The mitotic spindle checkpoint represents a signal transduction pathway that prevents the onset of anaphase until all chromosomes are properly aligned on a metaphase plate. Partial inactivation of this checkpoint allows premature separation of sister chromatids and results in aneuploidy, which might contribute to tumorigenesis. Unlike other cell cycle checkpoints, the spindle checkpoint is essential for cell viability, giving rise to the idea that the spindle checkpoint itself might represent a valuable target for anticancer therapy. We used a cell-based screen and identified the indolocarbazole compound Gö6976 as a pharmacologic inhibitor of the spindle checkpoint. Gö6976 potently overrides a spindle checkpoint–mediated mitotic arrest by abrogating the phosphorylation and kinetochore localization of several spindle checkpoint proteins. We identified the Aurora-A and Aurora-B kinases, which have been previously implicated in proper mitotic progression and spindle checkpoint function, as targets for Gö6976. Accordingly, Gö6976 treatment causes severe mitotic abnormalities and chromosome alignment defects, which are not properly detected by the drug-inactivated spindle checkpoint. This results in an aberrant progression of mitosis, leading to apoptosis in various human cancer cell lines, including spindle checkpoint–compromised cancer cells. Thus, our work describes a novel and promising strategy for anticancer treatment that targets the mitotic spindle checkpoint. [Cancer Res 2009;69(9):3874–83]

Antimitotic drugs, including various taxanes and Vinca alkaloids, are frequently used for anticancer treatment (1). It is well established that these drugs activate the spindle assembly checkpoint, a signal transduction pathway that monitors the proper alignment of chromosomes during mitosis (2, 3). The drug-induced and spindle checkpoint–mediated mitotic arrest is followed by the induction of apoptosis, which occurs either in mitosis or in the postmitotic G1 phase on escape from a prolonged mitotic arrest (4). The latter decision might depend on two separate pathways, which control the activation of caspases and a slow but continuous degradation of cyclin B during mitosis, respectively (5, 6). The drug-induced activation of caspases during mitosis might be a key event required for efficient tumor cell killing (6, 7), but the mechanisms leading to caspase activation remain elusive. Interestingly, the mitotic spindle checkpoint is required for the efficient induction of apoptosis on antimitotic drug treatment, regardless of whether a cell dies in mitosis or in the subsequent G1 phase. Thus, compromising the mitotic spindle checkpoint results in a significant decrease in sensitivity of tumor cells toward antimitotic drugs (812). These findings were recently supported by an unbiased small interfering RNA (siRNA) screen that identified spindle checkpoint proteins as major determinants for the sensitivity to paclitaxel (13). The molecular basis for a requirement of the spindle checkpoint in the activation of the apoptotic machinery is unclear, but it might be related to inhibition of Akt during a mitotic arrest (13) or to a dephosphorylation and activation of caspase-9 that requires a prolonged duration of mitosis (14).

The spindle checkpoint pathway requires the function of various proteins, including the kinases Bub1, BubR1, Mps1, Mad1, Mad2, Bub3, and several others, which are recruited to kinetochores on activation of the checkpoint (2, 3). Furthermore, inhibition of the centromeric Aurora-B kinase or overexpression of the centrosomal Aurora-A kinase led to spindle checkpoint malfunction (1517). Activation of the spindle checkpoint in response to misaligned chromosomes either in the early phases of a regular mitosis or chronically on antimitotic drug treatment causes the inhibition of the ubiquitin ligase activity within the anaphase-promoting complex/cyclosome (APC/C), which is required for the degradation of various mitotic substrates, including securin and cyclin B. Accordingly, failure of the spindle checkpoint results in a premature separation of sister chromatids and an unscheduled exit from mitosis, which leads to the generation of aneuploid progenitors (2, 3).

In human cancer, a partial dysfunction of the spindle checkpoint (15, 1822) is a frequent event and might contribute to tumorigenesis (18, 2328). In fact, mice heterozygous and hypomorphic for spindle checkpoint genes show gross aneuploidy and carcinogen-induced or spontaneous tumorigenesis (20, 29). Importantly, the analyses of homozygous knockout mice as well as siRNA studies in human cells revealed that the spindle checkpoint is essential for cell viability (3033). The essential nature of the spindle checkpoint raises the interesting idea to use the spindle checkpoint as a novel target for anticancer therapy.

To test this concept, we performed a cell-based screen and identified the indolocarbazole Gö6976 that inhibits the mitotic spindle checkpoint by restraining the phosphorylation and kinetochore localization of several spindle checkpoint proteins. Interestingly, we identified the mitotic Aurora-A and Aurora-B kinases as targets for Gö6976 in vitro and in vivo and show that the pharmacologic inactivation of the spindle checkpoint efficiently kills human cancer cells.

Cell culture and treatments. All cell lines were maintained under standard culture conditions (11) and treated with 300 nmol/L nocodazole (Sigma), 100 nmol/L Taxol (Sigma), or 68 μmol/L monastrol (Biomol), 200 μmol/L ALLN (Calbiochem), 20 μmol/L MG132 (Calbiochem), 5 to 100 μmol/L roscovitine (Sigma), 0.2 to 20 μmol/L Gö6976 (Calbiochem), 0.05 to 2 μmol/L ZM447439 (Tocris), 50 μmol/L H89 (Calbiochem), 100 μmol/L protein kinase A (PKA) inhibitor (Calbiochem), 1 μmol/L Ro31-8220 (Calbiochem), 1 μmol/L Ro32-0432 (Calbiochem), 40 μmol/L rottlerin (Calbiochem), 50 μmol/L PD98059 (Calbiochem), 10 μmol/L U0126 (Calbiochem), 30 μmol/L SB202190 (Calbiochem), 30 μmol/L SB203580 (Calbiochem), 50 nmol/L staurosporine (Sigma), 20 μmol/L LY294002 (Sigma), 0.1 to 2 μmol/L UCN-01 (a gift from the Developmental Therapeutics Program of the National Cancer Institute), and 10 μmol/L Raf inhibitor (BAY 43-9006). Cells were synchronized in G1-S by a double-thymidine block using 2 mmol/L thymidine (Sigma).

Flow cytometry and determination of the mitotic index. The DNA content and the mitotic index were determined as described (11).

Western blotting. Cell lysates were prepared, and SDS-PAGE, semidry Western blotting, and detection were performed as described (11) using the following antibodies: Bub1 (Bethyl; Stephen Taylor, University of Manchester, Manchester, United Kingdom), BubR1 (Chemicon; Stephen Taylor), Bub3 (BD Biosciences), Mps1 (Santa Cruz Biotechnology), Plk1 (Santa Cruz Biotechnology), Aurora-A (Santa Cruz Biotechnology; pT288: Cell Signaling), Aurora-B (BD Biosciences; pT232: Cell Signaling), cyclin B (Santa Cruz Biotechnology), tubulin (Sigma), actin (Sigma), securin (NeoMarkers), histone H3 (pS-10; Cell Signaling), and poly(ADP-ribose) polymerase (PARP; Pharmingen). Signals were quantitated using the ImageJ software (NIH).

Chromosome spread analysis. Premature sister chromatid separation and karyotype analysis were performed as described (11).

Microscopy. Cells were fixed/permeabilized in 2% paraformaldehyde/methanol and incubation with antibodies was carried out for 2 h. Images were taken with a Leica DM6000B microscope and a charge-coupled device camera (Orca-ER, Hamamatsu) with a Z-optical spacing of 0.2 μm. Images were deconvolved using the Leica LAS-AF software and maximum projection images are shown. Pixel quantitations of CREST and Bub1/BubR1 signals on kinetochores were performed using the Leica LAS-AF software.

Kinase assays.In vitro kinase assays using recombinant Aurora kinases (34) were performed for 30 min at 30°C in kinase buffer [50 mmol/L Tris-HCl (pH 7.7), 100 mmol/L KCl, 5 mmol/L MgCl2, 1 mmol/L DTT] in the presence of 50 to 2,000 μmol/L of ATP (Sigma), 1 μCi [32P]ATP (6,000 Ci/mmol; Amersham), and 7 μg MBP (Sigma). Substrate phosphorylation was visualized by autoradiography and quantitated using a phosphoimager (Fuji). For immunoprecipitation kinase assays, Aurora-A or Aurora-B was immunoprecipitated from lysates and subjected to in vitro kinase assays.

Apoptosis assays. Caspase activity assays, detection of PARP cleavage, and chromosomal DNA fragmentation assays as well as determination of the sub-G1 DNA content were performed as described previously (11). For caspase inhibition, the pan-caspase inhibitor Boc-D-FMK (100 μmol/L; Calbiochem) was used.

Identification of the indolocarbazole compound Gö6976 as a potent abrogator of a spindle damage–induced mitotic arrest. We performed a cell-based screen, in which HCT116 cells were first treated with nocodazole to arrest cells in mitosis in a spindle checkpoint–dependent manner. Subsequently, cells were treated with small-molecule inhibitors followed by determination of the mitotic index to monitor the mitotic arrest (Fig. 1A). Because several kinases are involved in spindle checkpoint signaling, we screened commercially available protein kinase 1inhibitors, including established inhibitors for PKA, protein kinase C (PKC), cyclin-dependent kinase (CDK), Chk1, mitogen-activated protein/extracellular signal-regulated kinase kinase, p38, Raf, and phosphatidylinositol 3-kinase (Fig. 1B). Among the small-molecule inhibitors tested, we identified the indolocarbazole Gö6976 (Supplementary Fig. S1A) as a compound that potently abrogated the induced mitotic arrest and forced cells to exit mitosis with a 4N DNA content (Fig. 1B and C). In HCT116 and HeLa cells, Gö6976 showed a half-maximal effect at 0.8 to 1.2 μmol/L in the presence of nocodazole and Taxol, which is ∼25 times more potent than the CDK1 inhibitor roscovitine in abrogating the mitotic arrest (Fig. 1D). Very similar results were obtained after treatment with the KSP/Eg5 inhibitor monastrol and in various human cell lines (Supplementary Fig. S1B and C). We also tested established structurally related and similarly potent PKC inhibitors (Ro31-8220 and Ro32-0432) as well as the structurally unrelated PKC inhibitor rottlerin and found no effect on the spindle damage–induced mitotic arrest, suggesting that Gö6976 acts independently of PKC inhibition. In addition, other indolocarbazole compounds, including UCN-01 or staurosporine, had no or little effects on the spindle damage–induced mitotic arrest (Fig. 1B). No significant inhibition of CDK1 by Gö6976 was observed (Supplementary Fig. S1D).

Figure 1.

Gö6976 overrides a spindle damage–induced mitotic arrest. A, a cell-based screen performed to identify pharmacologic spindle checkpoint inhibitors. B, Gö6976 is an inhibitor of spindle damage–induced mitotic arrest. HCT116 cells were arrested in mitosis with 300 nmol/L nocodazole for 14 h and subsequently treated with various protein kinase inhibitors. After 2 h of incubation, the mitotic index was determined. C, representative examples of fluorescence-activated cell sorting (FACS) profiles of HCT116 cells treated with nocodazole alone or followed by Gö6976 treatment as in B. D, dose-response curves for Gö6976 and roscovitine after treatment with nocodazole or Taxol according to B. Points, mean of at least three independent experiments; bars, SD.

Figure 1.

Gö6976 overrides a spindle damage–induced mitotic arrest. A, a cell-based screen performed to identify pharmacologic spindle checkpoint inhibitors. B, Gö6976 is an inhibitor of spindle damage–induced mitotic arrest. HCT116 cells were arrested in mitosis with 300 nmol/L nocodazole for 14 h and subsequently treated with various protein kinase inhibitors. After 2 h of incubation, the mitotic index was determined. C, representative examples of fluorescence-activated cell sorting (FACS) profiles of HCT116 cells treated with nocodazole alone or followed by Gö6976 treatment as in B. D, dose-response curves for Gö6976 and roscovitine after treatment with nocodazole or Taxol according to B. Points, mean of at least three independent experiments; bars, SD.

Close modal

Gö6976 inhibits the mitotic spindle checkpoint. In the presence of nocodazole or Taxol, treatment with 2 μmol/L Gö6976 induced a rapid exit from mitosis, which was associated with the rapid degradation of cyclin B and securin that could be blocked by the proteasome inhibitor ALLN, indicating that Gö6976 relieves the inhibition of the APC/C (Fig. 2A). Gö6976 allowed the entry into mitosis but suppressed the accumulation of mitotic cells in the presence of nocodazole, whereas roscovitine treatment prevented the entry into mitosis (Supplementary Fig. S2A). Furthermore, premature exit from mitosis led to endoreduplication (Supplementary Fig. S2B), which is associated with spindle checkpoint impairment as shown previously (35). Gö6976 treatment forced cells out of mitosis and a single nucleus with a 4N DNA content was reformed, whereas CDK inhibitors caused a high rate of multinucleation (Supplementary Fig. S2C), supporting the notion that Gö6976 acts independently from CDK1.

Figure 2.

Gö6976 is an inhibitor of the mitotic spindle checkpoint. A, Gö6976 induces mitotic exit and proteasome-dependent proteolysis of cyclin B and securin. HCT116 cells were treated with nocodazole (N) or Taxol (T) and incubated with DMSO (D) or 2 μmol/L Gö6976 (G) in the presence or absence of 200 μmol/L ALLN (A). The mitotic index was determined (left) and the protein levels of cyclin B and securin were determined on Western blot (right). B, Gö6976 induces premature sister chromatid separation. Nocodazole-arrested HCT116 cells were treated with DMSO or Gö6976 for 10 and 15 min and metaphase spreads were prepared. Examples of typical metaphase spreads and the quantitation of cells showing premature sister chromatid separations (n = 400) are shown. C, Gö6976 reduces kinetochore localization of Bub1 and BubR1. HCT116 cells were treated with nocodazole to arrest cells in mitosis followed by incubation with 20 μmol/L MG132 and 2 μmol/L Gö6976 for 2 h. Cells were fixed and immunofluorescence staining was performed for Bub1, BubR1, and CREST and chromosomes were stained with Hoechst dye. Pixel intensities for Bub1 and BubR1 at kinetochores were quantified relative to the CREST signals. The graphs represent quantitations from 80 kinetochores from six cells. D, Gö6976 inhibits phosphorylation of Bub1, Mps1, Aurora-A, and Aurora-B. Cells were treated as in C in the presence or absence of ALLN and the indicated mitotic proteins were detected on Western blots. For each cell population, the mitotic index was determined.

Figure 2.

Gö6976 is an inhibitor of the mitotic spindle checkpoint. A, Gö6976 induces mitotic exit and proteasome-dependent proteolysis of cyclin B and securin. HCT116 cells were treated with nocodazole (N) or Taxol (T) and incubated with DMSO (D) or 2 μmol/L Gö6976 (G) in the presence or absence of 200 μmol/L ALLN (A). The mitotic index was determined (left) and the protein levels of cyclin B and securin were determined on Western blot (right). B, Gö6976 induces premature sister chromatid separation. Nocodazole-arrested HCT116 cells were treated with DMSO or Gö6976 for 10 and 15 min and metaphase spreads were prepared. Examples of typical metaphase spreads and the quantitation of cells showing premature sister chromatid separations (n = 400) are shown. C, Gö6976 reduces kinetochore localization of Bub1 and BubR1. HCT116 cells were treated with nocodazole to arrest cells in mitosis followed by incubation with 20 μmol/L MG132 and 2 μmol/L Gö6976 for 2 h. Cells were fixed and immunofluorescence staining was performed for Bub1, BubR1, and CREST and chromosomes were stained with Hoechst dye. Pixel intensities for Bub1 and BubR1 at kinetochores were quantified relative to the CREST signals. The graphs represent quantitations from 80 kinetochores from six cells. D, Gö6976 inhibits phosphorylation of Bub1, Mps1, Aurora-A, and Aurora-B. Cells were treated as in C in the presence or absence of ALLN and the indicated mitotic proteins were detected on Western blots. For each cell population, the mitotic index was determined.

Close modal

Consistent with a spindle checkpoint inhibition by Gö6976, we found a high rate of premature chromatid separation on Gö6976 treatment (Fig. 2B). Prolonged treatment led to a significant increase in aneuploid cells, whereas the overall cell cycle progression was not significantly disturbed (Supplementary Fig. S2D and E), although dead cells became increasingly apparent.

Gö6976 inhibits the proper phosphorylation and localization of spindle checkpoint proteins. We investigated whether Gö6976 interferes with the localization or amount and phosphorylation of spindle checkpoint proteins. Significantly, Gö6976 treatment reduced the kinetochore localization of Bub1 and BubR1 by ∼80% (Fig. 2C). Aurora-B was also found to be mislocalized from the centromere region to chromosome arms on Gö6976 treatment (Supplementary Fig. S3), whereas Aurora-A retained its centrosomal localization (data not shown). Furthermore, on Western blots, we found that the hyperphosphorylation of Bub1, Mps1, Aurora-A, and Aurora-B, which is closely associated with spindle checkpoint activation, was abolished on Gö6976 treatment both on forced exit from mitosis and in mitotic cells (Fig. 2D).

Gö6976 causes aberrant spindle morphology and chromosome alignment defects. Next, we investigated the mitotic progression in the presence of Gö6976. Treatment of asynchronously growing HCT116 cells with 2 μmol/L Gö6976 resulted in a transient accumulation of mitotic cells (Fig. 3A) and this was associated with profound alterations in the spindle structure and severely misaligned chromosomes (Fig. 3B). The spindles often seemed cylindrical and flattened and groups of chromosomes were frequently found near the spindle poles, suggesting that those chromosomes are not properly attached to microtubules. Similar chromosome alignment defects were observed when cells were released from a monastrol block into Gö6976 (Supplementary Fig. S4A).

Figure 3.

Gö6976 causes chromosome alignment defects and premature and accelerated exit from mitosis. A, asynchronously growing HCT116 cells were treated with 2 μmol/L Gö6976 for the indicated times and the DNA content and the mitotic index were determined. B, cells were treated with 2 μmol/L Gö6976 for 6 h and fixed cells were stained for α-tubulin, CREST, and DNA. Typical examples of maximal projections of deconvolved Z-stacks are shown. C, HCT116 cells were synchronized in mitosis by a nocodazole block and released into medium with or without 2 μmol/L Gö6976. At the indicated time points, cells were fixed and stained for α-tubulin and DNA. D, cells were treated as in C and the DNA content and the mitotic index were determined. Representative FACS profiles are shown. Points, mean of at least three independent experiments; bars, SD.

Figure 3.

Gö6976 causes chromosome alignment defects and premature and accelerated exit from mitosis. A, asynchronously growing HCT116 cells were treated with 2 μmol/L Gö6976 for the indicated times and the DNA content and the mitotic index were determined. B, cells were treated with 2 μmol/L Gö6976 for 6 h and fixed cells were stained for α-tubulin, CREST, and DNA. Typical examples of maximal projections of deconvolved Z-stacks are shown. C, HCT116 cells were synchronized in mitosis by a nocodazole block and released into medium with or without 2 μmol/L Gö6976. At the indicated time points, cells were fixed and stained for α-tubulin and DNA. D, cells were treated as in C and the DNA content and the mitotic index were determined. Representative FACS profiles are shown. Points, mean of at least three independent experiments; bars, SD.

Close modal

Remarkably, the severe alignment defects induced by Gö6976 did not cause a robust mitotic arrest as would be expected in the presence of a functional spindle checkpoint. Instead, the accumulation of mitotic cells was only weak and transient (Fig. 3A) but dependent on the spindle checkpoint (Supplementary Fig. S4B), suggesting that the mitotic arrest cannot be maintained, possibly mediated by Gö6976-induced spindle checkpoint inhibition.

Gö6976 accelerates exit from an aberrant mitosis. To test whether Gö6976 treatment indeed allows an exit from mitosis in the presence of unaligned chromosomes, we released cells from a nocodazole block in the presence or absence of Gö6976 and followed the progression through mitosis qualitatively by immunofluorescence (Fig. 3C) and quantitatively by monitoring the mitotic index (Fig. 3D). Whereas control cells progressed normally through mitosis and exited mitosis properly, Gö6976-treated cells exhibited a significant acceleration of mitotic progression and exited from mitosis in the presence of misaligned chromosomes. Most Gö6976-treated cells underwent cytokinesis despite the presence of unaligned and missegregated chromosomes, producing daughter cells with highly unequal DNA content (Fig. 3D,, top). By the end of mitosis, apoptotic cells became readily apparent, suggesting that the severe missegregation observed causes cell death (Fig. 3C).

The mitotic Aurora-A and Aurora-B kinases are inhibited by Gö6976 in vitro and in vivo. To gain insights into the mitotic target kinase of Gö6976, we evaluated candidate mitotic kinases known to be involved in spindle checkpoint function. Because we observed a lack of phosphorylation of Bub1, Mps1, Aurora-A, and Aurora-B (Fig. 2D), which might relate to their functional inactivation, we evaluated these kinases in particular. In vitro, no inhibition of Bub1, Mps1, Plk1, and p38 by Gö6976 was found. However, Aurora-B kinase activity was significantly inhibited by Gö6976 in these assays (Fig. 4A). We determined the IC50 values for Gö6976 and ZM447439, which is a potent and well-characterized inhibitor for Aurora-A and Aurora-B kinases in vitro (36), and found that Gö6976 inhibited Aurora-B with similar potency as ZM447439 (100 versus 80 nmol/L at 50 μmol/L ATP; Fig. 4B). However, our analyses revealed that Gö6976 was nearly five times more potent than ZM447439 in inhibiting the Aurora-A kinase (120 versus 580 nmol/L at 50 μmol/L ATP; Fig. 4B). As expected for an ATP-competitive inhibitor, the inhibitory activity of Gö6976 decreased with higher ATP concentrations (Supplementary Fig. S5A; Supplementary Text). To test whether Gö6976 also inhibits the Aurora kinases in vivo, we evaluated the activation loop–specific phosphorylation of Aurora-A and Aurora-B kinases (pT288 for Aurora-A and pT232 for Aurora-B), which is indicative of their activation. In agreement with previous data (36), treatment of mitotic cells with ZM447439 abolished selectively the activation of cellular Aurora-B (75% inhibition) but left the activity of Aurora-A unchanged. However, in agreement with our in vitro data, Gö6976 treatment inhibited the activity of both Aurora-A and Aurora-B, with similar potency in vivo (60% and 58% inhibition, respectively; Fig. 4C; Supplementary Fig. S5B).

Figure 4.

Gö6976 inhibits the Aurora-A and Aurora-B kinases in vitro and in vivo. A, in vitro kinase assays using the indicated purified recombinant kinases in the presence of different concentrations of Gö6976. Representative autoradiographs are shown. B, the in vitro activity of purified recombinant Aurora-A and Aurora-B was determined in the presence of different concentrations of Gö6976 or ZM447439 and the IC50 values were calculated in the presence of 50 μmol/L ATP. C, detection of phosphorylated Aurora-A and Aurora-B kinases on Western blots using lysates from mitotic HCT116 cells treated with nocodazole for 14 h followed by incubation with MG132 in the presence or absence of 2 μmol/L Gö6976 or 2 μmol/L ZM447439 for 2 h. D, Aurora-A and Aurora-B kinases were immunopurified from mitotic HeLa cells treated with nocodazole, ALLN, and 1 or 2 μmol/L of Gö6976 and their kinase activities were determined. Columns, mean of at least three independent experiments; bars, SD.

Figure 4.

Gö6976 inhibits the Aurora-A and Aurora-B kinases in vitro and in vivo. A, in vitro kinase assays using the indicated purified recombinant kinases in the presence of different concentrations of Gö6976. Representative autoradiographs are shown. B, the in vitro activity of purified recombinant Aurora-A and Aurora-B was determined in the presence of different concentrations of Gö6976 or ZM447439 and the IC50 values were calculated in the presence of 50 μmol/L ATP. C, detection of phosphorylated Aurora-A and Aurora-B kinases on Western blots using lysates from mitotic HCT116 cells treated with nocodazole for 14 h followed by incubation with MG132 in the presence or absence of 2 μmol/L Gö6976 or 2 μmol/L ZM447439 for 2 h. D, Aurora-A and Aurora-B kinases were immunopurified from mitotic HeLa cells treated with nocodazole, ALLN, and 1 or 2 μmol/L of Gö6976 and their kinase activities were determined. Columns, mean of at least three independent experiments; bars, SD.

Close modal

We also determined the activity of Aurora-A and Aurora-B immunoprecipitated from drug-treated cells. Again, Gö6976 treatment caused significant inhibition of Aurora-A and Aurora-B (Fig. 4D), whereas immunoprecipitated CDK1-cyclin B complexes were not inhibited by Gö6976 (Supplementary Fig. S5C). These results provide strong evidence that Gö6976 is a potent inhibitor of Aurora-A and Aurora-B in vitro and in vivo, although we cannot exclude the possibility that Gö6976 targets additional kinases during mitosis.

In addition to its crucial function in chromosome alignment and spindle checkpoint function, Aurora-B is also required for cytokinesis (16). Indeed, treatment of asynchronously growing cells with ZM447439 prevents cytokinesis and results in profound polyploidization. Surprisingly, although Gö6976 inhibits Aurora-B in vivo, neither profound defects in cytokinesis nor polyploidization was detected (Supplementary Fig. S6).

The aberrant progression through mitosis induced by Gö6976 causes apoptosis in tumor cells. To test whether the Gö6976-induced aberrant progression of mitosis causes apoptosis, we synchronized HCT116 cells at G1-S, treated the cells with Gö6976 before entry into mitosis, and evaluated the activation of caspase-3 in intact cells. Indeed, Gö6976 treatment induced caspase-3 activity, PARP cleavage, and DNA fragmentation after progression through a single mitosis and this was significantly suppressed when entry into mitosis was prevented by inhibiting CDK1 (Fig. 5A; Supplementary Fig. S7A). Further, Gö6976-induced apoptosis was caspase dependent (Fig. 5B).

Figure 5.

The aberrant progression through mitosis induced by Gö6976 causes apoptosis in human cancer cells. A, Gö6976-induced apoptosis requires progression through mitosis. HCT116 cells were synchronized at the G1-S transition by a double-thymidine block and released into fresh medium. Seven hours after release, cells were treated with various concentrations of Gö6976, roscovitine, or roscovitine plus Gö6976, and another 7 h later, caspase-3 activity was determined. B, Gö6976-induced apoptosis is dependent on caspase activity. HCT116 cells were treated with 20 μmol/L Gö6976 in the presence or absence of 100 μmol/L Boc-D-FMK for 24 and 48 h and the DNA content and proportion of apoptotic cells were determined. Representative FACS profiles are shown. C, Gö6976 induces caspase activity in various human cancer cell lines. The indicated human cancer cell lines and two nontransformed cell lines were treated with 15 μmol/L Gö6976 for 24 h and caspase-3 activity was measured in intact cells. D, synergistic effect of Taxol and Gö6976. HCT116 cells were treated with 100 nmol/L Taxol for 14 h followed by the addition of DMSO or 2 μmol/L Gö6976 for additional 2 h. The DNA content was determined by FACS analyses and representative DNA profiles and quantitation of apoptotic cells are shown. E, quantitative analysis of cell death on combined treatment with 2 μmol/L Gö6976 and 100 nmol/L Taxol for 38 h. Columns, mean of at least three independent experiments; bars, SD.

Figure 5.

The aberrant progression through mitosis induced by Gö6976 causes apoptosis in human cancer cells. A, Gö6976-induced apoptosis requires progression through mitosis. HCT116 cells were synchronized at the G1-S transition by a double-thymidine block and released into fresh medium. Seven hours after release, cells were treated with various concentrations of Gö6976, roscovitine, or roscovitine plus Gö6976, and another 7 h later, caspase-3 activity was determined. B, Gö6976-induced apoptosis is dependent on caspase activity. HCT116 cells were treated with 20 μmol/L Gö6976 in the presence or absence of 100 μmol/L Boc-D-FMK for 24 and 48 h and the DNA content and proportion of apoptotic cells were determined. Representative FACS profiles are shown. C, Gö6976 induces caspase activity in various human cancer cell lines. The indicated human cancer cell lines and two nontransformed cell lines were treated with 15 μmol/L Gö6976 for 24 h and caspase-3 activity was measured in intact cells. D, synergistic effect of Taxol and Gö6976. HCT116 cells were treated with 100 nmol/L Taxol for 14 h followed by the addition of DMSO or 2 μmol/L Gö6976 for additional 2 h. The DNA content was determined by FACS analyses and representative DNA profiles and quantitation of apoptotic cells are shown. E, quantitative analysis of cell death on combined treatment with 2 μmol/L Gö6976 and 100 nmol/L Taxol for 38 h. Columns, mean of at least three independent experiments; bars, SD.

Close modal

We treated 15 human cell lines with Gö6976 and found a significant but varying induction of caspase activity in most cancer cell lines (Fig. 5C), whereas nontransformed human cells (human BJ-hTert fibroblasts and primary human umbilical vein endothelial cells) exhibited low caspase-3 activity. The reason for the different sensitivities observed is not known at present but could be recapitulated in colony formation assays comparing the sensitivity of HeLa and BJ-hTert fibroblasts toward Gö6976 (Supplementary Fig. S7B; Supplementary Text). Accordingly, we found that HeLa and HCT116 cancer cells showed a higher rate of cell killing than BJ-hTert fibroblasts after treatment with Gö6976 (Supplementary Fig. S7C and D). We also compared the efficacy of Gö6976 with ZM447439 and Taxol and found a higher efficacy of Taxol but a lower efficacy of ZM447439 toward HeLa and HCT116 cells (Supplementary Fig. S7D and E).

Gö6976 acts synergistically with Taxol. Recent data showed that the efficacy of antimitotic drugs could be greatly enhanced by subsequent inactivation of the spindle checkpoint (7, 37). To investigate whether Taxol and a Gö6976-mediated inactivation of the spindle checkpoint act synergistically, cells were treated for 14 hours with Taxol to arrest cells in mitosis followed by treatment with Gö6976 to induce exit from mitosis. In fact, a synergistic effect of Gö6976 and Taxol was clearly observed (Fig. 5D and E). It is important to note that a reversed order of drug treatment caused an inhibition of the spindle checkpoint before Taxol treatment and reduced sensitivity toward Taxol (data not shown; Supplementary Fig. S2B; refs. 11, 35, 38).

A spindle checkpoint override is required for efficacy of Gö6976. To investigate if the efficacy of Gö6976 requires the override of the mitotic spindle checkpoint, we took advantage of a human colon carcinoma cell line, HT29, which exhibits a very strong and prolonged mitotic arrest in response to spindle damage compared with most cancer cell lines, including HCT116 cells (Supplementary Fig. S8). Accordingly, Gö6976 could only weakly override the spindle checkpoint in HT29 cells (Fig. 6A), which was directly associated with resistance toward Gö6976 (Fig. 6B), suggesting that the spindle checkpoint override is directly associated with the sensitivity toward Gö6976. It is important to note that HT29 cells are not resistant per se because they are capable of inducing apoptosis on chemotherapeutic treatment (data not shown).

Figure 6.

The role of the spindle checkpoint for the efficacy of Gö6976. A, a spindle damage–induced mitotic arrest is not overridden by Gö6976 in HT29 cells. HCT116 and HT29 cells were arrested in mitosis by treatment with 150 nmol/L nocodazole for 16 h followed by the addition of different concentrations of Gö6976. After 2 h of incubation, the mitotic index was determined. B, reduced caspase activation in HT29 cells on Gö6976 treatment. HCT116 and HT29 cells were synchronized at G1-S and released into the cell cycle. Seven hours after release, cells were treated with various concentrations of Gö6976 and the activity of caspase-3 was determined after 7 h of treatment. C, spindle checkpoint–compromised cancer cells show reduced caspase activation on spindle damage. HCT116 and isogenic derivatives expressing lower levels of Mad2 (HCT116-MAD2+/−) or Mad1 (HCT-MAD1-kd) were treated with 150 nmol/L nocodazole for 38 h and caspase-3 activity was determined in intact cells. D, Gö6976 reduces the already lowered mitotic arrest in spindle checkpoint–compromised cancer cells on spindle damage. Spindle checkpoint–compromised and parental HCT116 cells were treated with 150 nmol/L nocodazole in the presence or absence of 2 μmol/L Gö6976 and the mitotic index was determined at various time points. E, activation of caspase-3 in spindle checkpoint–compromised cancer cells in response to Gö6976 treatment. The indicated HCT116 cell lines were treated with different Gö6976 concentrations and the induction of caspase-3 activity was determined. Columns, mean of at least three independent experiments; bars, SD.

Figure 6.

The role of the spindle checkpoint for the efficacy of Gö6976. A, a spindle damage–induced mitotic arrest is not overridden by Gö6976 in HT29 cells. HCT116 and HT29 cells were arrested in mitosis by treatment with 150 nmol/L nocodazole for 16 h followed by the addition of different concentrations of Gö6976. After 2 h of incubation, the mitotic index was determined. B, reduced caspase activation in HT29 cells on Gö6976 treatment. HCT116 and HT29 cells were synchronized at G1-S and released into the cell cycle. Seven hours after release, cells were treated with various concentrations of Gö6976 and the activity of caspase-3 was determined after 7 h of treatment. C, spindle checkpoint–compromised cancer cells show reduced caspase activation on spindle damage. HCT116 and isogenic derivatives expressing lower levels of Mad2 (HCT116-MAD2+/−) or Mad1 (HCT-MAD1-kd) were treated with 150 nmol/L nocodazole for 38 h and caspase-3 activity was determined in intact cells. D, Gö6976 reduces the already lowered mitotic arrest in spindle checkpoint–compromised cancer cells on spindle damage. Spindle checkpoint–compromised and parental HCT116 cells were treated with 150 nmol/L nocodazole in the presence or absence of 2 μmol/L Gö6976 and the mitotic index was determined at various time points. E, activation of caspase-3 in spindle checkpoint–compromised cancer cells in response to Gö6976 treatment. The indicated HCT116 cell lines were treated with different Gö6976 concentrations and the induction of caspase-3 activity was determined. Columns, mean of at least three independent experiments; bars, SD.

Close modal

Targeting of spindle checkpoint–compromised cancer cells by Gö6976. Several reports have indicated that the spindle checkpoint is required for the efficient induction of cell death after treatment with antimitotic drugs (8, 9, 11, 12, 39). Accordingly, spindle checkpoint–compromised HCT116 cells with lowered expression of MAD1 or MAD2 showed a lowered mitotic arrest and reduced caspase-3 activity in response to nocodazole treatment (Fig. 6C; ref. 11). In both spindle checkpoint–impaired cell lines, Gö6976 treatment further lowered the mitotic arrest in response to spindle damage (Fig. 6D) and induced caspase-3 activity similar to parental cells (Fig. 6E), suggesting that spindle checkpoint–compromised cancer cells are sensitive toward Gö6976 treatment.

Due to its essential nature, the spindle checkpoint has been suggested as a potential novel anticancer drug target (32, 33). We wished to prove this conceptual idea and have identified the indolocarbazole Gö6976 as an inhibitor of the spindle checkpoint. Importantly, we show that this inhibitor induces a highly aberrant and accelerated mitosis that results in efficient killing of human cancer cells. Thus, we propose to add the spindle checkpoint to the growing list of attractive mitotic drug targets.

The “classic” mitotic drug target is the mitotic spindle, whose function is inhibited by microtubule-targeting drugs including various taxanes or Vinca alkaloids (40). However, due to the ability to inhibit microtubule function in interphase or even in differentiated cells, a plethora of unwanted side effects, including neutropenia and neuropathy, is observed after treatment with these drugs. Therefore, novel mitotic drug targets that function exclusively in mitosis, including the Plk1 and the Aurora kinases, but also mitotic Eg5/KSP or Cenp-E kinesins, have gained considerable interest (1, 41). Similarly, no function for the mitotic spindle checkpoint pathway outside of mitosis has been described thus far. Thus, inhibition of the spindle checkpoint is expected to have no effect on interphase or differentiated cells. However, it is important to note that targeting mitosis per se might harbor the risk of chromosome missegregation in surviving cells that could contribute to de novo tumorigenesis.

At present, is not clear how antimitotic drugs induce tumor cell death. Most recent results have indicated that cells activate caspase-dependent pathways during mitosis and die either in mitosis or on an unscheduled exit from mitosis (57). However, it is unknown how caspases are activated on drug treatment, but the activation of the spindle checkpoint associated with a transient mitotic delay seems to be necessary for the efficient induction of cell death (813). Interestingly, during mitosis, caspase-9 is inhibited by CDK1/cyclin B–mediated phosphorylation. Prolonged mitotic arrest, which requires the spindle checkpoint, may lead to a progressive dephosphorylation, activation of caspase-9, and the induction of apoptosis, thereby providing a timer mechanism monitoring the length of mitosis (14). In contrast to this mechanism, we show here when targeting the spindle checkpoint pathway that the induction of apoptosis is not dependent on a functional spindle checkpoint. Similarly, because Aurora-B is required for the spindle checkpoint, ZM447439 also induces spindle checkpoint–independent apoptosis.3

3

A. Stolz, C. Vogel, and H. Bastians, unpublished results.

Thus, the therapeutic concept of spindle checkpoint inhibition is expected to be efficient in tumor cells harboring a compromised spindle checkpoint.

We show that Gö6976 targets the mitotic Aurora kinases, which might explain the phenotypes observed after treatment with Gö6976. Gö6976 treatment causes spindle formation defects and severe chromosome misalignment, phenotypes attributable to Aurora-A inhibition (42). In addition, inhibition of Aurora-B might explain the checkpoint override in the presence of misaligned chromosomes because Aurora-B has been implicated in spindle checkpoint function (36, 43). Thus, the efficacy of Gö6976 might depend on a two-step mechanism: first, the induction of mitotic damage associated with a transient mitotic delay, and second, the override of the spindle checkpoint allowing exit from an aberrant mitosis. The latter might be particularly supportive for the induction of apoptosis. In fact, when tumor cells are first treated with Taxol followed by the inactivation of the spindle checkpoint, the induction of cell death is greatly enhanced (Fig. 5; refs. 7, 37). Interestingly, the reverse order of events, Taxol treatment of cells in which the spindle checkpoint is already inhibited, led to endoreduplication and a reduced sensitivity toward Taxol3 (13, 35, 38), suggesting that a therapeutic combination of Taxol and spindle checkpoint inhibitors strictly depends on the order of drug treatment.

Although we show that Gö6976 inhibits Aurora-B in vitro and in vivo, we found some remarkable differences to the inhibition by selective Aurora-B inhibitors, including ZM447439 or hesperadin (36, 43). Whereas ZM447439 and hesperadin inhibit selectively a Taxol-induced spindle checkpoint, Gö6976 overrides the spindle checkpoint on Taxol or nocodazole treatment. Moreover, in contrast to ZM447439 or hesperadin, Gö6976 did not cause defects in cytokinesis or induce polyploidization. At present, we cannot explain these interesting differences, but they might be related to the concomitant inhibition of Aurora-A by Gö6976 or due to different inhibitor selectivity for distinct Aurora-B–containing protein complexes.

Previous work has identified the c-Jun NH2-terminal kinase inhibitor SP600125 as an inhibitor of the spindle checkpoint kinase Mps1 (44). Remarkably, SP600125-mediated inhibition of Mps1 led to an inactivation of the spindle checkpoint in cancer cells but not in nontransformed fibroblasts. Although we have not investigated the spindle checkpoint activity in nontransformed cells, we found that Gö6976 killed many cancer cells more efficiently than nontransformed human cells. The molecular basis for this selectivity is not clear at present but deserves further detailed investigation in future studies.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

A. Stolz and C. Vogel contributed equally to this work.

Grant support: University Medical Center Giessen and Marburg and Deutsche Forschungsgemeinschaft Heisenberg fellowship (H. Bastians) and Behring-Röntgen-Stiftung.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Stephen Taylor, Paolo Sassone-Corsi, Bert Vogelstein, Robert Benezra, Qi Min Zhan, Stefan Dimitrov, Stefan Gaubatz, Martin Eilers, Matthias Dobbelstein, and Mathias Schmidt for providing antibodies, cell lines, and plasmids; Rolf Müller for support; Katja Scheffler for experimental help; the Developmental Therapeutics Program of the National Cancer Institute for the gift of UCN-01; Gary Gorbski for discussions; and Heike Krebber and all members of the Bastians lab for comments on the manuscript.

1
Schmidt M, Bastians H. Mitotic drug targets and the development of novel anti-mitotic anticancer drugs.
Drug Resist Updat
2007
;
10
:
162
–81.
2
Bharadwaj R, Yu H. The spindle checkpoint, aneuploidy, and cancer.
Oncogene
2004
;
23
:
2016
–27.
3
Musacchio A, Salmon ED. The spindle-assembly checkpoint in space and time.
Nat Rev Mol Cell Biol
2007
;
8
:
379
–93.
4
Rieder CL, Maiato H. Stuck in division or passing through: what happens when cells cannot satisfy the spindle assembly checkpoint.
Dev Cell
2004
;
7
:
637
–51.
5
Brito DA, Rieder CL. Mitotic checkpoint slippage in humans occurs via cyclin B destruction in the presence of an active checkpoint.
Curr Biol
2006
;
16
:
1194
–200.
6
Gascoigne KE, Taylor SS. Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs.
Cancer Cell
2008
;
14
:
111
–22.
7
Kim M, Liao J, Dowling ML, et al. TRAIL inactivates the mitotic checkpoint and potentiates death induced by microtubule-targeting agents in human cancer cells.
Cancer Res
2008
;
68
:
3440
–9.
8
Tao W, South VJ, Zhang Y, et al. Induction of apoptosis by an inhibitor of the mitotic kinesin KSP requires both activation of the spindle assembly checkpoint and mitotic slippage.
Cancer Cell
2005
;
8
:
49
–59.
9
Sudo T, Nitta M, Saya H, Ueno NT. Dependence of paclitaxel sensitivity on a functional spindle assembly checkpoint.
Cancer Res
2004
;
64
:
2502
–8.
10
Masuda A, Maeno K, Nakagawa T, Saito H, Takahashi T. Association between mitotic spindle checkpoint impairment and susceptibility to the induction of apoptosis by anti-microtubule agents in human lung cancers.
Am J Pathol
2003
;
163
:
1109
–16.
11
Kienitz A, Vogel C, Morales I, Muller R, Bastians H. Partial downregulation of MAD1 causes spindle checkpoint inactivation and aneuploidy, but does not confer resistance towards taxol.
Oncogene
2005
;
24
:
4301
–10.
12
Kasai T, Iwanaga Y, Iha H, Jeang KT. Prevalent loss of mitotic spindle checkpoint in adult T-cell leukemia confers resistance to microtubule inhibitors.
J Biol Chem
2002
;
277
:
5187
–93.
13
Swanton C, Marani M, Pardo O, et al. Regulators of mitotic arrest and ceramide metabolism are determinants of sensitivity to paclitaxel and other chemotherapeutic drugs.
Cancer Cell
2007
;
11
:
498
–512.
14
Allan LA, Clarke PR. Phosphorylation of caspase-9 by CDK1/cyclin B1 protects mitotic cells against apoptosis.
Mol Cell
2007
;
26
:
301
–10.
15
Anand S, Penrhyn-Lowe S, Venkitaraman AR. AURORA-A amplification overrides the mitotic spindle assembly checkpoint, inducing resistance to Taxol.
Cancer Cell
2003
;
3
:
51
–62.
16
Vader G, Maia AF, Lens SM. The chromosomal passenger complex and the spindle assembly checkpoint: kinetochore-microtubule error correction and beyond.
Cell Div
2008
;
3
:
10
.
17
Ruchaud S, Carmena M, Earnshaw WC. Chromosomal passengers: conducting cell division.
Nat Rev Mol Cell Biol
2007
;
8
:
798
–812.
18
Wang X, Jin DY, Ng RW, et al. Significance of MAD2 expression to mitotic checkpoint control in ovarian cancer cells.
Cancer Res
2002
;
62
:
1662
–8.
19
Hernando E, Nahle Z, Juan G, et al. Rb inactivation promotes genomic instability by uncoupling cell cycle progression from mitotic control.
Nature
2004
;
430
:
797
–802.
20
Sotillo R, Hernando E, Diaz-Rodriguez E, et al. Mad2 overexpression promotes aneuploidy and tumorigenesis in mice.
Cancer Cell
2007
;
11
:
9
–23.
21
Guardavaccaro D, Frescas D, Dorrello NV, et al. Control of chromosome stability by the β-TrCP-REST-Mad2 axis.
Nature
2008
;
452
:
365
–9.
22
Perez de Castro I, de Carcer G, Malumbres M. A census of mitotic cancer genes: new insights into tumor cell biology and cancer therapy.
Carcinogenesis
2007
;
28
:
899
–912.
23
Cahill DP, Lengauer C, Yu J, et al. Mutations of mitotic checkpoint genes in human cancers.
Nature
1998
;
392
:
300
–3.
24
Saeki A, Tamura S, Ito N, et al. Frequent impairment of the spindle assembly checkpoint in hepatocellular carcinoma.
Cancer
2002
;
94
:
2047
–54.
25
Yoon DS, Wersto RP, Zhou W, et al. Variable levels of chromosomal instability and mitotic spindle checkpoint defects in breast cancer.
Am J Pathol
2002
;
161
:
391
–7.
26
Takahashi T, Haruki N, Nomoto S, et al. Identification of frequent impairment of the mitotic checkpoint and molecular analysis of the mitotic checkpoint genes, hsMAD2 and p55CDC, in human lung cancers.
Oncogene
1999
;
18
:
4295
–300.
27
Weaver BA, Cleveland DW. Does aneuploidy cause cancer?
Curr Opin Cell Biol
2006
;
18
:
658
–67.
28
Shichiri M, Yoshinaga K, Hisatomi H, Sugihara K, Hirata Y. Genetic and epigenetic inactivation of mitotic checkpoint genes hBUB1 and hBUBR1 and their relationship to survival.
Cancer Res
2002
;
62
:
13
–7.
29
Ricke RM, van Ree JH, van Deursen JM. Whole chromosome instability and cancer: a complex relationship.
Trends Genet
2008
;
24
:
457
–66.
30
Dobles M, Liberal V, Scott ML, Benezra R, Sorger PK. Chromosome missegregation and apoptosis in mice lacking the mitotic checkpoint protein Mad2.
Cell
2000
;
101
:
635
–45.
31
Kalitsis P, Earle E, Fowler KJ, Choo KH. Bub3 gene disruption in mice reveals essential mitotic spindle checkpoint function during early embryogenesis.
Genes Dev
2000
;
14
:
2277
–82.
32
Michel L, Diaz-Rodriguez E, Narayan G, Hernando E, Murty VV, Benezra R. Complete loss of the tumor suppressor MAD2 causes premature cyclin B degradation and mitotic failure in human somatic cells.
Proc Natl Acad Sci U S A
2004
;
101
:
4459
–64.
33
Kops GJ, Foltz DR, Cleveland DW. Lethality to human cancer cells through massive chromosome loss by inhibition of the mitotic checkpoint.
Proc Natl Acad Sci U S A
2004
;
101
:
8699
–704.
34
Tang Z, Shu H, Oncel D, Chen S, Yu H. Phosphorylation of Cdc20 by Bub1 provides a catalytic mechanism for APC/C inhibition by the spindle checkpoint.
Mol Cell
2004
;
16
:
387
–97.
35
Vogel C, Kienitz A, Hofmann I, Muller R, Bastians H. Crosstalk of the mitotic spindle assembly checkpoint with p53 to prevent polyploidy.
Oncogene
2004
;
23
:
6845
–53.
36
Ditchfield C, Johnson VL, Tighe A, et al. Aurora B couples chromosome alignment with anaphase by targeting BubR1, Mad2, and Cenp-E to kinetochores.
J Cell Biol
2003
;
161
:
267
–80.
37
Lee EA, Keutmann MK, Dowling ML, Harris E, Chan G, Kao GD. Inactivation of the mitotic checkpoint as a determinant of the efficacy of microtubule-targeted drugs in killing human cancer cells.
Mol Cancer Ther
2004
;
3
:
661
–9.
38
Vogel C, Kienitz A, Muller R, Bastians H. The mitotic spindle checkpoint is a critical determinant for topoisomerase-based chemotherapy.
J Biol Chem
2005
;
280
:
4025
–8.
39
Vogel C, Hager C, Bastians H. Mechanisms of mitotic cell death induced by chemotherapy-mediated G2 checkpoint abrogation.
Cancer Res
2007
;
67
:
339
–45.
40
Jordan MA, Wilson L. Microtubules as a target for anticancer drugs.
Nat Rev Cancer
2004
;
4
:
253
–65.
41
Keen N, Taylor SS. Aurora-kinase inhibitors as anticancer agents.
Nat Rev Cancer
2004
;
4
:
927
–36.
42
Marumoto T, Zhang D, Saya H. Aurora-A—a guardian of poles.
Nat Rev Cancer
2005
;
5
:
42
–50.
43
Hauf S, Cole RW, LaTerra S, et al. The small molecule Hesperadin reveals a role for Aurora B in correcting kinetochore-microtubule attachment and in maintaining the spindle assembly checkpoint.
J Cell Biol
2003
;
161
:
281
–94.
44
Schmidt M, Budirahardja Y, Klompmaker R, Medema RH. Ablation of the spindle assembly checkpoint by a compound targeting Mps1.
EMBO Rep
2005
;
6
:
866
–72.