A simplified synthesis of novel dictyostatin analogs with in ......2011/04/13  · A simplified...

41
1 A simplified synthesis of novel dictyostatin analogs with in vitro activity against epothilone B resistant cells and antiangiogenic activity in zebrafish embryos Laura L. Vollmer 1,$ , Maria Jiménez 2,$ , Daniel P. Camarco 1 , Wei Zhu 2 , Hikmat N. Daghestani 3 , Raghavan Balachandran 4 , Celeste E. Reese 1 , John S. Lazo 1, , Neil A. Hukriede 5 , Dennis P. Curran 2 , Billy W. Day 1,2,4 , and Andreas Vogt 1,6 * University of Pittsburgh Drug Discovery Institute 1 and Departments of Chemistry 2 , Molecular Biophysics & Structural Biology 3 , Pharmaceutical Sciences 4 , Developmental Biology 5 , and Computational and Systems Biology 6 , Pittsburgh, Pennsylvania 15260 $ These authors contributed equally to the work Running title: Potent dictyostatin analogs by convergent synthesis Key words: dictyostatin, high-content screening, multidrug resistance, zebrafish, angiogenesis. Abbreviations: Cy3, indocarbocyanine dye with three-methine linker; DDQ, 2,3-dichloro-5,6- dicyano-1,4-benzoquinone; ddGTP, 2’,3’-dideoxyguanosine-5’-triphosphate; DMSO, dimethylsulfoxide; DA, dorsal aorta; DLAV, dorsal longitudinal anastomotic vessel; Et2BOMe, diethylmethoxyborane; FITC, fluorescein isothiocyanate; HBSS, Hank’s Balanced Salt Solution; HCS, high-content screening; hpf, hours post fertilization; HWE, Horner-Wadsworth-Emmons; ISV, intersegmental vessels; MDEC, minimal detectable effective concentration; Mes, on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

Transcript of A simplified synthesis of novel dictyostatin analogs with in ......2011/04/13  · A simplified...

  • 1

    A simplified synthesis of novel dictyostatin analogs with in vitro activity against epothilone B

    resistant cells and antiangiogenic activity in zebrafish embryos

    Laura L. Vollmer1,$, Maria Jiménez2,$ , Daniel P. Camarco1, Wei Zhu2, Hikmat N. Daghestani3,

    Raghavan Balachandran4, Celeste E. Reese1, John S. Lazo1,, Neil A. Hukriede5, Dennis P. Curran2,

    Billy W. Day1,2,4, and Andreas Vogt1,6*

    University of Pittsburgh Drug Discovery Institute1 and Departments of Chemistry2, Molecular

    Biophysics & Structural Biology3, Pharmaceutical Sciences4, Developmental Biology5,

    and Computational and Systems Biology6, Pittsburgh, Pennsylvania 15260

    $ These authors contributed equally to the work

    Running title: Potent dictyostatin analogs by convergent synthesis

    Key words: dictyostatin, high-content screening, multidrug resistance, zebrafish, angiogenesis.

    Abbreviations: Cy3, indocarbocyanine dye with three-methine linker; DDQ, 2,3-dichloro-5,6-

    dicyano-1,4-benzoquinone; ddGTP, 2’,3’-dideoxyguanosine-5’-triphosphate; DMSO,

    dimethylsulfoxide; DA, dorsal aorta; DLAV, dorsal longitudinal anastomotic vessel; Et2BOMe,

    diethylmethoxyborane; FITC, fluorescein isothiocyanate; HBSS, Hank’s Balanced Salt Solution;

    HCS, high-content screening; hpf, hours post fertilization; HWE, Horner-Wadsworth-Emmons;

    ISV, intersegmental vessels; MDEC, minimal detectable effective concentration; Mes,

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 2

    morpholineethanesulfonate; MSG, monosodium glutamate; MTs, microtubules; MTS, 3-(4,5-

    dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)2H-tetrazolium; NaHMDS ,

    sodium bis(trimethylsilyl)amide; NHK, Nozaki-Hiyama-Kishi; NiCl2(dppf), dichloro[1,1'-

    bis(diphenylphosphino)ferrocene]nickel(II); PCV, posterior cardinal vein; PMB, para-

    methoxybenzyl; SAR, structure activity relationship; TBS, tert-butyldimethylsilyl; TBSOTf, tert-

    butyldimethylsilyl trifluoromethanesulfonate

    Notes

    This work was supported by the National Institutes of Health [Grants CA78039 to A.V and J.S.L.,

    HD053287 to N.A.H.], and the Fiske Drug Discovery Fund.

    *Corresponding author: Andreas Vogt, Department of Computational and Systems Biology,

    University of Pittsburgh Drug Discovery Institute, 10047 Biomedical Science Tower 3, University

    of Pittsburgh, Pittsburgh, PA 15260; Telephone: 412-383-5856; Fax: 412-648-9009; Email:

    [email protected].

    Present address for J.S. Lazo : University of Virginia School of Medicine, Charlottesville, VA 22908

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 3

    Abstract

    The natural product (–)-dictyostatin is a microtubule stabilizing agent that potently

    inhibits the growth of human cancer cells including paclitaxel-resistant clones. Extensive

    structure-activity relationship studies have revealed several regions of the molecule that could

    be altered without loss of activity. The most potent synthetic dictyostatin analog described to

    date, 6-epi-dictyostatin, has in vivo antitumor activity against human breast cancer xenografts

    superior to paclitaxel. Despite their encouraging preclinical activities, the complex chemical

    structure of the dictyostatins presents a major obstacle in their development into novel

    antineoplastic therapies. We recently reported a streamlined synthesis of 16-desmethyl-25,26

    dihydrodictyostatins and found several agents that compared with 6-epi-dictyostatin retained

    nanomolar activity in cellular microtubule bundling assays but showed cross-resistance to

    paclitaxel in cells with mutations in beta-tubulin. Extending these studies, we applied the new,

    highly convergent synthesis to generate 25,26-dihydrodictyostatin and 6-epi-25,26-

    dihydrodictyostatin. Both compounds were potent microtubule perturbing agents that induced

    mitotic arrest and microtubule assembly in vitro and in intact cells. In vitro radioligand binding

    studies showed that 25,26-dihydrodictyostatin and its C-6 epimer were able to displace

    [3H]paclitaxel and [14C]epothilone B from microtubules with potencies comparable to (–)-

    dictyostatin and discodermolide. Both compounds inhibited the growth of paclitaxel- and

    epothilone B-resistant cell lines at low nanomolar concentrations, synergized with paclitaxel in

    MDA-MB-231 human breast cancer cells, and had antiangiogenic activity in transgenic zebrafish

    larvae. The data identify 25,26-dihydrodictyostatin and 6-epi-25,26-dihydrodictyostatin as

    candidates for scale-up synthesis and further preclinical development.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 4

    Introduction

    Microtubules (MTs) are an important component in cell division and mitosis.

    Interference with MT dynamics causes a block in cell cycle progression and eventually

    programmed cell death (apoptosis), desirable results for treating rapidly dividing cancer cells.

    MT perturbing agents such as taxanes, epothilones, or vinca alkaloids, which stabilize or

    destabilize MTs, are successfully used in the treatment of solid or hematologic malignancies (1).

    The clinical successes of these anticancer agents have made MTs one of the most validated

    molecular cancer targets. Current, FDA-approved MT stabilizing agents are the taxanes

    paclitaxel (TaxolTM), docetaxel (TaxotereTM), cabazitaxel (JevtanaTM), an albumin-bound form of

    paclitaxel (AbraxaneTM), and a semi-synthetic analog of epothilone B, ixabepilone (IxempraTM).

    Despite their success, the development of drug resistance reduces the effectiveness of these

    agents (2), resulting in a continued effort to develop novel MT perturbing agents.

    Several MT stabilizing agents are currently under investigation as potential anticancer

    therapies (3). A particularly promising agent, (+)-discodermolide, a potent microtubule

    stabilizer with activity superior to paclitaxel, entered into Phase I clinical trials, but

    disappointingly failed due to pulmonary toxicity (4). Previously overshadowed by (+)-

    discodermolide, (–)-dictyostatin, a closely related compound, has recently gained attention as a

    potential anticancer agent. A decade after isolation, the complex structure was finally resolved

    (5), and two total syntheses (6, 7) provided enough sample for a detailed characterization (7, 8).

    Extensive structure activity relationship (SAR) studies have provided important information for

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 5

    the development of several (–)-dictyostatin analogs (9-11). These studies culminated in the

    discovery of 6-epi-dictyostatin, which was shown to have antitumor activity superior to

    paclitaxel in mice bearing human breast cancer MDA-MB-231 xenografts (12). In spite of these

    promising preclinical results, the complex structure and difficult synthesis of (–)-dictyostatin

    and analogs present major obstacles in their further preclinical development.

    We recently reported a streamlined synthesis that generated new 16-desmethyl-25,26-

    dihydrodictyostatins that were considerably easier to make and in preliminary biological studies

    retained much of the potency of (–)-dictyostatin (13). Based on the biological activity of the

    series, which suggested reduction of the C25, C26 double bond is well tolerated but removal of

    the C16 methyl group results in loss of activity against paclitaxel-resistant cells (13), we applied

    the new streamlined synthesis to generate 25,26-dihydrodictyostatin (1a) and 6-epi-25,26-

    dihydrodictyostatin (1b). High-content cellular analysis revealed that 25,26-dihydrodictyostatin

    and 6-epi-25,26-dihydrodictyostatin induced mitotic arrest and stabilized cellular MTs with

    potencies similar to that of the natural product. In vitro, both agents caused tubulin assembly

    with potency similar to paclitaxel and displaced [3H]paclitaxel and [14C]epothilone B from

    preformed MTs. The new analogs inhibited the growth of human cancer cells at low nanomolar

    concentrations, retained antiproliferative activity in epothilone B- and paclitaxel-resistant

    cancer cell lines, were able to synergize with paclitaxel, and had antiangiogenic activity in a

    zebrafish model. The data validate 25,26-dihydrodictyostatin and 6-epi-25,26-

    dihydrodictyostatin as bona fide MT stabilizing agents and identify them as candidates for

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 6

    continued preclinical development.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 7

    Materials and Methods

    Compounds. The dictyostatin analogs 1a and 1b were prepared by full syntheses. The

    Supporting Information contains complete characterization details and copies of NMR spectra.

    Full experimental details of the synthesis will be published elsewhere. [3H]Paclitaxel was

    obtained from the Drug Synthesis and Chemistry Branch, NCI. [14C]Epothilone B was a gift from

    Novartis Pharma.

    Cells and culture. HeLa human cervical carcinoma cells (ATCC, Manassas, VA), A549 human

    lung cancer cells, and their epothilone B-resistant counterparts EpoB40/A549 (a gift from Susan

    Horwitz, Albert Einstein College of Medicine) were maintained in Dulbecco’s modified Eagle

    medium (DMEM; Invitrogen) containing 10% fetal bovine serum (FBS, Cellgro), 2 mM L-

    glutamine (Invitrogen), and 1% penicillin-streptomycin (Invitrogen). Maintenance medium for

    EpoB40/A549 cells contained 40 nM epothilone B, which was removed prior to experimental

    setup. The HeLa/DZR cell line was generated as previously described (14) using ethyl methane

    sulfonate mutagenesis followed by stepwise increased concentrations of the antimitotic, tubulin

    assembly inhibiting, macrocyclic polyketide disorazole C1 (0.1-10.8 nM), leading to ~30-fold

    resistance to disorazole C1. These cells were valuable in our studies because they are resistant

    to natural products at least in part due to the overexpression of the ATP-binding cassette

    ABCB1 transporter (14). Thus, HeLa/DZR cells are cross resistant to the natural products

    vinblastine, doxorubicin and paclitaxel but not to cisplatin (14). Cells were cultured as

    previously described (14).

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 8

    MDA-MB-231 human breast cancer cells (ATCC), 1A9 human ovarian carcinoma cells and their

    paclitaxel-resistant clones 1A9/PTX10 and 1A9/PTX22 (a gift from Drs. Tito Fojo and Paraskevi

    Giannakakou) were maintained in RPMI 1640 medium (Invitrogen) containing 10% fetal bovine

    serum. Maintenance medium for 1A9/PTX10 and 1A9/PTX22 cells was further supplemented

    with 17 nM paclitaxel and 10 μM verapamil. Forty-eight hours prior to test agent analyses,

    paclitaxel and verapamil were removed and the cells placed into phenol red-free RPMI 1640

    medium supplemented with 10% FBS and antibiotics. All cells were maintained in a humidified

    atmosphere of 95% air-5% CO2 at 37oC. The identities of the HeLa and MDA-MB-231 cell lines

    were confirmed by The Research Animal Diagnostic Laboratory (RADIL) at the University of

    Missouri, Columbia, MO (http://www.radil.missouri.edu), using a PCR based method that

    detects 9 short tandem repeat (STR) loci, followed by comparison of results to the ATCC STR

    database.

    High-content analysis of mitotic arrest and microtubule stabilization. We used our previously

    reported cell-based immunofluorescence assay (11, 15) for high-content analysis of mitotic

    arrest and microtubule stabilization. In brief, 7,500 HeLa cells per well were seeded into the

    wells of two 384-well collagen-coated microplates (Becton Dickinson), allowed to adhere for 5

    h, and treated for an additional 21 h with either vehicle control (DMSO) or test agents. Cells

    were fixed with 4% formaldehyde containing 20 μg/mL Hoechst 33342, permeabilized with

    0.2% Triton X-100 and immunostained with the following antibody combinations: anti- -tubulin

    (Sigma Aldrich, T9026, mouse monoclonal, 1:3000 dilution)/ fluorescein isothiocyanate (FITC)-

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 9

    labeled donkey anti-mouse IgG (Jackson ImmunoResearch, 715-095-150, 1:500 dilution) and

    anti-phosphohistone H3 (Millipore, 06-570, rabbit polyclonal, 1:500 dilution)/Cy3-labeled

    donkey anti-rabbit IgG (Jackson ImmunoResearch, 711-165-152, 1:500 dilution) for mitotic

    arrest, or anti-acetylated tubulin (Sigma Aldrich, T7451, mouse monoclonal, 1:1000

    dilution)/Cy-3-labeled donkey anti-mouse IgG (Jackson ImmunoResearch, 715-165-150, 1:500

    dilution) for quantitation of stabilized cellular MTs. Cells were imaged on the ArrayScan II HCS

    reader (Thermo Fisher Cellomics, Pittsburgh, PA) using a 20X objective and an Omega XF93

    filter set at excitation/emission wavelengths of 350/461 nm (Hoechst), 494/519 nm (FITC), and

    556/573 nm (Cy3). For each condition images of 1,000 cells were acquired and analyzed using a

    Target Activation Bioapplication algorithm (Thermo Fisher Cellomics, Pittsburgh, PA) essentially

    as described (16). An image mask was generated from the Hoechst-stained nuclei. MT density

    and acetylation were defined as the average pixel intensity in an area defined by the nuclear

    mask. For determination of mitotic index and nuclear condensation, thresholds for Hoechst

    33342 and phosphohistone-H3 intensities were defined as one S.D. above the average Hoechst

    33342 or Cy3 intensity obtained from 28 vehicle-treated wells located in the center of the

    microplate. Cells were classified as positive if their average Hoechst 33342 or Cy3 intensity

    exceeded this threshold. Minimal detectable effective concentrations (MDEC) were estimated

    from concentration-response curves as described (17).

    Antiproliferative activities.

    Epothilone B-resistant cells. Growth inhibition of A549 and EpoB40/A549 cells was assessed

    over three days using a modified version of our previously described high-content cytotoxicity

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 10

    assay (18). Cells were plated in 384-well collagen-coated plates at 1,000 cells per well, allowed

    to adhere overnight, and treated in quadruplicate with 10-point 2-fold serial dilutions of

    individual test agents or vehicle control (DMSO) for an additional 72 h. After the 72 h

    treatment period, cells were fixed and nuclei stained with 10 μg/mL Hoechst 33342. Four

    imaging fields were acquired on the ArrayScan II at excitation/emission wavelengths of 350/461

    nm using a 10x objective, and nuclei enumerated as described (18). Cell densities were

    calculated as objects per imaging field and normalized to vehicle control density at the end of

    the study.

    Paclitaxel-resistant cells. Growth inhibition of 1A9 human ovarian cancer cells and the

    paclitaxel-resistant clones 1A9/PTX10 and 1A9/PTX22 was assessed over three days using a

    previously described colorimetric assay (8). Cells were seeded at a low density into 96-well

    plates. Following a 48 h attachment and growth period, the cells were treated with a

    concentration range of individual test agents in quadruplicate or vehicle control (DMSO, n=8)

    for an additional 72 h. Cell proliferation was assessed spectrophotometrically after exposure to

    3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)2H-tetrazolium and

    N-methyloxyphenazine methylsulfate (MTS) followed by an absorbance reading at 490 nm

    minus the absorbance reading at 630 nm. One full microplate was developed at the end of the

    attachment period to determine cell numbers at the time of treatment. The 50% growth

    inhibitory concentrations (GI50) of test agents were calculated from the spectrophotometrically

    determined expansion of the control cells over the 72 h period.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 11

    siRNA knockdown of ABCB1. HeLa/DZR cells were transfected with 20 nM ABCB1 siRNA or

    scrambled siRNA (Stealth siRNA Negative Control Hi GC, both from Invitrogen) as described

    previously (14). Treatment with this ABCB1 siRNA caused >75% decrease in ABCB1 protein

    levels at 24- and 72-h after transfection as measured by Western blotting (14). Briefly,

    HeLa/DZR cells were plated at a density of 7.5 x 104 cells/well into a six-well tissue culture plate

    and transfected 24 h thereafter with 20 nM ABCB1 siRNA or scrambled siRNA using 5 μL/well

    Dharmafect 1 reagent (Dharmacon, Lafayette, CO) and 480 μL/well Optimem transfection

    medium (Invitrogen) in a total volume of 2 mL/well. After 5 h, the transfection medium was

    replaced with fresh medium. Twenty-four h later cells were detached with 0.05% trypsin,

    seeded into 96 well plates at a density of 1,000 cells/well, and allowed to attach overnight. Cells

    were then treated with test agents or vehicle control for 72 h. Growth inhibition was

    determined by measuring Hoechst 33342-stained nuclei as described above.

    Combination cytotoxicity studies. Combination cytotoxicity studies were performed

    essentially as described (19). MDA-MB-231 cells were treated in quadruplicate for 96 h with

    10-point 2-fold serial dilutions of paclitaxel, test agents, or a fixed ratio of test agent and

    paclitaxel based on the GI50 values of the individual agents. Images were acquired on the

    ArrayScan II and nuclei enumerated as described above. Affected fractions (Fa) were calculated

    as Fa = cell density of drug treated cells / cell density of vehicle treated cells. The data were

    analyzed using the median-effect analysis of Chou and Talalay (20), assuming mutually exclusive

    drug effects. The degree of synergism, additivity, and antagonism was measured by calculating

    combination indices (CI) over a range of affected fractions exactly as described previously (19).

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 12

    Radioligand displacement studies. Experiments were performed as previously described (11)

    using tubulin purified in our laboratory from bovine brains by the method of Hamel (21). MTs

    were preformed by incubating 2 μM bovine tubulin with 40 μM ddGTP in 0.75 M MSG, pH 6.6,

    at 37 °C for 30 min. In separate tubes, a 50 μL solution of 8 μM test agent and 4 μM

    radiolabeled [3H]paclitaxel or [14C]epothilone B in 0.75 M MSG, pH 6.6, with a final DMSO

    content of 1%, was incubated for 10 min at 37 °C. An aliquot (50 μL) of the preformed MTs was

    added to the radioligand/test agent mixture and incubated at 37 °C for an additional 30 min.

    Final concentrations of tubulin, radioligand and test agents were 1 μM, 2 μM, and 4 μM,

    respectively. Reaction mixtures were then centrifuged at 17,000 x g for 30 min at room

    temperature and the amount of unbound radioligand determined by analyzing 50 μL of the

    supernatant by scintillation spectrometry (Beckman-Coulter LS6500). To account for non-

    specific radioligand binding, the amount of bound radioligand was calculated by subtracting the

    amount of radioligand in the supernatant in the presence of test agent from the amount of

    radioligand in the supernatant in the presence of a large molar excess of the agent with the

    highest binding affinity (20 μM (–)-dictyostatin) (8, 11). The extent of displacement was then

    calculated as percent inhibition = (1-(radioligand bound with test agent/radioligand bound with

    DMSO))*100.

    Tubulin assembly assay. Tubulin assembly was monitored turbidimetrically at 350 nm in

    temperature-controlled, multichannel Beckman-Coulter 7400 or Gilford 250

    spectrophotometers as described previously (8, 22). Reaction mixtures without test

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 13

    compounds consisted of bovine brain tubulin (1 mg/mL) in 0.1 M (4-morpholino)ethane

    sulfonate (Mes) and were cooled to 2.5oC to establish baselines. Compounds predissolved in

    DMSO were added to give the indicated final concentrations and each reaction mixture (0.25

    mL final volume) was subjected to a temperature gradient. From the precooled state, the

    temperature was rapidly raised to 30°C (in approximately 1 min) and maintained for 20 min.

    The temperature was then rapidly lowered back to 0.25–2.5°C. Absorbance at 350 nm was

    monitored every 15 s.

    Antiangiogenesis assay. The Tg(Fli1:EGFP)y1 transgenic zebrafish line (obtained from Dr. Brant

    Weinstein) was maintained as described (23). Embryos were collected at 24 h post fertilization

    (hpf) and staged according to (24). For each condition, five Tg(Fli1:EGFP)y1 transgenic zebrafish

    embryos were placed in 500 μL E3 medium (5 mM NaCl, 0.33 mM CaCl2, 0.17 mM KCl, 0.33 mM

    MgSO4) and treated with vehicle (DMSO, 0.5%) or various concentrations of test agents (1 μM

    to 25 μM) for an additional 24 h. After manual removal of the chorions, single embryos were

    transferred to wells of a 96-well half area plate (Greiner, Monroe, NC) containing 40 μg/ml

    MS222 (tricaine methanesulfonate, Sigma) in E3 for imaging.

    Photomicrographs of fluorescent ISV were acquired with the ImageXpress Ultra high-content

    reader (Molecular Devices, Sunnyvale, CA) using a 4X objective and the 488 nm argon laser.

    Images were uploaded into the Definiens Developer software suite (Definiens AG, Germany)

    and analyzed with a custom designed Cognition Network Technology (CNT) ruleset as described

    (25). Thresholding modifications were made to the CNT ruleset to accommodate the higher

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 14

    resolution and pixel depth of the ImageXpress system compared with the previously used

    ArrayScan (25). Total embryo size and intensity measurements were used to identify dead

    embryos, plate-loading artifacts, and autofluorescent compounds. Wells that contained no

    embryos, or embryos in which no dorsal region could be detected were eliminated. For the

    remaining wells, the ruleset provided numerical measurements of ISV development (area,

    length, and shape). The parameter that most robustly measured ISV development was the total

    ISV area (in pixels). Data were normalized to vehicle controls. Experiments were repeated at

    least three times.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 15

    Results

    Synthesis of novel dictyostatins analogs. We recently reported a streamlined synthesis of

    dictyostatin and used it to prepare two 16-desmethyl-25,26-dihydrodictyostatins epimeric at C6

    (13). Based on the biological activity of the series, we concluded that the reduction of the C25–

    C26 double bond is well tolerated but that removal of the C16 methyl group causes loss of

    activity against paclitaxel resistant cells (13). Accordingly, we selected 25,26-

    dihydrodictyostatin 1a and 6-epi-25,26-dihydrodictyostatin 1b as target compounds.

    The streamlined route, which features high convergence, modularity, a relative ease with which

    structurally complex new analogs of DCT can be prepared without ambiguity in the C2-C3

    configuration, and reliability of the fragment couplings, was used to make the new analogs 1a

    and 1b. Fragment couplings and completion of the syntheses are summarized in Figure 1.

    Briefly, a Horner-Wadsworth-Emmons (HWE) reaction (26) was used to couple the known top

    fragment 4 (13) with new middle fragment 3 to give 5. 1,4-Reduction of the enone, removal of

    the para-methoxybenzyl (PMB) group, stereoselective ketone reduction and mono-silylation

    then provided 6. Intermolecular esterification with epimeric acid chlorides 7a,b incorporated

    the bottom fragment (27) to give 8a,b. Selective removal of the primary tert-butyldimethylsilyl

    (TBS) group and oxidation provided aldehydes 9a,b that were substrates for an intramolecular

    Nozaki-Hiyama-Kishi (NHK) reaction (13) to give macrolactone 10a,b. Selectivity in the

    formation of the new stereocenter at C9 depended on the configuration at C6 with the b

    isomer being more selective (10b, 10/1; 10b, 3/1). Desilylation and careful purification to

    remove the C9 epimers provided the target products 1a and 1b. The strategy enabled the total

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 16

    synthesis of both analogs in a total of 39 steps, with a longest linear sequence of 11 steps from

    commercially available starting material.

    High-content analysis of mitotic arrest. We first characterized the novel agents for mitotic

    arrest and microtubule perturbation using our multiparameter high-content analysis assay (11,

    15) as described in the Materials and Methods Section. Immunofluorescence images of HeLa

    cells treated with test agents for 21 h show that the new analogs, like 6-epi-dictyostatin, caused

    MT bundling (shown in green), chromatin condensation (blue), and elevated levels of phospho-

    histone H3 (red) at nanomolar concentrations (Figure 1B). All agents showed concentration-

    dependent changes (Figure 1C). From the range of concentrations tested, a minimum

    detectable effective concentration (MDEC) value was determined (28). The data indicate that

    the new agents were equipotent to 6-epi-dictyostatin and paclitaxel. A detailed summary of

    the mitotic arrest assay results can be found in Table S1 in the Data Supplements Section.

    Stabilization of cellular MTs and tubulin assembly in vitro. We next asked if the new agents

    stabilized MTs in cells and caused MT assembly of isolated tubulin in vitro. It was previously

    shown that acetylated tubulin is a marker for stabilized cellular MTs (29). Cells were stained

    with antibodies against alpha tubulin or acetylated tubulin, respectively, to visualize cellular

    MTs and MT acetylation. Figure 2A shows distinct differences in the concentration-response

    curves of tubulin and acetylated tubulin staining obtained with (–)-dictyostatin, a known MT

    stabilizer, or vincristine, a known MT destabilizer. In cells treated with (–)-dictyostatin, we

    observed a steady increase in cellular MT density as well as acetylated MTs that plateaued at

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 17

    high concentrations. In contrast, vincristine caused an initial increase in cellular MT density and

    MT acetylation at low concentrations that was lower in magnitude and that reversed at higher

    concentrations. This bimodal response is characteristic for MT destabilizing agents: the initial

    increase results from morphological changes (i.e., cell rounding); the subsequent decrease is

    due to extraction of monomeric tubulin into the permeabilization buffer during cell processing

    and staining (15). Both the shape and the magnitude of MT and acetylated MT density curves

    caused by the dictyostatin analogs (Figure 2A) were identical to that elicited by (–)-dictyostatin,

    suggesting 25,26-dihydrodictyostatin (1a) and 6-epi-25,26-dihydrodictyostatin (1b) caused MT

    stabilization. Immunofluorescence micrographs of acetylated MTs confirmed the results of the

    automated analysis (Figure 2B).

    In vitro tubulin assembly. To further confirm the MT stabilizing activity of the new analogs, we

    performed in vitro tubulin assembly studies using a turbidity assay (22) and paclitaxel as a

    positive control. Isolated tubulin from bovine brain was incubated with vehicle (DMSO) or

    various concentrations of test agents and subjected to a temperature gradient as shown in

    Figure 2C. The new agents induced rapid and vigorous tubulin assembly with potency similar to

    paclitaxel and (–)-dictyostatin (Figure 2C). Assembly was concentration-dependent and the

    resulting polymer was cold-stable, similar to paclitaxel and consistent what we had previously

    observed with 6-epi-dictyostatin (11).

    In vitro radioligand displacement. We previously showed that (–)-dictyostatin competes with

    [3H]paclitaxel and [14C]epothilone B for binding to tubulin polymer formed in the presence of

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 18

    ddGTP (8). We therefore tested whether the new analogs retained this ability. Discodermolide,

    (–)-dictyostatin, and the new analogs were incubated with preformed MTs labeled with

    [3H]paclitaxel and [14C]epothilone, and the amount of unbound tracer measured by scintillation

    spectrometry. Table 1 shows that the new analogs displaced [3H]paclitaxel and [14C]epothilone

    B with similar potency to discodermolide or (–)-dictyostatin. These experiments provided

    conclusive evidence that the new dictyostatin analogs bind the taxoid site on tubulin polymer

    with affinities similar to that of (–)-dictyostatin.

    Antiproliferative activity in paclitaxel-, epothilone B-, and disorazole C1-resistant cell lines.

    (–)-Dictyostatin has antiproliferative activity in paclitaxel-resistant cells (11). To assess if the

    analogs remained active in drug resistant cancer cell lines, we tested 25,26-dihydrodictyostatin

    and 6-epi-25,26-dihydrodictyostatin in paclitaxel-resistant 1A9 human ovarian cancer cells with

    beta-tubulin mutations (Phe270 –> Val) and (Ala364 –>Thr) (30) induced by long-term culture

    with paclitaxel, and in epothilone B-resistant A549 human lung cancer cells that harbor a point

    mutation in beta-tubulin (292Gln–>Glu) as a result of long-term exposure to epothilone (31).

    Table 2 shows that cross-resistance to paclitaxel in the 1A9/PTX10 cells was reduced from 49-

    fold, to 15-fold with (–)-dictyostatin and further reduced with the new analogs (7- and 8-fold for

    1a and 1b, respectively). Similarly, cross resistance to epothilone B was reduced with (–)-

    dictyostatin (from 94-fold for epothilone B and 18-fold for paclitaxel to 10-fold with (–)-

    dictyostatin), and further diminished with the new analogs (5-fold and 3-fold, respectively, for

    1a and 1b). Diminished cross-resistance was also observed in a recently described disorazole

    C1-resistant human cervical carcinoma cell line that overexpresses the ABCB1 P-glycoprotein

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 19

    pump (14). Consistent with previously published data (14), these cells were 1395- and 502-fold

    resistant to paclitaxel and vinblastine, respectively (Table 2). In contrast, the new dictyostatin

    analogs showed greatly reduced cross-resistance to disorazole C1 compared with paclitaxel and

    vinblastine, with a residual 12- and 18-fold resistance respectively, for 1a and 1b. To

    investigate further if the new analogs were affected by multidrug transport proteins, we

    performed siRNA knockdown of ABCB1, which reversed the residual cross-resistance in the

    disorazole C1 resistant cells (Table 2).

    Combination cytotoxicity studies of dictyostatins and paclitaxel. Discodermolide and

    paclitaxel represent a synergistic drug combination in human cancer cells (32). We therefore

    examined the novel dictyostatin analogs in combination with paclitaxel to determine if they

    also resulted in synergy. We used our previously described growth inhibition assay (18)

    together with median effect analysis (20) to quantify synergism, additivity, and antagonism.

    MDA-MB-231 cells were treated with comprehensive concentration gradients of paclitaxel,

    discodermolide, 6-epi-dictyostatin, 25,26-dihydrodictyostatin 1a, 6-epi-25,26-

    dihydrodictyostatin 1b, or equipotent, fixed mixtures thereof with paclitaxel for four days, and

    cell densities quantified by counting Hoechst 33342-stained nuclei. Median effect (Dm), slopes

    (m), and correlation coefficients (r) for the individual agents and the combinations can be found

    in Table S2 in the Supporting Information Section. Combination indices were then calculated for

    various effect levels by the method of Chou and Talalay (20, 33) as described previously (18).

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 20

    As shown in Figure 3, we reproduced the results of Martello et al. (32), who found the

    combination of paclitaxel and discodermolide to be synergistic at lower effect levels and

    antagonistic at high effect levels. The dictyostatins had combination index profiles similar to

    that of discodermolide, although the degree of synergism was lower. The least potent

    combination was with 6-epi-25,26-dihydrodictyostatin 1b (Figure 3D), which was additive over

    much of the effect range. The data consistently repeated over the course of multiple

    independent experiments. The data suggest that (–)-dictyostatin and the new analogs share

    the ability of discodermolide to synergize with paclitaxel, a feature that is potentially favorable

    for clinical use.

    Inhibition of angiogenesis in zebrafish embryos. Some MT perturbing agents have

    antiangiogenic activity that contributes to in vivo anticancer activity (34). Solid tumors require

    an adequate supply of blood vessels to survive, grow, and metastasize (reviewed in (35)), and

    agents targeting tumor angiogenesis are now FDA-approved anti-cancer medicines (e.g.,

    bevacizumab, Avastin ®). We therefore asked if the dictyostatin analogs had antiangiogenic

    activity. We used the Tg(fli1:EGFP)y1 zebrafish line that expresses EGFP under the control of

    the Fli1 promoter, thereby labeling all blood vessels and providing a live visual marker for

    vascular development (36). Zebrafish have a stereotypical vertebrate vasculature that develops

    in response to the same signals that guide mammalian blood vessel development (37, 38).

    Zebrafish vasculature recruitment also occurs in response to human glioma xenografts (39, 40),

    mimicking conditions found in mammals.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 21

    Tg(fli1:EGFP)y1 zebrafish embryos at 24 hpf were treated for 24 h with vehicle or various

    concentrations of test agents and imaged. Figure 4A shows that, as expected, vehicle-treated

    embryos had well-established intersegmental vessels (ISV) that extended from the dorsal aorta

    (DA) and connected to the dorsal longitudinal anastomotic vessel (DLAV) (Figure 4A, (Isogai et

    al., 2001)). Visually, all of the dictyostatin analogs stunted ISV outgrowth and prevented the

    establishment of the DLAV (Figure 4A, upper panels). Our previously described image analysis

    algorithm (25) quantified the antiangiogenic phenotype (Figure 4A, lower panels). All agents

    concentration-dependently inhibited angiogenesis (Figure 4B), with concentrations required to

    reduce ISV area by 50% compared with control (IC50) of 8.8, 6.1, and 6.7 μM for 6-epi-

    dictyostatin, 25,26-dihydrodictyostatin 1a, and 6-epi-25,26-dihydrodictyostatin 1b, respectively.

    Importantly, at concentrations that were antiangiogenic, we observed no obvious signs of

    toxicity such as the appearance of necrotic opaque cells. At the highest concentration tested

    (25 μM, data not shown), the test agents caused a bent-tail phenotype, suggesting that the

    compounds at this concentration would likely cause developmental defects in the embryo.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 22

    Discussion

    An improved synthetic route to dictyostatin analogs. The complex chemical structure and

    difficult synthesis of the dictyostatins is a major impediment to their development into novel

    antineoplastic agents. This work validates that our recently described synthetic route (13) can

    be used to rapidly make new analogs. The streamlined route features a bimolecular

    esterification to make the C1–O21 bond in place of the usual macrolactonization. This bypasses

    a major problem of Z/E isomerization of the C2–C3 alkene that has plagued the

    macrolactonization. In turn, the large ring is closed by a mild Nozaki-Hiyama-Kishi reaction to

    make the C9–C10 bond. It should be possible to access many more analogs thanks to the

    modularity of this route and the reliability of the fragment couplings and end game steps.

    Predictions based on existing SAR are validated. Consistent with prior findings, removal of the

    C16 methyl moiety did not dramatically affect antiproliferative activity in human tumor cells

    expressing wild-type tubulin but diminished the ability of the compounds to inhibit the growth

    of paclitaxel-resistant clones harboring mutations within beta-tubulin (10). We therefore

    reasoned that retaining the C16 methyl group would preserve the lack of cross-resistance to

    paclitaxel and selected 25,26-dihydrodictyostatin and 6-epi-25,26-dihydrodictyostatin as target

    compounds. Consistent with existing SAR, both new agents showed low nanomolar

    antiproliferative activity in HeLa, A-549, and MDA-MB-231 cells, and reduced cross-resistance

    to paclitaxel and epothilone B in cells with mutant tubulin.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 23

    Dictyostatin analogs occupy the taxane binding site on tubulin. To confirm that the new

    analogs directly interact with their proposed target, we performed radioligand binding studies.

    These experiments show the new analogs have affinities for the taxane site similar to paclitaxel,

    epothilone B, or discodermolide. The precise location of the dictyostatin binding site has not

    been established, because the interaction of the dictyostatins or discodermolide with tubulin

    has not been solved by cryoelectron microscopy as it has for paclitaxel and epothilone A (41,

    42). Furthermore, two binding sites have been described for taxanes: an internal luminal

    binding site and an external transient binding site of unknown structure. The radioligand

    competition studies are unable to distinguish the two sites. However, growth inhibition studies

    of the natural product (8) and on the 16-desmethyl analogs using 1A9/PTX10 ovarian cancer

    cells with the Phe270 —>Val mutation that we performed previously (13) are consistent with

    dictyostatin and analogs binding to the internal site.

    Similarities and dissimilarities to (+)-discodermolide. The new analogs retained some but not

    all of the ability of discodermolide to synergize with paclitaxel in human breast cancer cells.

    Modeling studies based on NMR structures have suggested that the bound conformer of

    dictyostatin resembles that of discodermolide and provides similar contacts with tubulin (43).

    Because it is unusual for two drugs that bind to identical sites on the same target to show

    synergy, the combination cytotoxicity data do support the previously proposed model of

    overlapping binding sites for paclitaxel and the dictyostatins (43). The extent of synergy varied

    with the analogs; the least potent agent was 1b, although all of them showed a trend towards

    higher synergy at lower effect levels. Therefore, our results confirmed a synergistic

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 24

    relationship specifically at the lower concentrations of the two drugs as reported by Horwitz’s

    group (32). The reasons for the differential activity of the analogs in this assay are unknown.

    The fact that the dictyostatins were essentially equivalent in all of our assays, including the in

    vitro radioligand binding studies, makes it seem unlikely that differences in binding affinity or

    cellular distribution would account for the observed differences. To formulate a valid

    hypothesis based on structural terms, however, physical evidence such as a high resolution

    cryoelectron microscopy structure of the dictyostatins and discodermolide is needed.

    Alternatively, the different degree of synergy of the dictyostatins compared with

    discodermolide may be a result of off-target effects. As pointed out by Martello et al. (32),

    discodermolide induces apoptosis by mechanisms unrelated to MT binding, and it is currently

    not known whether the dictyostatins share these activities. The data do suggest, however, that

    the combination of paclitaxel with either 6-epi-dictyostatin or 1a merits exploration in in vivo

    antitumor studies.

    Dictyostatins lack cross-resistance to paclitaxel, epothilone B, and disorazole C1. Drug

    resistance is a major problem with MT perturbing agents in clinical use. One clinically

    important resistance mechanism is overexpression of p-glycoprotein efflux pumps (44). In

    cultured cells, additional resistance mechanisms have been observed that involve tubulin

    mutations induced by long-term culture of cell lines in the presence of MT perturbing agents

    (31, 45), although such drug-induced mutations have not been found in clinical samples. In

    three such cellular models with mutant tubulin, the new analogs retained activity against both

    paclitaxel- and epothilone B-resistant cells, and appeared less cross-resistant than the natural

    product. The 1A9/PTX10 cell line harbors a Phe270 —> Val mutation that is located within the

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 25

    taxane binding site (42) and confers 49-fold resistance to paclitaxel. Consistent with our

    previous studies with (–)-dictyostatin and 6-epi-dictyostatin (13), cross-resistance was reduced

    to Thr mutation that is adjacent to the taxane binding

    pocket. In epothilone B resistant A-549 cells with a 292Gln —> Glu mutation, which is located

    at the periphery of the taxane pocket and makes contact with epothilone but not paclitaxel

    (42), the analogs showed only a 12-18-fold cross resistance compared with epothilone B (94-

    fold resistance). The data indicate that reduction of the terminal double bond does not alter

    the mode of tubulin binding. They are consistent with a mode of binding to tubulin as

    proposed by Canales et al. (43) that involves the taxane binding pocket but not residues outside

    the pocket that make contact with the taxane side chain.

    The analogs showed a unique behavior toward cells with acquired resistance against the natural

    product disorazole C1 (14), which owe their resistance phenotype at least in part to

    overexpression of the ABCB1 p-glycoprotein pump. All agents were subnanomolar inhibitors of

    wild-type HeLa cells. Paclitaxel and vinblastine were 1395- and 502-fold less active,

    respectively, in the resistant cells (HeLa/DZR, Table 2). Knockdown of the P-glycoprotein pump,

    ABCB1, restored most, of their activity (HeLa/DZR/ABCB1 siRNA, Table 2). In contrast, the

    HeLa/DZR cells showed only minor cross-resistance to the dictyostatin analogs (12- to 18-fold,

    HeLa/DZR, Table 2) that was fully reversed by ABCB1 knockdown. The data suggest that the

    dictyostatins may be only weak substrates for ABCB1. Moreover, because the HeLa/DZR cells

    were generated by a single exposure to the mutagen ethyl methane sulfonate followed by a

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 26

    stepwise increased disorazole C1 exposure, it is likely that resistance mechanisms other than

    elevated ABCB1 exist, but these do not appear to influence cellular sensitivity to the

    dictyostatin analogs.

    Antiangiogenic activity in vivo. We had previously shown that microtubule-perturbing agents

    inhibit angiogenesis in Tg(fli1:EGFP)y1 transgenic fluorescent zebrafish embryos (15). Here we

    demonstrate that the new analogs also have this property, which is thought to be beneficial for

    clinical activity (34, 46). In the Tg(fli1:EGFP)y1 model, the agents appeared to have

    antiangiogenic rather than antivascular activity. During development, intersegmental vessels

    (ISV)s sprout from the dorsal aorta (DA) at 24 hpf, and at 48 hpf are fully established and

    connected to the dorsal longitudinal anastomotic vessel (DLAV). To assess the effect of test

    agents on new vessel outgrowth (angiogenesis), embryos were treated at 24 hpf (when ISV are

    just beginning to sprout and are barely visible (15)), and analyzed for ISV formation 24 h

    thereafter. While the analogs caused a concentration-dependent inhibition of new vessel

    growth, they did not affect existing blood vessels as the head and large trunk vessels were

    intact. Furthermore, heart beat, circulation, and twitch response (assessed visually) were all

    normal (data not shown). We also did not observe tissue necrosis, which would show as

    opaque cells in the fluorescence micrographs (see Figure 4). Test agent-treated embryos also

    showed little difference in gross morphology when compared with control embryos (Figure 4),

    although we did observe a bent tail phenotype at the highest concentration tested (25 μM).

    While the model is currently not well enough characterized to suggest therapeutic safety in the

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 27

    context of angiogenesis inhibition, the data indicate the new dictyostatins have antiangiogenic

    activity in a zebrafish model of angiogenesis at nontoxic concentrations.

    In summary, we have used our previously reported, highly convergent, streamlined

    synthesis (13) to generate 25,26-dihydrodictyostatin and 6-epi-25,26-dihydrodictyostatin, two

    new analogs of the highly complex natural product, (–)-dictyostatin. Consistent with existing

    SAR studies and a mode of action involving high affinity binding to the taxane site on tubulin,

    the new analogs retained essentially all of the biological activities of (–)-dictyostatin and 6-epi-

    dictyostatin, the only analog whose activity in adult mammals has been described to date (12).

    While the new analogs do not represent a significant simplification from a structural

    standpoint, reduction of the exposed double bond eliminates chemical reactivity and a

    potential metabolic “soft spot”, as has been shown for discodermolide (47). Future

    experiments should focus on this issue. The results identify 25,26-dihydrodictyostatin and 6-

    epi-25,26-dihydrodictyostatin as candidates for scale-up using the improved synthesis

    procedure and for further preclinical development.

    Acknowledgments

    We thank Dr. Brant Weinstein for the transgenic Tg(fli1:EGFP)y1 line, Dr. Susan B. Horwitz for

    the epothilone B-resistant cells, Drs. Tito Fojo and Paraskevi Giannakakou for the paclitaxel-

    resistant clones, the National Cancer Institute for [3H]paclitaxel, and Novartis Pharma for

    [14C]epothilone B.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 28

    References

    1. Perez EA. Microtubule inhibitors: Differentiating tubulin-inhibiting agents based on mechanisms of action, clinical activity, and resistance. Mol Cancer Ther 2009;8:2086-95.

    2. Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer 2004;4:253-65.

    3. Klar U, Hoffmann J, Giurescu M. Sagopilone (ZK-EPO): from a natural product to a fully synthetic clinical development candidate. Expert Opin Investig Drugs 2008;17:1735-48.

    4. Kingston DG. Tubulin-interactive natural products as anticancer agents. J Nat Prod 2009;72:507-15.

    5. Paterson I, Britton R, Delgado O, Wright AE. Stereochemical determination of dictyostatin, a novel microtubule-stabilising macrolide from the marine sponge Corallistidae. Chem Commun 2004:632-3.

    6. Paterson I, Britton R, Delgado O, Meyer A, Poullennec KG. Total synthesis and configurational assignment of (-)-dictyostatin, a microtubule-stabilizing macrolide of marine sponge origin. Angew Chem Int Edit 2004;43:4629-33.

    7. Shin Y, Fournier JH, Fukui Y, Bruckner AM, Curran DP. Total synthesis of (-)-dictyostatin: confirmation of relative and absolute configurations. Angew Chem Int Ed Engl 2004;43:4634-7.

    8. Madiraju C, Edler MC, Hamel E, Raccor BS, Balachandran R, Zhu G, et al. Tubulin assembly, taxoid site binding, and cellular effects of the microtubule-stabilizing agent dictyostatin. Biochemistry 2005;44:15053-63.

    9. Jung WH, Harrison C, Shin Y, Fournier JH, Balachandran R, Raccor BS, et al. Total synthesis and biological evaluation of C16 analogs of (-)-dictyostatin. J Med Chem 2007;50:2951-66.

    10. Shin Y, Fournier JH, Balachandran R, Madiraju C, Raccor BS, Zhu G, et al. Synthesis and biological evaluation of (-)-16-normethyldictyostatin: a potent analogue of (-)-dictyostatin. Org Lett 2005;7:2873-6.

    11. Raccor BS, Vogt A, Sikorski RP, Madiraju C, Balachandran R, Montgomery K, et al. Cell-based and biochemical structure-activity analyses of analogs of the microtubule stabilizer dictyostatin. Mol Pharmacol 2008;73:718-26.

    12. Eiseman JL, Bai L, Jung WH, Moura-Letts G, Day BW, Curran DP. Improved synthesis of 6-epi-dictyostatin and antitumor efficacy in mice bearing MDA-MB231 human breast cancer xenografts. J Med Chem 2008;51:6650-3.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 29

    13. Zhu W, Jimenez M, Jung WH, Camarco DP, Balachandran R, Vogt A, et al. Streamlined syntheses of (-)-dictyostatin, 16-desmethyl-25,26-dihydrodictyostatin, and 6-epi-16-desmethyl-25,26-dihydrodictyostatin. J Am Chem Soc 2010;132:9175-87.

    14. Lazo JS, Reese CE, Vogt A, Vollmer LL, Kitchens CA, Gunther E, et al. Identifying a resistance determinant for the antimitotic natural products disorazole C1 and A1. J Pharmacol Exp Ther 2010;332:906-11.

    15. Vogt A, McPherson PA, Shen X, Balachandran R, Zhu G, Raccor BS, et al. High-content analysis of cancer-cell-specific apoptosis and inhibition of in vivo angiogenesis by synthetic (-)-pironetin and analogs. Chem Biol Drug Des 2009;74:358-68.

    16. Wang Z, McPherson PA, Raccor BS, Balachandran R, Zhu G, Day BW, et al. Structure-activity and high-content imaging analyses of novel tubulysins. Chem Biol Drug Des 2007;70:75-86.

    17. Wipf P, Reeves JT, Balachandran R, Giuliano KA, Hamel E, Day BW. Synthesis and biological evaluation of a focused mixture library of analogues of the antimitotic marine natural product curacin A. J Am Chem Soc 2000;122:9391-5.

    18. Vogt A, Kalb EN, Lazo JS. A scalable high-content cytotoxicity assay insensitive to changes in mitochondrial metabolic activity. Oncol Res 2004;14:305-14.

    19. Vogt A, McDonald PR, Tamewitz A, Sikorski RP, Wipf P, Skoko JJ, 3rd, et al. A cell-active inhibitor of mitogen-activated protein kinase phosphatases restores paclitaxel-induced apoptosis in dexamethasone-protected cancer cells. Mol Cancer Ther 2008;7:330-40.

    20. Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 1984;22:27-55.

    21. Hamel E, Lin CM. Separation of active tubulin and microtubule-associated proteins by ultracentrifugation and isolation of a component causing the formation of microtubule bundles. Biochemistry 1984;23:4173-84.

    22. ter Haar E, Kowalski RJ, Hamel E, Lin CM, Longley RE, Gunasekera SP, et al. Discodermolide, a cytotoxic marine agent that stabilizes microtubules more potently than taxol. Biochemistry 1996;35:243-50.

    23. Westerfield M. The zebrafish book. A guide for the laboratory use of zebrafish (Danio rerio): Univ. of Oregon Press, Eugene; 2000.

    24. Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF. Stages of Embryonic-Development of the Zebrafish. Dev Dyn 1995;203:253-310.

    25. Vogt A, Cholewinski A, Shen X, Nelson SG, Lazo JS, Tsang M, et al. Automated image-based phenotypic analysis in zebrafish embryos. Dev Dyn 2009;238:656-63.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 30

    26. Paterson I, Yeung KS, Smaill JB. The Horner-Wadsworth-Emmons reaction in natural-products synthesis - Expedient construction of complex (E)-enones using barium hydroxide. Synlett 1993:774-6.

    27. Moura-Letts G, Curran DP. Selective synthesis of (2Z,4E)-dienyl esters by ene-diene cross metathesis. Org Lett 2007;9:5-8.

    28. Minguez JM, Kim SY, Giuliano KA, Balachandran R, Madiraju C, Day BW, et al. Synthesis and biological assessment of simplified analogues of the potent microtubule stabilizer (+)-discodermolide. Bioorg Med Chem 2003;11:3335-57.

    29. Mohan R, Panda D. Kinetic stabilization of microtubule dynamics by estramustine is associated with tubulin acetylation, spindle abnormalities, and mitotic arrest. Cancer Res 2008;68:6181-9.

    30. Giannakakou P, Sackett DL, Kang YK, Zhan Z, Buters JT, Fojo T, et al. Paclitaxel-resistant human ovarian cancer cells have mutant beta-tubulins that exhibit impaired paclitaxel-driven polymerization. J Biol Chem 1997;272:17118-25.

    31. Yang CP, Verdier-Pinard P, Wang F, Lippaine-Horvath E, He L, Li D, et al. A highly epothilone B-resistant A549 cell line with mutations in tubulin that confer drug dependence. Mol Cancer Ther 2005;4:987-95.

    32. Martello LA, McDaid HM, Regl DL, Yang CP, Meng D, Pettus TR, et al. Taxol and discodermolide represent a synergistic drug combination in human carcinoma cell lines. Clin Cancer Res 2000;6:1978-87.

    33. Chou J, Chou TC. Dose-effect analysis with microcomputers: quantitation of ED 50 , LD 50 , synergism/antagonism, low-dose risk, receptor ligand binding and enzyme kinetics. Elsevier/North-Holland, Amsterdam; 1987.

    34. Belotti D, Vergani V, Drudis T, Borsotti P, Pitelli MR, Viale G, et al. The microtubule-affecting drug paclitaxel has antiangiogenic activity. Clin Cancer Res 1996;2:1843-9.

    35. Folkman J. Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Disc 2007;6:273-86.

    36. Lawson ND, Weinstein BM. In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev Biol 2002;248:307-18.

    37. Childs S, Chen JN, Garrity DM, Fishman MC. Patterning of angiogenesis in the zebrafish embryo. Development 2002;129:973-82.

    38. Lawson ND, Weinstein BM. Arteries and veins: making a difference with zebrafish. Nat Rev Genet 2002;3:674-82.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 31

    39. Nicoli S, Ribatti D, Cotelli F, Presta M. Mammalian tumor xenografts induce neovascularization in zebrafish embryos. Cancer Res 2007;67:2927-31.

    40. Geiger GA, Fu W, Kao GD. Temozolomide-mediated radiosensitization of human glioma cells in a zebrafish embryonic system. Cancer Res 2008;68:3396-404.

    41. Nogales E, Wolf SG, Downing KH. Structure of the alpha beta tubulin dimer by electron crystallography. Nature 1998;391:199-203.

    42. Nettles JH, Li H, Cornett B, Krahn JM, Snyder JP, Downing KH. The binding mode of epothilone A on alpha,beta-tubulin by electron crystallography. Science 2004;305:866-9.

    43. Canales A, Matesanz R, Gardner NM, Andreu JM, Paterson I, Diaz JF, et al. The bound conformation of microtubule-stabilizing agents: NMR insights into the bioactive 3D structure of discodermolide and dictyostatin. Chem-Eur J 2008;14:7557-69.

    44. Kavallaris M. Microtubules and resistance to tubulin-binding agents. Nat Rev Cancer 2010;10:194-204.

    45. Giannakakou P, Gussio R, Nogales E, Downing KH, Zaharevitz D, Bollbuck B, et al. A common pharmacophore for epothilone and taxanes: molecular basis for drug resistance conferred by tubulin mutations in human cancer cells. Proc Natl Acad Sci U S A 2000;97:2904-9.

    46. Vacca A, Iurlaro M, Ribatti D, Minischetti M, Nico B, Ria R, et al. Antiangiogenesis is produced by nontoxic doses of vinblastine. Blood 1999;94:4143-55.

    47. Fan Y, Schreiber EM, Day BW. Human liver microsomal metabolism of (+)-discodermolide. J Nat Prod 2009;72:1748-54.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 32

    Table 1. In vitro radioligand displacement.

    Percent inhibition of[3H]Paclitaxel binding

    Percent inhibition of [14C]Epothilone B binding

    4 μM test agent 20 μM test agent 4 μM test agent 20 μM test agent25,26-dihydrodictyostatin 1a 61 ± 4 (2) 68 ± 5 (2) 66 ± 13 (3) 101 ± 9 (3) 6-epi-25,26-dihydrodictyostatin 1b 83 ± 2 (2) 98 ± 11 (2) 41 ± 3 (3) 70 ± 9 (3) (–)-dictyostatin 75 ± 6 (2) 100 (2) 53 ± 6 (3) 100 (3) (+)-discodermolide 86 ± 13 (2) ND 65 ± 3 (3) 112 ± 17 (3)

    Preformed MTs were labeled with 2 μM [3H]paclitaxel or 2 μM [14C]epothilone in the presence or absence of test agents, and the

    amount of unbound radioligand measured by scintillation spectrometry as described in the Materials and Methods Section. Data

    were normalized to free radiolabel measured in the presence of vehicle (DMSO) and in the presence of 20 μM (–)-dictyostatin. Data

    represent the average ± S.D. of (n) independent experiments. ND, not determined.

    on June 7, 2021. © 2011 A

    merican A

    ssociation for Cancer R

    esearch. m

    ct.aacrjournals.org D

    ownloaded from

    Author m

    anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

    Author M

    anuscript Published O

    nlineFirst on A

    pril 13, 2011; DO

    I: 10.1158/1535-7163.MC

    T-10-1048

    http://mct.aacrjournals.org/

  • 33

    Table 2. Antiproliferative activity in paclitaxel-, epothilone B-, and disorazole C1-resistant cell lines.

    A549 aEpoB40/ A549 a

    1A9 b 1A9/PTX10 b 1A9/PTX22b HeLa c HeLa/DZR cHeLa/DZR

    ABCB1siRNA c

    25,26-dihydrodictyostatin1a

    2.3 ± 0.5 12.4 ± 1.5 (5)

    6.8 ± 0.7 44.9 ± 12.9 (7)

    12.2 ± 4.8 (2)

    0.6 ± 0.3 7.0 ± 3.2 (12)

    0.4 ± 0.1 (1)

    6-epi-25,26-dihydrodictyostatin 1b

    4.5 ± 1.0 15.1 ± 1.8 (3)

    5.0 ± 3.5 42.1 ± 20.1 (8)

    9.6 ± 4.8 (2) 1.0 ± 0.7 17.5 ± 9.1 (18)

    0.9 ± 0.5 (1)

    6-epi-dictyostatin0.7 ± 0.3 4.5 ± 1.0

    (6)0.8 ± 0.1 14.2 ± 5.6

    (18)1.9 ± 0.7 (2) 0.4 ± 0.0 5.9 ± 2.6

    (15)0.7 ± 0.2

    (2)

    (–)-dictyostatin 0.5 ± 0.0 5.1 ± 0.5

    (10)1.3 ± 1.0 18.8 ± 2.2

    (15)5.1 ± 0.6 (4) ND d ND ND

    (+)-discodermolide 2.2 ± 0.1 22.0 ± 2.2

    (10)ND ND ND ND ND ND

    Paclitaxel0.4 ± 0.1 7.0 ± 1.0

    (18)1.7 ± 0.3 83.5 ± 6.4

    (49)58.7 ± 13.7

    (35)0.2 ± 0.0 279 ± 13

    (1395) 4.5 ± 2.4

    (23)

    Epothilone B 0.2 ± 0.1 18.7 ± 3.8

    (94)ND ND ND ND ND ND

    VinblastineND ND ND ND ND 0.4 ± 0.0 201 ± 1

    (502)8.3 ± 0.5

    (21)

    Cells were exposed to vehicle or test agents for 72-120 h and cell growth determined as described in the Materials and Methods

    Section. Data represent average 50% growth inhibitory concentrations (GI50) ± S.D. (nM) from at least three independent

    experiments. Values in parentheses denote fold resistance compared with wild-type cells. a 3-day HCS assay; b 3-day MTS assay; c 5-

    day HCS assay; ND, not determined

    on June 7, 2021. © 2011 A

    merican A

    ssociation for Cancer R

    esearch. m

    ct.aacrjournals.org D

    ownloaded from

    Author m

    anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

    Author M

    anuscript Published O

    nlineFirst on A

    pril 13, 2011; DO

    I: 10.1158/1535-7163.MC

    T-10-1048

    http://mct.aacrjournals.org/

  • 34

    Figure Legends

    Figure 1. Mitotic arrest and alterations in MT morphology by 25,26-dihydrodictyostatin (1a)

    and 6-epi-25,26-dihydrodictyostatin (1b) prepared by a highly convergent synthesis. A) A

    highly convergent chemical synthesis of 25,26-dihydrodictyostatin (1a) and its C6-epimer (1b).

    Reaction conditions: (a) Ba(OH)2, 70%; (b) [Ph3PCuH]6; (c) 2,3-dichloro-5,6-dicyano-1,4-

    benzoquinone (DDQ), 76% over 2 steps; (d) NaBH4, Diethylmethoxyborane (Et2BOMe), 90%; (e)

    tert-butyldimethylsilyl trifluoromethanesulfonate (TBSOTf), 86%. (f) sodium

    bis(trimethylsilyl)amide (NaHMDS), b isomer 57%; (g) HF•pyridine, a isomer 70% (over 2 steps),

    b isomer 84%; (h) Dess-Martin, NaHCO3, a isomer 95%%, b isomer 94%; (i) CrCl2, dichloro[1,1'-

    bis(diphenylphosphino)ferrocene]nickel(II) (NiCl2(dppf)), 4,4'-di-tert-butyl-2,2'-dipyridyl, a

    isomer 22%, b isomer 42%; (f) HF•pyridine, a isomer 54%, b isomer 82%. (B-C),

    Multiparametric analysis of mitotic arrest. HeLa cells were treated for 21 h with concentration

    gradients of 6-epi-dictyostatin, 25,26-dihydrodictyostatin 1a, or 6-epi-25,26-dihydrodictyostatin

    1b, and stained with anti-tubulin and anti-phosphohistone H3 antibodies. Nuclei were

    counterstained with Hoechst 33342. B) Images documenting microtubule perturbation and

    mitotic arrest. Fluorescence micrographs of tubulin (green), nuclei (blue), and phospho-histone

    H3 (red) were acquired on the ArrayScan II. Images are from one representative field of view.

    All agents are shown at 15.6 nM. C) Quantification of cellular response. MT density, nuclear

    condensation, and histone H3 phosphorylation were measured in 1,000 individual cells after

    treatment with 6-epi-dictyostatin ( ), 25,26-dihydrodictyostatin 1a ( ), or 6-epi-25,26-

    dihydrodictyostatin 1b ( ). Data points represent the average of quadruplicate wells ± S.E.

    from one representative experiment that was repeated at least three times.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 35

    Figure 2. Stabilization of cellular MTs and tubulin assembly in vitro. A) HeLa cells were

    treated for 21 h with vincristine, (–)-dictyostatin, 25,26-dihydrodictyostatin, or 6-epi-25,26-

    dihydrodictyostatin. MT density ( ) or acetylated MT density ( ) was quantified on the

    ArrayScan II as described in Materials and Methods. Data are the averages ± S.E. of

    quadruplicate wells from a single experiment that has been repeated three times. B)

    Immunofluorescence images of acetylated MTs in cells treated with dictyostatin analogs (31

    nM) or vincristine (25 nM). C) In vitro tubulin assembly. Electrophoretically homogenous

    bovine brain tubulin was incubated with vehicle (DMSO) or the indicated concentrations of test

    agents and subjected to a temperature gradient as indicated. Tubulin assembly was measured

    by turbidimetry at 350 nm.

    Figure 3. Combination cytotoxicity of dictyostatin analogs and paclitaxel. MDA-MB-231 cells

    were treated with concentration gradients of paclitaxel, discodermolide, 6-epi-dictyostatin,

    25,26-dihydrodictyostatin, 6-epi-25,26-dihydrodictyostatin, or equipotent combinations of

    paclitaxel and dictyostatin analog for four days. Cell densities were quantified on the ArrayScan

    II as described in Materials and Methods. Data were analyzed by the median-effect analysis of

    Chou and Talalay, assuming mutually exclusive drug effects. Combination Indices (CI) were

    calculated for each effect level. CI values of < 1, 1 (solid line), and >1 are synergistic, additive,

    and antagonistic, respectively. Data points represent average CI ± S.E. of at least three

    independent experiments.

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • 36

    Figure 4. Antiangiogenic activity in zebrafish embryos. Tg(fli1:EGFP)y1 larvae at 24 hpf were

    treated with vehicle (DMSO) or various concentrations of test compounds for an additional 24 h

    and imaged on a Molecular Devices ImageXpress high-content confocal imager. (A-D)

    representative fluorescence micrographs of embryos treated with A) vehicle (DMSO) B) 6-epi-

    dictyostatin (12.5 μM), C) 25,26-dihydrodictyostatin 1a (5 μM) or D) 6-epi-25,26-

    dihydrodictyostatin 1b (5 μM). Top panel, raw fluorescence micrographs (shown inverted for

    visual clarity); Bottom panel, images with Cognition Network Technology (CNT) analysis applied.

    DMSO concentration was 0.5% in all cases. ISVs are shown in red. Green, purple, pink, yellow,

    and white areas represent the zebrafish body, large trunk vessels, head, yolk, and dorsal

    regions, respectively. E) Quantification of ISVs. Images were analyzed with a CNT ruleset as

    described (25). Total ISV area was calculated for each condition, and normalized to pooled

    vehicle controls. Each data point represents the average ± S.E. from at least three independent

    experiments, performed in quintuplicate

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • B

    Microtubule Density Chromatin Condensation Mitotic IndexC

    DMSO (–)-dictyostatin 1a 1b

    6-epi-dictyostatin1a1b

    0 1 2 30

    200

    400

    log nM

    mea

    n pi

    xel i

    nten

    sity

    , nu

    clea

    r reg

    ion

    0 1 2 30

    20

    40

    60

    log nM

    %co

    nden

    sed

    nucl

    ei

    0 1 2 30

    20

    40

    60

    log nM

    %ph

    osph

    o-hi

    ston

    eH

    3po

    sitiv

    ece

    lls

    OPMB

    O(MeO)2OP

    OTBS

    TBSO

    O

    I

    OH

    TBSO

    O

    I

    +

    4 top fragmentC18–C26

    a

    5

    b–e

    6

    O

    TBSO

    I

    3 middle fragmentC10–C17

    TBS

    O

    TBSO

    O

    R

    O

    OTBSI

    OTBSTBSO

    ClO

    7a,b bottom fragmentsC1–C9, both epimers at C6

    f

    8a,b, R = CH2OTBS9a,b, R = CHO

    O

    HO

    OH

    O

    OH OH

    O

    TBSO

    O

    O

    OH OTBS

    10a,b

    TBS TBS

    g-i j

    1a, C6-(R), Me (down)1b, C6-(S), Me (up)C25-C26 saturated

    (–)-dictyostatin, C6-(R), Me (down)C25-C26 unsaturated

    9

    1625

    26

    6 6 6

    6

    A

    Vollmer et al. Figure 1

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • (–)-dictyostatin

    DMSO

    PTX (10 μM)5 μM10 μM20 μM

    1a(–)-dictyostatin 1b

    C

    (–)-dictyostatin

    0 1 2 30

    200

    400

    600

    800

    mea

    n pi

    xel i

    nten

    sity

    , nu

    clea

    r reg

    ion

    vincristine

    0 1 2 30

    200

    400

    600

    800

    log nM

    1a

    0 1 2 30

    200

    400

    600

    800

    1b

    0 1 2 30

    200

    400

    600

    800

    B

    1a 1b

    A

    log nM log nM log nM

    vincristine

    0 10 20 30

    0.0

    0.4

    0.8

    2.5 oC 2.5 oC30 oC

    Abs

    350

    nm

    time (min)

    0 10 20 30

    0.0

    0.4

    0.8

    2.5 oC 2.5 oC30 oC

    0 10 20 30

    0.0

    0.4

    0.8

    2.5 oC 2.5 oC30 oC

    time (min) time (min)

    Vollmer et al. Figure 2

    on June 7, 2021. © 2011 A

    merican A

    ssociation for Cancer R

    esearch. m

    ct.aacrjournals.org D

    ownloaded from

    Author m

    anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

    Author M

    anuscript Published O

    nlineFirst on A

    pril 13, 2011; DO

    I: 10.1158/1535-7163.MC

    T-10-1048

    http://mct.aacrjournals.org/

  • (+)-discodermolide

    0.0 0.2 0.4 0.6 0.8 1.00.0

    0.5

    1.0

    1.5

    2.0

    affected fraction

    Com

    bina

    tion

    Inde

    x

    6-epi-dictyostatin

    0.0 0.2 0.4 0.6 0.8 1.00.0

    0.5

    1.0

    1.5

    2.0

    affected fraction

    Com

    bina

    tion

    Inde

    x

    25,26-dihydro-dictyostatin

    0.0 0.2 0.4 0.6 0.8 1.00.0

    0.5

    1.0

    1.5

    2.0

    affected fraction

    Com

    bina

    tion

    Inde

    x

    6-epi-25,26-dihydro-dictyostatin

    0.0 0.2 0.4 0.6 0.8 1.00.0

    0.5

    1.0

    1.5

    2.0

    affected fraction

    Com

    bina

    tion

    Inde

    x

    Vollmer et al. Figure 3

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/

  • A. B. C. D.

    E.

    ISV

    DA

    DLAV

    0

    50

    100

    totalISV

    area

    (%ofvehiclecontrol)

    6-epi- 1a 1bdictyostatin

    1 μM2.5 μM5 μM12.5 μM25 μM

    Vollmer et al. Figure 4

    on June 7, 2021. © 2011 A

    merican A

    ssociation for Cancer R

    esearch. m

    ct.aacrjournals.org D

    ownloaded from

    Author m

    anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

    Author M

    anuscript Published O

    nlineFirst on A

    pril 13, 2011; DO

    I: 10.1158/1535-7163.MC

    T-10-1048

    http://mct.aacrjournals.org/

  • Published OnlineFirst April 13, 2011.Mol Cancer Ther Laura L Vollmer, Maria Jimenez, Daniel P Camarco, et al. activity in zebrafish embryosactivity against epothilone B resistant cells and antiangiogenic A simplified synthesis of novel dictyostatin analogs with in vitro

    Updated version

    10.1158/1535-7163.MCT-10-1048doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mct.aacrjournals.org/content/suppl/2011/04/14/1535-7163.MCT-10-1048.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://mct.aacrjournals.org/content/early/2011/04/13/1535-7163.MCT-10-1048To request permission to re-use all or part of this article, use this link

    on June 7, 2021. © 2011 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 13, 2011; DOI: 10.1158/1535-7163.MCT-10-1048

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-10-1048http://mct.aacrjournals.org/content/suppl/2011/04/14/1535-7163.MCT-10-1048.DC1http://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/early/2011/04/13/1535-7163.MCT-10-1048http://mct.aacrjournals.org/