Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Sep 15.
Published in final edited form as: Bioorg Med Chem. 2011 Jul 30;19(18):5446–5453. doi: 10.1016/j.bmc.2011.07.054

Synthesis and characterization of tritylthioethanamine derivatives with potent KSP inhibitory activity

Delany Rodriguez a, Chinnasamy Ramesh b, Lauren H Henson a, Lori Wilmeth a, Bj K Bryant b, Samuel Kadavakollu b, Rebecca Hirsch a, Johnelle Montoya a, Porsha R Howell a, Jon M George b, David Alexander b, Dennis L Johnson b, Jeffrey B Arterburn b, Charles B Shuster a,
PMCID: PMC3171608  NIHMSID: NIHMS316121  PMID: 21855351

Abstract

Assembly of a bipolar mitotic spindle requires the action of class 5 kinesins, and inhibition or depletion of this motor results in mitotic arrest and apoptosis. S-trityl-L-cysteine is an allosteric inhibitor of vertebrate kinesin spindle protein (KSP) that has generated considerable interest due to its anticancer properties, however, poor pharmacological properties have limited the use of this compound. We have modified the triphenylmethyl and cysteine groups, guided by biochemical and cell-based assays, to yield new cysteinol and cysteamine derivatives with increased inhibitory activity, greater efficacy in model systems, and significantly enhanced potency against the NCI60 tumor panel. These results reveal a promising new class of conformationally-flexible small molecules as allosteric KSP inhibitors for use as research tools, with activities that provide impetus for further development as antitumor agents.

Keywords: Kinesin Spindle Protein, KSP, Mitosis, Kinesin, Anti-mitotic, Cell division

1. Introduction

Natural and synthetic small molecules that target the microtubule cytoskeleton have proven invaluable for investigating the mechanisms of cell division. Modulators of microtubule polymerization (such as colcimid and paciltaxel) interfere with cell division by altering microtubule dynamics, resulting in extended mitotic arrest and cell death.5, 6 Microtubule disrupters have proven to be effective chemotherapeutics, but because microtubules are essential for many cellular functions, their toxicity imposes limitations to their use as anti-cancer agents. More recently, attention has turned toward kinesins as potential targets for anti-proliferative drugs.711 There are roughly 14 classes of kinesin motors,12 whose functions range from vesicular transport to cell division, and RNA interference analysis of 25 Drosophila kinesins revealed that four members were involved in mitotic spindle assembly.13 Most prominent of these is the class five kinesin, Kinesin Spindle Protein (KSP; also known as Kif11 or Eg5), a plus end-directed motor that separates the spindle poles early in mitosis.14 KSP is antagonized by a minus end-directed kinesin known as KifC1,15 and inhibition of KSP results in spindle collapse and formation of a monopolar spindle. Because chromosomes are unable to establish bioriented attachments to the spindle, the cell arrests in mitosis, and eventually dies by apoptosis.16 Although there are reports that KSP plays a role in neuronal development,17 there are no other known functions outside of mitosis in adults, making KSP an attractive target for chemotherapeutic intervention.

Anticancer drug screening and chemical biological approaches have identified a number of KSP inhibitors, including monastrol,18 S-trityl-L-cysteine (STLC, 1),19, 20 3QC,21 MK-0731,22 ispinesib,23 and K85824 (Figure 1) that exhibit no activity against the other kinesin family members. The allosteric site located between α helix 3 and Loop 5 of the KSP motor domain in vertebrate organisms has been identified as the common site of action for monastrol, 1 and ispinesib.1, 4, 2529 The specificity of these compounds for KSP is associated with the extended Loop 5 found only in class 5 kinesins.29 Kinetic analyses indicate that these allosteric inhibitors act through a mechanism whereby ADP and motor rebinding to the microtubule are blocked.16, 23, 30 Loop 5 undergoes dynamic conformational shifts during the ATP binding and hydrolysis cycle, and it is thought that interactions with small molecules such as monastrol “lock” this loop into an ADP bound-like conformation.1, 4, 31, 32 While monastrol has not demonstrated efficacy at pharmacologically relevant concentrations, several compounds that exhibit anticancer activity have advanced to clinical trials.22, 3338 However, recent characterization of drug-resistant variants of KSP raises concern that tumor cells may develop resistance over time.1, 3, 4

Figure 1.

Figure 1

Representative structures of known KSP inhibitors, including STLC, monastrol, K858, 3QC, ispinesib, and MK-0731.

The cysteine derivative 1 represents a relatively simple scaffold with potent and reversible KSP inhibitory activity. Recent structure-activity analyses of STLC derivatives have been described and revealed the critical role of the trityl group for inhibitory activity.39, 40 The amphiphilic character of 1 results in poor water solubility and reduced permeability that affect bioavailability. We were interested in identifying related structures with increased inhibitory activity and improved physicochemical properties that would confer better water solubility and other drug-like characteristics. Herein, we describe the development of a new class of small, conformationally flexible KSP inhibitors based on the 2-(tritylthio)ethanamine scaffold. These compounds were effective inhibitors of human KSP in both biochemical and cell-based assays, as well as in nonmammalian model organisms. Two of the lead thioethanamine compounds 5 and 7 exhibited elevated anti-proliferative activity against the NCI60 panel of tumor cell lines and demonstrate the potential of this scaffold for the development of new anticancer therapeutics.

2. Chemistry

A series of STLC analogs have recently been evaluated in attempts to characterize structure-activity effects on KSP inhibition.3941 While structural alterations of the trityl group generally result in reduced inhibitory activity, the substitution of methyl, chloro, and methoxy groups in the para-position of the triphenylmethyl appendage was shown to increase the potency of STLC analogs. Carboxylic amides were found to function with similar inhibitory concentration values to the acids, but have not exhibited increased potency in the cellular assays.42 Additionally, the individual R- and S-enantiomers of trityl-cysteine derived from L- and D-cysteine have previously been shown to inhibit KSP with equivalent potency.16 Therefore, we focused our investigation on a series of novel thioethanamine compounds 47 related to 1, but lacking the carboxylic group, seeking to achieve increased KSP inhibitory properties, more favorable physicochemical characteristics and increased cell permeability by eliminating the zwitterionic structure of the parent compound. The individual enantiomers of 4′-methoxytrityl derivatives 2 and 3 were prepared to allow comparison of their activities (Figure 2) with 47.

Figure 2.

Figure 2

Enantiomers of S-4-methoxytritylcysteine evaluated as KSP inhibitors.

The thioethanamine derivatives were synthesized by the procedures shown in Scheme 1. Compounds 4–6 were prepared by in situ reduction of cysteine to cysteinol with BH3-THF, followed by quenching the excess borane reagent with DMF and selective S-alkylation with the appropriate trityl chloride. This efficient, two-step procedure avoids the formation of disulfide byproducts, and provided convenient access to the enantiomeric compounds 5 and 6 derived from L- and D-cysteine respectively. The achiral ethanamine derivative 7 was prepared using the published procedure for S-alkylation of cysteamine with 4-methoxytrityl chloride in trifluoroacetic acid. 43

Scheme 1.

Scheme 1

a

a Reagents and conditions: (i) BH3-THF 0 °C–RT, DMF quench ii) 4, 88% yield from L-cysteine and trityl chloride; 5, 91% yield from L-cysteine and 4-methoxytrityl chloride; 6, 92% yield from D-cysteine and 4-methoxytrityl chloride; (iii) 7, 96% yield from 4-methoxytrityl chloride, CF3CO2H.43

3. Results

Compounds 1–7 were tested for their ability to block human KSP activity in a biochemical ATPase assay44 and a phenotypic assay in cultured HeLa cells (Table I). The two assays provided a complementary assessment of direct inhibition of the motor, as well as indirectly reflecting membrane permeability and demonstrating efficacy in whole cells. The parent molecule 1 blocked the ATPase activity of recombinant human KSP with an IC50 of 0.62 μM, and in mitotic cells generated the stereotypical arrangement of microtubules radiating from the collapsed spindle poles with chromosomes arranged in a rosette pattern (Figure 3, Panel G). The 4′-methoxytrityl-derivative 2 exhibited increased potency as expected from recent reports.39, 40 The enantiomeric compound 3 derived from D-cysteine gave similar results in both assays, confirming the lack of stereochemical effects on inhibitory activity previously demonstrated for 1.16

Table 1.

IC50 Values for Inhibition of Microtubule-Stimulated KSP ATPase Activity and EC50 Values for Mitotic Arrest in Cell-Based Assays for Compounds 17.

Compound ATPase activity [μM] Cell-based assay [μM]
1 0.62 1.72
2 0.27 0.23
3 0.23 0.60
4 0.31 0.294
5 0.127 0.190
6 0.130 0.160
7 0.136 0.115

Figure 3.

Figure 3

STLC analogs block bipolar spindle formation without affecting interphase microtubule organization. HeLa cells were incubated in the presence of 0.1% DMSO (A and F) or 1 μM STLC 1 (B and G), 4 (C& H), 5 (D& I), 7 (E and J) and then fixed and stained for microtubules (green) and DNA (blue).

Evaluation of the inhibitory activity of the reduced cysteinol derivative 4 revealed an increase in potency associated with transformation of the carboxylic acid functional group to a primary alcohol that was comparable to incorporation of the 4′-methoxy-trityl group in 2. An additive effect was observed for 4-methoxy-substituted cysteinol compound 5 that incorporates both of these modifications to achieve increased KSP inhibitory activity in both the biochemical and cellular assays. The enantiomer 6 derived from D-cysteine displayed equivalent activity in both biochemical and cell-based assays. The simple ethanamine compound 7 derived from cysteamine displayed the highest potency with an EC50 = 115 nM.

The parent compound 1 affects bipolar spindle formation without altering interphase microtubule dynamics or organization (Figure 3B).16 As illustrated in Figure 3, interphase microtubule morphology was normal for compounds 4,5 and 7 tested in the presence of 1 μM compound (Figure 3C–E), whereas spindle morphology mirrored that of the parent compound (Figure 3H–J). Additionally, it has been established that 1 displays no activity against other kinesin family members.16, 19 Compounds 4 and 5 were similarly tested for inhibitory activity against a selected panel of mitotic kinesins (mammalian centromere-associated kinesin (MCAK), centromeric protein E (CENP-E) and chromokinesin), with no measurable inhibition detected (not shown), suggesting that selectivity for class 5 kinesins was retained by the cysteinol derivatives.

KSP is a bipolar, homotetrameric motor whose nucleotide hydrolysis cycle is disrupted when treated with allosteric inhibitors such as 1, monastrol and ispinesib.16, 23, 30 Kinetic analysis of motor inhibition predicts that KSP remains locked in the ADP bound state,16, 23, 45 blocking motor rebinding and leading to a predicted loss of motor from spindle microtubules. To determine whether the cysteinol derivatives had a similar effect on KSP dynamics, cells were treated with 1 or 5 and probed for KSP localization with anti-KSP antibodies, using EB1 antibodies to label microtubules (Figure 4). Similar to reports of monastrol-treated cells,46 KSP was observed clustering at the center of the monopole upon treatment with either compound (Figure 4A, D, G, J, M and P). Both compounds displayed a dose-dependent depletion of KSP from the spindle, with 5 exhibiting a 1.63-fold increase in clearance of KSP over 1 (Figure 4FJ, M and P), consistent with the biochemical- and cell-based assays (Table 1). Thus, the behavior of KSP in the presence of this cysteinol derivative was consistent with a mechanism of action by which these allosteric modulators promote disengagement of the motor from its microtubule track.

Figure 4.

Figure 4

Dose-dependent depletion of KSP from monopolar spindles in 1 and 5-treated cells. HeLa cells were incubated in either 1 (A–I) or 5 (J–R) for 4 hrs and the fixed and processed for KSP (green), the microtubule-binding protein EB1 (red) and DNA (blue) localization, and images were acquired using equivalent exposure times for the green channel.

The hydrophobic domain created by an extended loop 5 and alpha helix 3 in the KSP motor domain accommodates a structurally diverse group of allosteric KSP inhibitors (Figure 1) and is unique to class 5 kinesins.1 Moreover, it has been proposed that the nonpolar nature of this pocket is unique to vertebrates.1 Sequence alignments comparing the binding pockets of human, Xenopus laevis, and sea urchin Strongylocentrotus purpuratus revealed a high degree of sequence conservation between urchin and vertebrate KSP proteins, particularly at residues identified by mutational analysis to be critical for inhibition by monastrol and STLC analogs (Figure 5A).14 In contrast, the Drosophila homolog Klp61F was more divergent, particularly within loop 5. To determine whether 1 or the cysteamine- or cysteinol derivatives were effective in sea urchin eggs or Drosophila S2 cells, cells were treated over a three log order range of concentrations, and scored for bipolar spindle formation by immunofluorescence. Compounds (1–7) were screened in S2 cells, however, no activity was detected at doses up to 100 μM and treatments extending up to 7 hours (not shown).

Figure 5.

Figure 5

Efficacy of STLC derivatives in non-vertebrate organisms. A) Alignment of the loop 5 (top) and helix 3 (bottom) of kinesin 5 homologs from Homo sapiens (NM_004523), Xenopus laevis (NM_001085956), Strongylocentrotus purpuratus (AF292395), and Drosophila melanogaster (NM_057470). Asterisks denote the residues reported to be required for drug inhibition.14 B) Micrographs of S. purpuratus eggs incubated in 100 μM Monastrol, 1, or 5 and 0.1% DMSO as control. Eggs were fixed and processed for microtubule (green) and DNA (blue) localization.

In contrast, monopolar spindles were detected in Strongylocentrotus purpuratus eggs treated during the first cell cycle following fertilization (Figure 5B). Compound 5 blocked bipolar spindle formation with an EC50 of 28 μM, whereas the parent molecule 1 and monastrol were weakly active with EC50 ≥ 100 μM. The response to all compounds was phenotypically identical, causing spindle collapse following nuclear envelope breakdown resulting in a large monopolar spindle (Figure 5B, panels B-D). Thus, the observed activity of these compounds in the highly ionic environment of seawater provides an additional assessment of the compounds biological efficacy using an invertebrate model organism, and the The possible binding modes of derivatives 5–7 were evaluated by molecular docking experiments using the crystal structure of human KSP (PBD: 2FME), and Genetic Optimization for Ligand Docking (GOLD) scores were derived from 30 poses for each compound, with the highest differences in activity between the compounds 1 and 5 in sea urchin eggs parallel the activities measured in human cultured cells.

Compounds 5 (MSTCO, NSC 747880) and 7 (MSTNH2, NSC 753791) were evaluated for anti-proliferative activity against the NCI60 tumor panel. Growth inhibitory concentrations (GI50) of 5 and 7 ranged from 10 nM to 3 μM across the panel, with average GI50’s of 360 nM and 350 nM for 5 and 7, respectively (Table 2). This represented roughly a 143- and 35-fold average increase in potency over Monastrol (NSC 716782) and 1 (NSC 83265), respectively. There were, however, individual tumor lines such as melanoma M14 where 5 and 7 displayed a 1200-fold increase in activity over monastrol and the ovarian SK-OV-3 line where 5 and 7 displayed a 1250-fold increase in potency over the parent compound 1. Thus, the tritylthioethanamine derivatives 5 and 7 had activities over a broad range of tumor types and represented a significant increase in antiproliferative potency.

Table 2.

GI50 values from NCI60 panel screens* of Monastrol, 1, 5 and 7

Cell line Monastrol 1 5 7
Leukemia CCRF-CEM 31.6 1.58 0.1 0.16
Leukemia HL-60(TB) 25.1 2.51 0.19 0.25
Leukemia K-562 31.6 1.58 0.06 0.04
Leukemia MOLT-4 31.6 7.94 0.28 0.50
Leukemia RPMI-8226 31.6 3.16 0.03 0.31
Leukemia SR 31.6 6.31 0.01 0.13
NSC Lung A549/ATCC 50.1 5.01 0.33 0.10
NSC Lung EKVX 63.1 3.16 0.51 0.39
NSC Lung HOP-62 63.1 19.9 0.51 0.66
NSC Lung NCI-H226 50.1 100 2.75 1.74
NSC Lung NCI-H23 63.1 2.51 0.26 0.51
NSC Lung NCI-H322M 39.8 1.26 0.11 0.06
NSC Lung NCI-H522 31.6 0.5 0.22 0.04
Colon COLO 205 31.6 3.16 0.08 0.21
Colon HCC-2998 39.8 2.51 0.05 0.24
Colon HCT-116 31.6 0.5 0.03 0.04
Colon HCT-15 39.8 2.51 0.21 0.05
Colon HT29 79.4 3.98 0.34 0.35
Colon KM12 31.6 5.01 0.22 0.31
Colon SW-620 39.8 1.58 0.07 0.07
CNS SF-268 63.1 3.98 0.11 0.15
CNS SF-295 31.6 0.5 0.28 0.18
CNS SF-539 50.1 0.79 0.26 0.25
CNS SNB-19 79.4 2.51 0.25 0.34
CNS SNB-75 39.8 1.99 0.26 0.60
CNS U251 31.6 1 0.19 0.05
Melanoma LOX IMVI 79.4 6.31 0.49 0.50
Melanoma MALME-3M 63.1 1.26 0.36 0.22
Melanoma M14 25.1 0.398 0.02 0.03
Melanoma SK-MEL-2 31.6 1.5 0.24 0.06
Melanoma SK-MEL-28 79.4 12.6 0.62 0.64
Melanoma SK-MEL-5 39.8 0.63 0.06 0.05
Melanoma UACC-257 63.1 1.26 0.54 0.70
Melanoma UACC-62 39.8 1.99 0.25 0.40
Ovarian IGROV1 50.1 3.98 0.26 0.15
Ovarian OVCAR-3 50.1 2.51 0.2 0.15
Ovarian OVCAR-4 63.1 12.6 1.62 1.40
Ovarian OVCAR-5 79.4 100 0.39 0.57
Ovarian OVCAR-8 63.1 1.99 0.42 0.12
Ovarian SK-OV-3 63.1 100 0.08 0.08
Renal 786-0 63.1 1.26 0.44 0.40
Renal ACHN 79.4 19.9 0.49 0.69
Renal CAKI-1 63.1 15.8 0.45 0.19
Renal SN12C 39.8 3.16 0.32 0.13
Renal TK-10 100 6.31 0.46 1.56
Renal UO-31 100 100 1.1 0.50
Average 51.5 12.6 0.36 0.35
*

cell lines shown are those common to all four screens

The possible binding modes of derivatives 5–7 were evaluated by molecular docking experiments using the crystal structure of human KSP (PBD:2FME), and Genetic Optimization for Ligand Docking (GOLD) scores were derived from 30 poses for each compound, with the highest scores illustrated in Figure 6. All three molecules docked into the binding pocket in the same manner, with the ammonium group forming hydrogen-bonding interactions with GLU 116 and GLY 117, consistent with previous docking studies for 1.39 Indeed, a structural of overlay of 1 with 7 revealed an identical alignment within the binding pocket (Figure 6D). Similarly, the aromatic rings of the trityl groups for all three molecules fit into the previously described hydrophobic pockets (Hy1, comprised of TYR 211 and ALA 218 of α3; Hy2, comprised of ALA 133 of α2 at the base of loop 5; and Hy3, comprised of ILE 136 from α2, LEU 160, LEU 214 from α3). The highest scoring poses of 5, 6 and 7 gave Gold scores of 85.74, 83.61 and 85.79 respectively. No specific interactions of the primary alcohol groups of 5 and 6 with the backbone of KSP were identified in these binding models.

Figure 6.

Figure 6

Docking studies of compounds 5–7. Highest scoring poses of 5 (A), 6 (B) and 7 (C) in the allosteric binding site of KSP (PDB: 2FME). All inhibitors bind to the same pocket formed by loop 5, α2 and α3. Helix α3 is shown to the left and loop 5 defines the right side and upper lid of the site. D) Overlay of optimal poses for 1 and 7; E) overlay of the R- and S- enantiomers 5 and 6.

Previously described studies as well as data reported here (Table 1) demonstrate an increase of potency for methoxytrityl derivatives of 1.39, 40 While no specific interactions were identified for the methoxy group and any side chains within the binding pocket, 5 and 7 demonstrated a slight preference for positioning the methoxyphenyl group in pocket Hy3 (Figures 6A and C), while 6 preferred to occupy Hy1 (Figure 6B), as illustrated by structural overlays of 5 and 6 (Figure 6E). However, no poses showed the methoxyphenyl ring occupying pocket Hy2, the area nearest Loop 5.

4. Discussion

The potential of kinesin inhibitors for use in the clinical arena is an area of active investigation, with several candidate drugs in clinical trials.34, 37, 38, 47 This study has identified S-trityl- derivatives of cysteinol and cysteamine as promising classes of selective KSP inhibitors. These compounds incorporate a small, flexible thioethanamine scaffold and exhibited increased activity as inhibitors of both purified recombinant motor activity as well as bipolar spindle assembly (Table 1). The increased potency of para-substitution of the trityl group in derivatives of 1 was maintained in the tritylthioethanamine compounds developed in this study. Previous modeling studies of 1 with KSP have indicated that these substituents are oriented towards the Hy3 hydrophobic pocket.39 The tritylthioethanamine derivatives described here are able to establish additional interactions of the methoxy substituent with hydrophobic pockets 1 and 3, and preserve hydrogen bonding with the polar backbone contacts GLU116 and GLY117 that were identified as part of the pharmacophore.

The steric volume and hydrophobicity of the triphenylmethyl group are major determinants of the physicochemical and pharmacological properties of these compounds. It is intriguing to note the presence of the trityl moiety in a variety of compounds that have been identified as potential anticancer agents and function through different mechanisms of action.48 5′-O-Tritylinosine functions an an allosteric inhibitor of the angiogenic enzyme thymidine phosphorylase.49 The antifungal imidazole agent clotrimazole and synthetic analogs exhibit anticancer activity in cell culture and animal models, with treatment resulting in G1-phase arrest in the cell cycle, affecting intracellular Ca2++ levels and inducing detachment of mitochondrial-bound hexokinase that inhibits glycolysis.5053 Triphenylmethylamide derivatives have been shown to exhibit activity against multiple melanoma cell lines, causing G1-phase arrest and inducing apoptosis.54, 55 The related trityl-phosphonate compound TPMP-III-2 exhibits anticancer activity and induces arrest in the M-phase of the cell cycle with characteristic formation of fragmented mitotic spindles, through inhibition of tubulin polymerization.48 In contrast, inhibition of KSP by STLC derivatives results in M phase cell cycle arrest and is characterized by the formation of the monopolar spindle. Synthetic diphenyl-heterocyclic carbinols derivatives were recently shown to exhibit antiproliferative effects against carcinoma cell lines.56 The cysteamine and cysteinol derivatives described herein are shown to be effective inhibitors of KSP and exhibit the archetypal monopolar phenotype, demonstrating the importance of both the trityl- and ethanamine elements of the pharmacophore.

Methoxytritylethanamine derivatives demonstrated improved efficacy in blocking KSP ATPase activity and bipolar spindle assembly in HeLa cells (Table 1). The zwitterionic character and neutral overall charge of carboxylic derivatives 1–3 represents major physicochemical differences, with respect to compounds 4–7, that possess positive charges due to their protonated amine groups at physiological pH. In order to evaluate our lead compounds against an expanded panel of tumor cell types, 5 and 7 were submitted to the NCI Developmental Therapeutics Program to be screened against the NCI60 tumor panel. Examination of the growth inhibitory activities in cell lines common to screens of monastrol, 1, 5 and 7 revealed that the ethanamine derivatives showed greater efficiency in blocking tumor cell growth in comparison to 1 and monastrol (Table 2). Leukemia and colon tumor cell lines were generally the most susceptible, with average GI50 values ≤ 230 nM for both compounds. Indeed, the differential sensitivity of hematological neoplasms to KSP inhibitors has been noted for other compounds, several of these compounds have advanced through preclinical evaluation. 47, 5861 Thus, the cysteinol and cysteamine scaffolds hold promise for further development as therapeutically useful antiproliferative agents.

As an additional means of studying the interaction between these compounds and the KSP motor domain, analogs were tested against diverse species that contain varying degrees of conservation within the inhibitor-binding pocket (Figure 5). A single report has described monastrol- and 1 efficacy in a brown algae,62 but there have been no other examples of KSP inhibitors functioning in invertebrate cells. Examination of the Loop 5 and helix 3 reveal that while sea urchin KSP shares a high degree of identity with human KSP throughout the binding pocket, including the residues identified as being essential for inhibition by 1, as well as monastrol and ispinesib (Figure 5A, asterisks). In contrast, the Drosophila homolog Klp61F shares only partial identity with echinoderm and vertebrate loop 5, with notable variation at positions 116, 117, and 130, residues identified as crucial for monastrol- and STLC inhibition in human KSP.14 Indeed, whereas we found that derivatives of STLC were effective in sea urchin eggs (Figure 5B), Drosophila S2 cells were resistant to all compounds tested (not shown). However, because Klp61F contains a serine residue position 129 that is a proline in vertebrate KSP homologs, it is possible that this variation alters the structure of Loop 5 and thus Hy2. Thus, understanding the structural differences between vertebrate and invertebrate KSP homologs may lead to opportunities for the rational design of inhibitors that selectively target non-mammalian pathogens.

5. Conclusions

Derivatization of both the cysteine and triphenylmethyl groups of known KSP inhibitor 1 has provided a new class of compounds that display increased potency in both biochemical and cell-based assays. The structure-based mechanism of action exhibited by this class of triphenylmethyl derivatives with respect to 1 is maintained by these compounds, eliciting cell cycle arrest and producing the characteristic monoaster spindle phenotype that results from KSP inhibition. The cysteinol and cysteamine derivatives 5 and 7 displayed an average thirty five-fold increase anti-proliferative activity against the NCI60 panel of cancer cell lines in comparison with 1, as well as broadened species specificity. The potent activity and conformational flexibility of these small molecule allosteric inhibitors of KSP offers real promise for applications as research tools and potential for further development as clinically useful antitumor agents.

6. Experimental

6.1 General Methods

All reactions were performed in an efficient fume hood. Commercially available solvents and reagents were used without further purification. Preparative chromatographic separations were performed using medium pressure flash chromatography and ethyl acetate/hexanes or methanol/dichloromethane as eluent. Reverse phase chromatography employed C-18 columns and water-acetonitrile or water-methanol mobile phase. NMR spectra were acquired at ambient temperatures (18 ± 2 °C) unless otherwise noted. Reactions were monitored by thin-layer chromatography on silica gel (60 Å pore size, 5–17 μm) polyester backed sheets that were visualized under a UV lamp, iodine vapor, phosphomolybdic acid, or anisaldehyde. The 1H NMR spectra in CDCl3 were referenced to TMS unless otherwise noted. The 13C {1H} NMR spectra were recorded at 75 or 100 MHz and referenced relative to the 13C {1H} peaks of the solvent. NMR spectra are reported as ppm (δ), (multiplicity, coupling constants (Hz), and number of protons). Infrared spectra were recorded as KBr pellets or neat films and are reported in cm−1. Melting points are uncorrected. The purity of all compounds used in biological studies was determined to be >95% by analytical HPLC equipped with Photodiode Array (PDA) and ESI-MS detection. The compounds (1mg/mL CH3CN, 20 μL) were injected into a Waters Symmetry® C18 5 μm 3.0 × 150 mm column and eluted as specified. Compound STLC (1) was obtained from Novabiochem, Compounds 2,40 3,40 and 743 were prepared following reported procedures. The other compounds were synthesized as follows.

6.1.1 General Procedure for the Preparation of S-Triarylmethyl-cysteinol (4–6)

Borane-THF (4 mL, 4 mmol) was added dropwise to cysteine (0.121 g, 1 mmol) in dry THF (5 mL) at 0 °C under an argon atmosphere, and stirred at room temperature for 7 h. The reaction mixture was quenched with dry DMF (1 mL) and stirred for 1 h. Triarylmethylchloride (0.5 mmol) was added to the reaction mixture and stirred at room temperature for 3 h. The volatiles were removed in vacuo. The residue was dissolved in CH2Cl2 (20 mL), washed with H2O (20 mL), saturated NaCl solution (10 mL), and dried over Na2SO4. The solvents were evaporated under reduced pressure and the residue was purified by silica gel chromatography using CH3OH/CH2Cl2 (3:97) as eluent to isolate the product.

6.1.2 (R)-2-amino-3-(tritylthio)propan-1-ol (4)

From L-cysteine and trityl chloride; yield (88%) as colorless viscous oil. IR (KBr, cm−1): 3425, 3054, 2917, 1593, 742; 1H NMR (300 MHz, CDCl3) δ 7.44–7.39 (m, 6H), 7.31–7.17 (m, 9H), 3.38 (dd, J = 10.86, 4.26 Hz, 1H), 3.15 (dd, J = 10.71, 6.89 Hz, 1H), 2.61–2.53 (m, 1H), 2.31 (dd, J = 12.47, 5.14 Hz, 1H), 2.20 (dd, J = 12.47, 7.63 Hz, 1H), 1.52 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 144.60, 129.50, 127.84, 126.67, 66.77, 65.53, 51.99, 36.62. HPLC-MS: Elution with CH3CN/H2O (20:80) exhibited a single peak at 3.92 min. ESI-MS m/z [ES+] calcd for C22H24NOS [M+H]+ 350.44; found 350.16.

6.1.3 (R)-2-amino-3-((4-methoxyphenyl)diphenylmethylthio)propan-1-ol (5)

From L-cysteine and 4-methoxytrityl chloride; yield (91%) as colorless viscous oil. [α]20 D +6.91° (c 0.57, CHCl3); IR (Neat, cm−1): 3447, 2925, 1508, 1250, 1033; 1H NMR (300 MHz, CDCl3) δ 7.42–7.17 (m, 12H), 6.81 (d, J = 8.8 Hz, 2H), 3.79 (s, 3H), 3.44–3.39 (dd, J = 10.71, 4.11 Hz, 1H), 3.21–3.15 (m, 1H), 2.65–2.55 (m, 1H), 2.36–2.19 (m, 2H), 1.55 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 158.10, 144.95, 136.76, 130.72, 129.46, 127.87, 126.64, 113.14, 66.36, 65.72, 55.11, 52.06, 36.85. HPLC-MS: Elution with CH3CN/H2O (20:80), exhibited a single peak at 2.18 min. ESI-MS m/z [ES+] calcd for C29H26NO2S [M+H]+ 380.09; found 380.16). HRMS calcd 380.1679, found: 380.1678.

6.1.4 (S)-2-amino-3-((4-methoxyphenyl)diphenylmethylthio)propan-1-ol (6)

From D-cysteine and 4-methoxytrityl chloride; yield (92%) as colorless viscous oil. [α] 20D -6.04° (c 0.57, CHCl3); IR (Neat, cm−1): 3447, 2925, 1508, 1250, 1033; 1H NMR (300 MHz, CDCl3) δ 7.42–7.17 (m, 12H), 6.81 (d, J = 8.8 Hz, 2H), 3.79 (s, 3H), 3.44–3.39 (dd, J = 10.71, 4.11 Hz, 1H), 3.21–3.15 (m, 1H), 2.65–2.55 (m, 1H), 2.36–2.19 (m, 2H), 1.54 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 158.10, 144.89, 136.70, 130.72, 129.46, 127.87, 126.67, 113.14, 66.36, 65.72, 55.20, 52.06, 36.81. HPLC-MS: Elution with 20–80% CH3CN in H2O (gradient 1.5 % min−1), exhibited a single peak at 2.18 min. ESI-MS m/z [ES+] calcd for C29H26NO2S [M+H]+ 380.09; found 380.16).

6.2 Analysis of analogs using an in vitro kinesin end-point ATPase assay

Inhibitory activity of STLC analogs were measured using a microtubule-activated ATPase end-point assay from Cytoskeleton (Denver, CO). The reagents and kinesin motor domains were added to wells of a 96-well microtitre dish (Greiner Bio-One, Monroe, NC) in a total reaction volume of 30 μl. Reactions were started by the addition of 2mM ATP (Sigma Co. St Louis, MO), incubated at room temperature for 5 minutes, and terminated by the addition of 70 μl of CytoPhos. The reactions were incubated for an additional 10 minutes at room temperature and absorbance was measured at 650 nm using a microplate reader Elx 800™ (BIO-TEK® Winooski, VT) at 650 nm. All kinesin domain motors were used at a final concentration of 0.08 μg/μl. STLC and its derivatives were assayed over a range of concentrations between 0.25 μM to 10 μM. Controls of DMSO were used at a final concentration of 0.1%. All conditions were performed in triplicate, and raw data was entered into Graphpad Prism software, and IC50 values were calculated using a four parameter non-linear regression.

6.3 Mammalian cell culture

HeLa cells (American Type Culture Collection, Manassas, VA) were cultured in minimum essential medium with Earle’s BSS (Lonza, Walkersville, MD), supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, fungicide, 1.5 g/L sodium bicarbonate, and 1.0 mM sodium pyruvate (Sigma Chemical Co., St. Louis, MO) in 5% CO2 at 37°C in a humidified incubator. STLC and its derivatives were screened for KSP inhibition by incubating cells for four hours with either carrier alone (0.1% DMSO) or DMSO-solublized compound in doses ranging from 50 nM to 100 μM. Cells were then processed for tubulin and DNA localization and 100 mitotic cells were scored for bipolar spindle formation per condition. EC50 values were calculated using Graphpad Prism software using a four parameter non-linear regression as described above. Growth inhibitory activity of tumor cell lines (NCI60 panel) was performed by the Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, U.S.A.

6.4 Sea urchin embryo culture

Strongylocentrotus purpuratus sea urchins were obtained from Marinus (Long Beach, CA) and maintained in chilled aquarium at 10°C. Gametes were obtained by intracoloemic injection of 0.5M KCl and eggs were fertilized in artificial seawater in the presence of 2 mM 3-amino triazole to prevent hardening of the fertilization envelope. Twenty minutes after fertilization, eggs were stripped of their fertilization envelops by passage through 80 μm Nitex, and cultured either in the presence of 0.1% DMSO or KSP inhibitors. At the time of first mitosis, eggs were fixed and processed for microtubule and DNA localization as previously described.63 To generate EC50 values for KSP inhibitor activity in sea urchin eggs, embryos were treated over a three log order range of concentrations from 100 nM to 100 μM, processed for microtubule localization, and then scored for bipolar spindle formation (100 cells scored per condition).

6.5 Immunofluorescence

Hela cells were incubated in the presence of compounds dissolved in DMSO at the specified concentration for four hours and then fixed by immersion in 100% MeOH for a minimum of thirty minutes at −20°C before rehydration in phosphate-buffered saline (PBS) for ten minutes. Hela cells were blocked by incubation in PBS containing 5% Bovine Serum Albumin (PBS-BSA) for one hour at room temperature. Cells were then probed with either mouse anti-tubulin (Sigma, Co, St. Louis, MO), or mouse anti-KSP (Abcam, Cambridge, MA) in blocking buffer overnight at 4°C. To counterstain the microtubule cytoskeleton in KSP localization experiments, cells were also probed for the presence of the microtubule end-binding protein EB1 using a custom rabbit polyclonal antibody against whole recombinant human EB1 (Pro-Sci, Poway, CA). Primary antibodies were detected using AlexaFluor-conjugated secondary antibodies (Molecular Probes, Eugene OR). After washing, cells were mounted in 90% glycerol/1x PBS and imaged using a Zeiss 200M inverted microscope equipped with epifluorescence optics and an Apotome structured illumination module, a 100W mercury arc fluorescent light source and DAPI, FITC and TRITC filter sets. Images were acquired using a 63X 1.4 NA Plan-aprochromat objective, an Axiocam MrM 12 bit CCD camera driven by Axiovision 4.5 software. Images were exported as 8 bit tif images and figures were prepared using Adobe Photoshop® software.

6.6 In silico Docking Studies

All docking calculations were performed on a PC equipped with a 1.6 GHz Core 2 duo processor and 4GB of RAM, running Windows 7. Ligand structures were generated using Spartan’ 10.0 (Wavefunction Inc., Irvine, CA), including expected protonation states at physiological pH, and geometry optimized. Binding site preparation of the KSP complexes and docking studies were performed with Gold Suite 5.0.1 (The Cambridge Crystallographic Data Centre: Cambridge U.K., 2010) and Hermes 1.4.1. Docking was performed in the binding site of PDB code: 2FME. All the water molecules present in the binding site were allowed to be replaced by the ligand or to change orientation. The standard function Gold Score was used for ranking the poses, including 30 solutions for each ligand generated. Ligand Scout v 3.0 (Inte:Ligand GmbH: Vienna, Austria, 2010) was used for visual inspection of the docked poses and for producing three-dimensional visual representations.

Supplementary Material

01

Acknowledgments

The authors would like to thank Dr. Kathy Hanley for her assistance in statistical analyses. This work is supported by National Science Foundation MCB0818729 (CBS), National Center for Research Resources P20 RR16480 (JBA and CBS), the Howard Hughes Medical Institute’sUndergraduate Education Improvement Program at NMSU, the Cowboys for Cancer Research Foundation, and an Interdisciplinary Research Grant from the NMSU Vice President for Research (JBA and CBS). We acknowledge the NCI Developmental Therapeutics Program for providing biological data including NCI60 screening services, see http://dtp.cancer.gov/.

Footnotes

Supporting Information Available: 1H and 13C NMR spectra for compounds 3-7. This material is available free of charge via the Internet at www.elsevier.com

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Maliga Z, Mitchison TJ. BMC Chem Biol. 2006;6:2. doi: 10.1186/1472-6769-6-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Marshall CG, Torrent M, Williams O, Hamilton KA, Buser CA. Arch Biochem Biophys. 2009;484:1. doi: 10.1016/j.abb.2009.01.015. [DOI] [PubMed] [Google Scholar]
  • 3.Tcherniuk S, van Lis R, Kozielski F, Skoufias DA. Biochem Pharmacol. 2009 doi: 10.1016/j.bcp.2009.11.001. [DOI] [PubMed] [Google Scholar]
  • 4.Brier S, Lemaire D, DeBonis S, Forest E, Kozielski F. J Mol Biol. 2006;360:360. doi: 10.1016/j.jmb.2006.04.062. [DOI] [PubMed] [Google Scholar]
  • 5.Jordan MA, Wilson L. Nat Rev Cancer. 2004;4:253. doi: 10.1038/nrc1317. [DOI] [PubMed] [Google Scholar]
  • 6.Zhou J, Giannakakou P. Curr Med Chem Anticancer Agents. 2005;5:65. doi: 10.2174/1568011053352569. [DOI] [PubMed] [Google Scholar]
  • 7.Bergnes G, Brejc K, Belmont L. Curr Top Med Chem. 2005;5:127. doi: 10.2174/1568026053507697. [DOI] [PubMed] [Google Scholar]
  • 8.Duhl DM, Renhowe PA. Curr Opin Drug Discov Devel. 2005;8:431. [PubMed] [Google Scholar]
  • 9.Huszar D, Theoclitou ME, Skolnik J, Herbst R. Cancer Metastasis Rev. 2009;28:197. doi: 10.1007/s10555-009-9185-8. [DOI] [PubMed] [Google Scholar]
  • 10.Knight SD, Parrish CA. Curr Top Med Chem. 2008;8:888. doi: 10.2174/156802608784911626. [DOI] [PubMed] [Google Scholar]
  • 11.Zhang Y, Xu W. Anticancer Agents Med Chem. 2008;8:698. [PubMed] [Google Scholar]
  • 12.Lawrence CJ, Dawe RK, Christie KR, Cleveland DW, Dawson SC, Endow SA, Goldstein LS, Goodson HV, Hirokawa N, Howard J, Malmberg RL, McIntosh JR, Miki H, Mitchison TJ, Okada Y, Reddy AS, Saxton WM, Schliwa M, Scholey JM, Vale RD, Walczak CE, Wordeman L. J Cell Biol. 2004;167:19. doi: 10.1083/jcb.200408113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Goshima G, Vale RD. J Cell Biol. 2003;162:1003. doi: 10.1083/jcb.200303022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Blangy A, Lane HA, d’Herin P, Harper M, Kress M, Nigg EA. Cell. 1995;83:1159. doi: 10.1016/0092-8674(95)90142-6. [DOI] [PubMed] [Google Scholar]
  • 15.Mountain V, Simerly C, Howard L, Ando A, Schatten G, Compton DA. J Cell Biol. 1999;147:351. doi: 10.1083/jcb.147.2.351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Skoufias DA, DeBonis S, Saoudi Y, Lebeau L, Crevel I, Cross R, Wade RH, Hackney D, Kozielski F. J Biol Chem. 2006;281:17559. doi: 10.1074/jbc.M511735200. [DOI] [PubMed] [Google Scholar]
  • 17.Haque SA, Hasaka TP, Brooks AD, Lobanov PV, Baas PW. Cell Motil Cytoskeleton. 2004;58:10. doi: 10.1002/cm.10176. [DOI] [PubMed] [Google Scholar]
  • 18.Mayer TU, Kapoor TM, Haggarty SJ, King RW, Schreiber SL, Mitchison TJ. Science. 1999;286:971. doi: 10.1126/science.286.5441.971. [DOI] [PubMed] [Google Scholar]
  • 19.DeBonis S, Skoufias DA, Lebeau L, Lopez R, Robin G, Margolis RL, Wade RH, Kozielski F. Mol Cancer Ther. 2004;3:1079. [PubMed] [Google Scholar]
  • 20.Zee-Cheng KY, Cheng CC. J Med Chem. 1970;13:414. doi: 10.1021/jm00297a019. [DOI] [PubMed] [Google Scholar]
  • 21.Tarby CM, Kaltenbach RF, 3rd, Huynh T, Pudzianowski A, Shen H, Ortega-Nanos M, Sheriff S, Newitt JA, McDonnell PA, Burford N, Fairchild CR, Vaccaro W, Chen Z, Borzilleri RM, Naglich J, Lombardo LJ, Gottardis M, Trainor GL, Roussell DL. Bioorg Med Chem Lett. 2006;16:2095. doi: 10.1016/j.bmcl.2006.01.056. [DOI] [PubMed] [Google Scholar]
  • 22.Cox CD, Coleman PJ, Breslin MJ, Whitman DB, Garbaccio RM, Fraley ME, Buser CA, Walsh ES, Hamilton K, Schaber MD, Lobell RB, Tao W, Davide JP, Diehl RE, Abrams MT, South VJ, Huber HE, Torrent M, Prueksaritanont T, Li C, Slaughter DE, Mahan E, Fernandez-Metzler C, Yan Y, Kuo LC, Kohl NE, Hartman GD. J Med Chem. 2008;51:4239. doi: 10.1021/jm800386y. [DOI] [PubMed] [Google Scholar]
  • 23.Lad L, Luo L, Carson JD, Wood KW, Hartman JJ, Copeland RA, Sakowicz R. Biochemistry. 2008;47:3576. doi: 10.1021/bi702061g. [DOI] [PubMed] [Google Scholar]
  • 24.Nakai R, Iida S, Takahashi T, Tsujita T, Okamoto S, Takada C, Akasaka K, Ichikawa S, Ishida H, Kusaka H, Akinaga S, Murakata C, Honda S, Nitta M, Saya H, Yamashita Y. Cancer Res. 2009;69:3901. doi: 10.1158/0008-5472.CAN-08-4373. [DOI] [PubMed] [Google Scholar]
  • 25.Brier S, Lemaire D, Debonis S, Forest E, Kozielski F. Biochemistry. 2004;43:13072. doi: 10.1021/bi049264e. [DOI] [PubMed] [Google Scholar]
  • 26.Brier S, Lemaire D, DeBonis S, Kozielski F, Forest E. Rapid Commun Mass Spectrom. 2006;20:456. doi: 10.1002/rcm.2329. [DOI] [PubMed] [Google Scholar]
  • 27.Maliga Z, Kapoor TM, Mitchison TJ. Chem Biol. 2002;9:989. doi: 10.1016/s1074-5521(02)00212-0. [DOI] [PubMed] [Google Scholar]
  • 28.Maliga Z, Xing J, Cheung H, Juszczak LJ, Friedman JM, Rosenfeld SS. J Biol Chem. 2006;281:7977. doi: 10.1074/jbc.M511955200. [DOI] [PubMed] [Google Scholar]
  • 29.Turner J, Anderson R, Guo J, Beraud C, Fletterick R, Sakowicz R. J Biol Chem. 2001;276:25496. doi: 10.1074/jbc.M100395200. [DOI] [PubMed] [Google Scholar]
  • 30.Cochran JC, Gilbert SP. Biochemistry. 2005;44:16633. doi: 10.1021/bi051724w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Behnke-Parks WM, Vendome J, Honig B, Maliga Z, Moores C, Rosenfeld SS. J Biol Chem. 2011;286:5242. doi: 10.1074/jbc.M110.192930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kim ED, Buckley R, Learman S, Richard J, Parke C, Worthylake DK, Wojcik EJ, Walker RA, Kim S. J Biol Chem. 2010;285:18650. doi: 10.1074/jbc.M109.092072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Beer TM, Goldman B, Synold TW, Ryan CW, Vasist LS, Van Veldhuizen PJ, Jr, Dakhil SR, Lara PN, Jr, Drelichman A, Hussain MH, Crawford ED. Clin Genitourin Cancer. 2008;6:103. doi: 10.3816/CGC.2008.n.016. [DOI] [PubMed] [Google Scholar]
  • 34.Lee CW, Belanger K, Rao SC, Petrella TM, Tozer RG, Wood L, Savage KJ, Eisenhauer EA, Synold TW, Wainman N, Seymour L. Invest New Drugs. 2008;26:249. doi: 10.1007/s10637-007-9097-9. [DOI] [PubMed] [Google Scholar]
  • 35.Blagden SP, Molife LR, Seebaran A, Payne M, Reid AH, Protheroe AS, Vasist LS, Williams DD, Bowen C, Kathman SJ, Hodge JP, Dar MM, de Bono JS, Middleton MR. Br J Cancer. 2008;98:894. doi: 10.1038/sj.bjc.6604264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kathman SJ, Williams DH, Hodge JP, Dar M. Cancer Chemother Pharmacol. 2009;63:469. doi: 10.1007/s00280-008-0760-4. [DOI] [PubMed] [Google Scholar]
  • 37.Knox JJ, Gill S, Synold TW, Biagi JJ, Major P, Feld R, Cripps C, Wainman N, Eisenhauer E, Seymour L. Invest New Drugs. 2008;26:265. doi: 10.1007/s10637-007-9103-2. [DOI] [PubMed] [Google Scholar]
  • 38.Tang PA, Siu LL, Chen EX, Hotte SJ, Chia S, Schwarz JK, Pond GR, Johnson C, Colevas AD, Synold TW, Vasist LS, Winquist E. Invest New Drugs. 2008;26:257. doi: 10.1007/s10637-007-9098-8. [DOI] [PubMed] [Google Scholar]
  • 39.Debonis S, Skoufias DA, Indorato RL, Liger F, Marquet B, Laggner C, Joseph B, Kozielski F. J Med Chem. 2008;51:1115. doi: 10.1021/jm070606z. [DOI] [PubMed] [Google Scholar]
  • 40.Ogo N, Oishi S, Matsuno K, Sawada J, Fujii N, Asai A. Bioorg Med Chem Lett. 2007;17:3921. doi: 10.1016/j.bmcl.2007.04.101. [DOI] [PubMed] [Google Scholar]
  • 41.Kaan HY, Weiss J, Menger D, Ulaganathan V, Tkocz K, Laggner C, Popowycz F, Joseph B, Kozielski F. J Med Chem. 2011;54:1576. doi: 10.1021/jm100991m. [DOI] [PubMed] [Google Scholar]
  • 42.Hapuarachchige S, Montano G, Ramesh C, Rodriguez D, Henson LH, Williams CC, Kadavakkollu S, Johnson DL, Shuster CB, Arterburn JB. J Am Chem Soc. 2011;133:6780. doi: 10.1021/ja2005175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Riddoch RW, Schaffer P, Valliant JF. Bioconjug Chem. 2006;17:226. doi: 10.1021/bc050216w. [DOI] [PubMed] [Google Scholar]
  • 44.Funk CJ, Davis AS, Hopkins JA, Middleton KM. Anal Biochem. 2004;329:68. doi: 10.1016/j.ab.2004.02.032. [DOI] [PubMed] [Google Scholar]
  • 45.Cochran JC, Gatial JE, 3rd, Kapoor TM, Gilbert SP. J Biol Chem. 2005;280:12658. doi: 10.1074/jbc.M413140200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kapoor TM, Mayer TU, Coughlin ML, Mitchison TJ. J Cell Biol. 2000;150:975. doi: 10.1083/jcb.150.5.975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Carol H, Lock R, Houghton PJ, Morton CL, Kolb EA, Gorlick R, Reynolds CP, Maris JM, Keir ST, Billups CA, Smith MA. Pediatr Blood Cancer. 2009;53:1255. doi: 10.1002/pbc.22056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Palchaudhuri R, Nesterenko V, Hergenrother PJ. J Am Chem Soc. 2008;130:10274. doi: 10.1021/ja8020999. [DOI] [PubMed] [Google Scholar]
  • 49.Casanova E, Hernandez AI, Priego EM, Liekens S, Camarasa MJ, Balzarini J, Perez-Perez MJ. J Med Chem. 2006;49:5562. doi: 10.1021/jm0605379. [DOI] [PubMed] [Google Scholar]
  • 50.Al-Qawasmeh RA, Lee Y, Cao MY, Gu X, Vassilakos A, Wright JA, Young A. Bioorg Med Chem Lett. 2004;14:347. doi: 10.1016/j.bmcl.2003.11.004. [DOI] [PubMed] [Google Scholar]
  • 51.Benzaquen LR, Brugnara C, Byers HR, Gatton-Celli S, Halperin JA. Nat Med. 1995;1:534. doi: 10.1038/nm0695-534. [DOI] [PubMed] [Google Scholar]
  • 52.Khalid MH, Tokunaga Y, Caputy AJ, Walters E. J Neurosurg. 2005;103:79. doi: 10.3171/jns.2005.103.1.0079. [DOI] [PubMed] [Google Scholar]
  • 53.Snajdrova L, Xu A, Narayanan N. J Biol Chem. 1998;273:28032. doi: 10.1074/jbc.273.43.28032. [DOI] [PubMed] [Google Scholar]
  • 54.Dothager RS, Putt KS, Allen BJ, Leslie BJ, Nesterenko V, Hergenrother PJ. J Am Chem Soc. 2005;127:8686. doi: 10.1021/ja042913p. [DOI] [PubMed] [Google Scholar]
  • 55.Palchaudhuri R, Hergenrother PJ. Bioorg Med Chem Lett. 2008;18:5888. doi: 10.1016/j.bmcl.2008.07.128. [DOI] [PubMed] [Google Scholar]
  • 56.Benaka Prasad S, Vinaya K, Ananda Kumar C, Swarup S, Rangappa K. Medicinal Chemistry Research. 2010;19:220. [Google Scholar]
  • 57.Shi J, Orth JD, Mitchison T. Cancer Res. 2008;68:3269. doi: 10.1158/0008-5472.CAN-07-6699. [DOI] [PubMed] [Google Scholar]
  • 58.Kantarjian HM, Padmanabhan S, Stock W, Tallman MS, Curt GA, Li J, Osmukhina A, Wu K, Huszar D, Borthukar G, Faderl S, Garcia-Manero G, Kadia T, Sankhala K, Odenike O, Altman JK, Minden M. Invest New Drugs. 2011 doi: 10.1007/s10637-011-9660-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Shimizu M, Ishii H, Ogo N, Unno Y, Matsuno K, Sawada J, Akiyama Y, Asai A. Cancer Lett. 298:99. doi: 10.1016/j.canlet.2010.06.007. [DOI] [PubMed] [Google Scholar]
  • 60.Tunquist BJ, Woessner RD, Walker DH. Mol Cancer Ther. 9:2046. doi: 10.1158/1535-7163.MCT-10-0033. [DOI] [PubMed] [Google Scholar]
  • 61.Carter BZ, Mak DH, Woessner R, Gross S, Schober WD, Estrov Z, Kantarjian H, Andreeff M. Leukemia. 2009;23:1755. doi: 10.1038/leu.2009.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Peters NT, Kropf DL. BMC Plant Biol. 2006;6:19. doi: 10.1186/1471-2229-6-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.George O, Bryant BK, Chinnasamy R, Corona C, Arterburn JB, Shuster CB. ACS Chem Biol. 2008;3:167. doi: 10.1021/cb700210u. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES