16, and C19 epimers, and two macrolactone isomers with (E,E) geometry at C2–C5. The biological evaluation of the compounds revealed a wide range of activities resulting from these very small structural changes. The SAR determined includes the following. The macrolactone is important but not a full requisite for microtubule stabilization and antiproliferative

Ethyl (4R,5S,2E)-5,7-bis(tert-butyldimethylsilyloxy)-4-methylhept-2-enoate (83)

From 82: a solution of triethyl phosphonoacetate (3.5 mL, 17.6 mmol) was added to a cooled (0 °C), stirred suspension of NaH (0.43 g, 17.0 mmol, 95% dispersion in mineral oil) in THF (46 mL) dropwise over 10 min. The mixture was brought to room temperature with a water bath (30 min) and then cooled back to −78 °C, and the aldehyde (2.73 g, 7.58 mmol) in THF (5 mL) was added. The resulting mixture was stirred for 1 h at 0 °C, and then pH≈7 phosphate buffer solution (10 mL) and Et2O (50 mL) were added. The

Acknowledgements

We thank the National Institutes of Health for support through grant CA078390. We thank Drs. Kenneth Bair and Fred Kinder for the gift of discodermolide, Prof. Ian Paterson and Dr. Robert Britton for conducting some of the 1H NMR and TLC comparison experiments of the dictyostatin samples, and Mr. W.-H. Jung for helpful suggestions on the manuscript.

References and notes (53)

  • R.J. Kowalski et al.

    Mol. Pharmacol.

    (1997)
  • D.M. Bollag et al.

    Cancer Res.

    (1995)
  • (a)Shin, Y.; Choy, N.; Turner, T. R.; Balachandran, R.; Madiraju, C.; Day, B. W.; Curran, D. P. Org. Lett. 2002, 4,...(b)Experimental details for the syntheses reported in this paper are found in Shin, Y. Ph.D. thesis, University of...I. Paterson et al.

    Chem. Commun.

    (2007)
  • Pettit, G. R.; Cichacz, Z. A. U.S. Patent 5,430,053,...
  • Y. Shin et al.

    Angew. Chem., Int. Ed.

    (2004)
  • Dandapani, S. Unpublished results; summarized in Shin, Y. Ph.D. thesis, University of Pittsburgh: Pittsburgh, PA,...
  • J.A. Marshall et al.

    Org. Lett.

    (2003)
    H. Lebel et al.

    J. Org. Chem.

    (1998)
  • S.D. Rychnovsky et al.

    Acc. Chem. Res.

    (1998)
  • S.J. Mickel et al.

    Org. Process Res. Dev.

    (2004)
  • P. Wipf et al.

    Chem. Biol. Drug Des.

    (2006)
  • I. Paterson et al.

    Science

    (2005)
  • C. Madiraju et al.

    Biochemistry

    (2005)
    R. Buey et al.

    Chem. Biol.

    (2005)
  • J. Lowe et al.

    J. Mol. Biol.

    (2001)
  • E. Hamel et al.

    Biochemistry

    (1999)
  • P. Verdier-Pinard et al.

    Mol. Pharmacol.

    (2000)
    T.L. Tinley et al.

    Cancer Res.

    (2003)
  • S. Kar et al.

    EMBO J.

    (2003)
  • J.H. Nettles et al.

    Science

    (2004)
  • G.R. Pettit et al.

    J. Chem. Soc., Chem. Commun.

    (1994)
  • (a)Wright, A. E.; Cummins, J. L.; Pomponi, S. A.; Longley, R. E.; Isbrucker, R. A. PCT application 0162239,...R.A. Isbrucker et al.

    Biochem. Pharmacol.

    (2003)
  • D.C. MylesS.P. Gunasekera et al.
  • N. Choy et al.

    J. Med. Chem.

    (2003)
    J.M. Minguez et al.

    Bioorg. Med. Chem.

    (2003)
    J.M. Minguez et al.

    Mol. Cancer. Ther.

    (2002)
  • I. Paterson et al.

    Chem. Commun.

    (2004)
  • I. Paterson et al.

    Angew. Chem., Int. Ed.

    (2004)
  • G.W. O'Neil et al.

    J. Am. Chem. Soc.

    (2006)
    P.V. Ramachandran et al.

    Org. Lett.

    (2007)
  • For related synthetic studies,...C. Gennari et al.

    Pure Appl. Chem.

    (2007)
    E. Prusov et al.

    Org. Lett.

    (2006)
    J. Jagel et al.

    Synlett

    (2006)
    C.O. Kangani et al.

    Org. Lett.

    (2005)
    G.W. O'Neil et al.

    Tetrahedron Lett.

    (2004)
  • Cited by (54)

    • Extractive biocatalysis in the asymmetric reduction of α-alkyl, β-aryl enones by Baker's yeast

      2017, Tetrahedron Asymmetry
      Citation Excerpt :

      Given our interest in the development of biocatalytic protocols for the enantioselective preparation of compounds of synthetic interest, here we report the chemo- and enantioselective Baker’s yeast-mediated double bond reduction of α-alkyl-, β-aryl-unsaturated ketones adsorbed onto the resin Amberlite XAD-7.20,22 The α-alkylketone moiety is present in a number of bioactive compounds, including natural products28 and drugs29,30 and the methodology describe herein should be an interesting strategy for the asymmetric preparation of these compounds. More specifically, optically active α-alkyl, β-aryl ketones could find application as intermediates for the asymmetric synthesis of odorant muguesia31,32 analogues.

    • An architectonic macrolide library based on a C2-symmetric macrodiolide toward pharmaceutical compositions

      2015, Tetrahedron
      Citation Excerpt :

      Among the macrolides, 14-membered ring compounds are considered to be the most important in medicinal chemistry, because a large number of 14-membered macrolides exist that have been shown to exhibit various useful bioactivity. Some 14-membered ring natural products are shown in Fig. 1,16 such as erythromycin A, rustmicin, clonostachydiol, sekothrixide, migrastatin, and these compounds possess significant biological activity, which varies–or is influenced by–the specific structure of each compound. Consequently, a 14-membered ring macrolide library covering multiple, varied substituents promising diversity and complexity would be an attractive and important research tool.

    • Reduction of Carboxylic Acids and their Derivatives to Aldehydes

      2014, Comprehensive Organic Synthesis: Second Edition
    View all citing articles on Scopus
    View full text