6
Molecular Pathways Anaphase Catastrophe Is a Target for Cancer Therapy Fabrizio Galimberti 1 , Sarah L. Thompson 2 , Saranya Ravi 1 , Duane A. Compton 2,3 , and Ethan Dmitrovsky 1,3,4 Abstract Neoplastic cells are genetically unstable. Strategies that target pathways affecting genome instability can be exploited to disrupt tumor cell growth, potentially with limited consequences to normal cells. Chromosomal instability (CIN) is one type of genome instability characterized by mitotic defects that increase the rate of chromosome mis-segregation. CIN is frequently caused by extra centrosomes that transiently disrupt normal bipolar spindle geometry needed for accurate chromosome segregation. Tumor cells survive with extra centrosomes because of biochemical pathways that cluster centrosomes and promote chromosome segregation on bipolar spindles. Recent work shows that targeted inhibition of these pathways prevents centrosome clustering and forces chromosomes to segregate to multiple daughter cells, an event triggering apoptosis that we refer to as anaphase catastrophe. Anaphase catastrophe specifically kills tumor cells with more than 2 centrosomes. This death program can occur after genetic or pharmacologic inhibition of cyclin dependent kinase 2 (Cdk2) and is augmented by combined treatment with a microtubule inhibitor. This proapoptotic effect occurs despite the presence of ras mutations in cancer cells. Anaphase catastrophe is a previously unrecognized mechanism that can be pharmacologically induced for apoptotic death of cancer cells and is, therefore, appealing to engage for cancer therapy and prevention. Clin Cancer Res; 17(6); 1218–22. Ó2011 AACR. Background Anaphase catastrophe is a proapoptotic death mechan- ism observed in cancer cells with extra centrosomes that segregate chromosomes in the presence of multipolar spindles (1). The number of spindle poles in mitosis is determined by centrosomes, discrete organelles that nucle- ate spindle microtubules. Centrosome copy number is under strict cell cycle regulation. Centrosomes duplicate in S phase so that normal cells enter mitosis with 2 centrosomes and equally segregate replicated chromo- somes using bipolar spindles (Figs. 1A and 2A; ref. 2). Cells with extra centrosomes can undergo anaphase with multipolar spindles and segregate chromosomes impro- perly to more than 2 daughter cells (Figs. 1C and 2B). We call this lethal event for each daughter cell (3, 4) anaphase catastrophe. Anaphase catastrophe selectively targets cancer cells with extra centrosomes and spares normal cells that enter mitosis with only 2 centrosomes and, therefore, are incapable of segregating chromosomes to more than 2 spindle poles. Targeting chromosomally unstable cancer cells Recent work revealed a mechanism that induces cell death preferentially in cancer cells with chromosomal instability (CIN; ref. 1). This mechanism can be exploited therapeutically. CIN is common in aneuploid tumor cells and is usually caused by the persistence of inappropriate attachments of chromosomes to spindle microtubules (5–7). The prevalence of attachment errors increases sharply in cells with extra centrosomes, owing to the key role of centrosomes in determining the num- ber of spindle poles during mitosis (3, 8). Cancer cells gain extra copies of centrosomes either from failure of cytokinesis or by deregulation of the strict cell cycle control of centrosome duplication. Because chromosome segregation is vital for cell survival, cancer cells with extra centrosomes assemble bipolar spindles in mitosis by clustering supernumerary centrosomes together at spin- dle poles (3, 4, 8). Many biochemical pathways promote centrosome clus- tering during mitosis. Initial evidence shows that these pathways can be pharmacologically targeted to induce anaphase catastrophe (Fig. 2C). A genome-wide screen using Drosophila-cultured cells identified 133 distinct genes required for centrosome clustering (4). The products of these genes participate in diverse cellular processes, includ- ing regulation of the actin cytoskeleton, spindle assembly, spindle assembly checkpoint (SAC) activity, and cell adhe- sion. Positive hits in that screen that represent potential Authors' Affiliations: 1 Department of Pharmacology and Toxicology, 2 Department of Biochemistry, 3 Norris Cotton Cancer Center, and 4 Depart- ment of Medicine, Dartmouth Medical School, Hanover, New Hampshire; and Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire Note: F. Galimberti and S. L. Thompson contributed equally to this work. Corresponding Author: Ethan Dmitrovsky, Department of Pharmacology and Toxicology, Dartmouth Medical School, Remsen 7650, Hanover, NH 03755. Phone: 603-650-1707; Fax: 603-650-1129; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-10-1178 Ó2011 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 17(6) March 15, 2011 1218 on February 14, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst February 2, 2011; DOI: 10.1158/1078-0432.CCR-10-1178

Anaphase Catastrophe Is a Target for Cancer Therapyclincancerres.aacrjournals.org/content/clincanres/17/6/1218.full.pdf · Note: F. Galimberti and S. L. Thompson contributed equally

Embed Size (px)

Citation preview

Molecular Pathways

Anaphase Catastrophe Is a Target for Cancer Therapy

Fabrizio Galimberti1, Sarah L. Thompson2, Saranya Ravi1, Duane A. Compton2,3, and Ethan Dmitrovsky1,3,4

AbstractNeoplastic cells are genetically unstable. Strategies that target pathways affecting genome instability can

be exploited to disrupt tumor cell growth, potentially with limited consequences to normal cells.

Chromosomal instability (CIN) is one type of genome instability characterized by mitotic defects that

increase the rate of chromosome mis-segregation. CIN is frequently caused by extra centrosomes that

transiently disrupt normal bipolar spindle geometry needed for accurate chromosome segregation. Tumor

cells survive with extra centrosomes because of biochemical pathways that cluster centrosomes and

promote chromosome segregation on bipolar spindles. Recent work shows that targeted inhibition of

these pathways prevents centrosome clustering and forces chromosomes to segregate to multiple daughter

cells, an event triggering apoptosis that we refer to as anaphase catastrophe. Anaphase catastrophe

specifically kills tumor cells with more than 2 centrosomes. This death program can occur after genetic

or pharmacologic inhibition of cyclin dependent kinase 2 (Cdk2) and is augmented by combined

treatment with a microtubule inhibitor. This proapoptotic effect occurs despite the presence of ras

mutations in cancer cells. Anaphase catastrophe is a previously unrecognized mechanism that can be

pharmacologically induced for apoptotic death of cancer cells and is, therefore, appealing to engage for

cancer therapy and prevention. Clin Cancer Res; 17(6); 1218–22. �2011 AACR.

Background

Anaphase catastrophe is a proapoptotic death mechan-ism observed in cancer cells with extra centrosomes thatsegregate chromosomes in the presence of multipolarspindles (1). The number of spindle poles in mitosis isdetermined by centrosomes, discrete organelles that nucle-ate spindle microtubules. Centrosome copy number isunder strict cell cycle regulation. Centrosomes duplicatein S phase so that normal cells enter mitosis with 2centrosomes and equally segregate replicated chromo-somes using bipolar spindles (Figs. 1A and 2A; ref. 2).Cells with extra centrosomes can undergo anaphase withmultipolar spindles and segregate chromosomes impro-perly to more than 2 daughter cells (Figs. 1C and 2B). Wecall this lethal event for each daughter cell (3, 4) anaphasecatastrophe. Anaphase catastrophe selectively targets cancercells with extra centrosomes and spares normal cells thatenter mitosis with only 2 centrosomes and, therefore, are

incapable of segregating chromosomes to more than 2spindle poles.

Targeting chromosomally unstable cancer cellsRecent work revealed a mechanism that induces cell

death preferentially in cancer cells with chromosomalinstability (CIN; ref. 1). This mechanism can beexploited therapeutically. CIN is common in aneuploidtumor cells and is usually caused by the persistence ofinappropriate attachments of chromosomes to spindlemicrotubules (5–7). The prevalence of attachment errorsincreases sharply in cells with extra centrosomes, owingto the key role of centrosomes in determining the num-ber of spindle poles during mitosis (3, 8). Cancer cellsgain extra copies of centrosomes either from failure ofcytokinesis or by deregulation of the strict cell cyclecontrol of centrosome duplication. Because chromosomesegregation is vital for cell survival, cancer cells with extracentrosomes assemble bipolar spindles in mitosis byclustering supernumerary centrosomes together at spin-dle poles (3, 4, 8).

Many biochemical pathways promote centrosome clus-tering during mitosis. Initial evidence shows that thesepathways can be pharmacologically targeted to induceanaphase catastrophe (Fig. 2C). A genome-wide screenusing Drosophila-cultured cells identified 133 distinct genesrequired for centrosome clustering (4). The products ofthese genes participate in diverse cellular processes, includ-ing regulation of the actin cytoskeleton, spindle assembly,spindle assembly checkpoint (SAC) activity, and cell adhe-sion. Positive hits in that screen that represent potential

Authors' Affiliations: 1Department of Pharmacology and Toxicology,2Department of Biochemistry, 3Norris Cotton Cancer Center, and 4Depart-ment of Medicine, Dartmouth Medical School, Hanover, New Hampshire;and Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire

Note: F. Galimberti and S. L. Thompson contributed equally to this work.

Corresponding Author: Ethan Dmitrovsky, Department of Pharmacologyand Toxicology, Dartmouth Medical School, Remsen 7650, Hanover,NH 03755. Phone: 603-650-1707; Fax: 603-650-1129; E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-10-1178

�2011 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 17(6) March 15, 20111218

on February 14, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2011; DOI: 10.1158/1078-0432.CCR-10-1178

targets for inhibition include the mitotic kinesin HSET,myosin 10A, and the enzyme tankyrase, which modifiesproteins involved in spindle pole organization. HSET is notrequired for mitosis in normal somatic cells with 2 centro-somes (9). Loss of HSET function in cells with supernu-merary centrosomes can induce anaphase catastrophespecifically within cells having extra centrosomes (4). Thisfinding provides a proof of principle that targeted inhibi-tion of these enzymes causes anaphase catastrophe andjustifies the search for inhibitors of these enzymes or othertargets that might cause anaphase catastrophe. The powerof the genetic screens is offset by the fact that the strategywill only identify target genes whose products function at aspecific phase of the cell cycle. Other biochemical pathwaysmight participate in centrosome clustering. These pathwaysrepresent additional opportunities for inducing anaphasecatastrophe. As one example, lung cancers often overex-press cyclin E, which can deregulate cyclin-dependentkinase (Cdk) activity (10). Targeting Cdk2, one of thesederegulated kinases, triggers anaphase catastrophe (1).Cell viability relies on the equal separation of replicated

chromosomes during mitosis. Mitotic fidelity is enhancedby the SAC (11). The SAC prevents sister chromatid separa-tion and anaphase onset until all chromosomes formappropriate bipolar attachments to spindle microtubules.A single unaligned or unattached chromosome is sufficientto maintain SAC activity and prevent entry into anaphase(12, 13). Thus, a defined sequence of events is strictlyfollowed to ensure accurate chromosome segregation in

normal cells. Chromosomes form attachments to spindlemicrotubules and align at the equator of a bipolar spindle;that satisfies the SAC leading to activation of the ubiquitinligase anaphase promoting complex/cyclosome (APC/C).APC/C subsequently triggers degradation of securin andthe mitotic cyclin B to induce sister chromatid separationand exit from mitosis, respectively, so that daughter cellsenter G1 phase of the next cell cycle with the appropriatenumbers of chromosomes (14).

Anaphase catastrophe versus mitotic catastropheChemotherapeutic drugs such as paclitaxel disrupt

microtubule dynamics and chromosome attachment tospindle microtubules (15); this prolongs mitosis by pre-venting satisfaction of the SAC. The cellular response toprolonged mitotic arrest varies depending on the cell lineand even among cells within the same cell line; the out-come of this response depends on processes that regulatecyclin B levels (16, 17). Cyclin B quantities graduallydecrease and apoptotic signals gradually increase in mitoticcells. In some cells, mitosis can be sufficiently prolonged byantimitotic drugs, which promote apoptotic signals thatcan exceed a critical threshold and induce death in mitosis.In other cells, cyclin B levels drop below a critical thresholdduringmitotic arrest and cells exit mitosis and reenter G1 astetraploid cells without chromosome segregation. Thesetetraploid cells typically enter senescence or undergo apop-tosis (Fig. 1B), although some propagate and endocycle(16). Exit from mitosis without chromosome segregation

© 2011 American Association for Cancer ResearchR2011 AAmerican Association for CancCA i A i ti f CA i ti f C

Mitotic

slippage

or

adaptation

Anaphase

catastrophe

Normal

mitosis

A

B

C

G2 M FATE

Normal

daughter

cells

Tetraploidy

Apoptosis

Cell

death

Figure 1. Fates of mitotic cells. Cells can undergo diverse fates according to their status at anaphase. A, proper segregation of chromosomes in mitosisleads to the generation of 2 genetically identical daughter cells. B, gradual degradation of cyclin B in the presence of prolonged spindle checkpoint activationcauses cells to exit mitosis without dividing chromosomes in anaphase, termed slippage. Cells that exit mitosis via slippage enter G1 as tetraploid cells. Thesecells may continue to cycle, senesce, or undergo apoptosis. C, anaphase catastrophe occurs when a cell with multiple centrosomes fails to coalescecentrosomes into 2 spindle poles and enters anaphasewith amultipolar spindle. Segregation of chromosomes tomore than 2 daughter cells causes cell death.

Anaphase Catastrophe Is a Target for Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(6) March 15, 2011 1219

on February 14, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2011; DOI: 10.1158/1078-0432.CCR-10-1178

following extended mitotic arrest has been termed adapta-tion because cells are said to "adapt" to prolonged check-point activity (18). Perhaps a more appropriate term forthis is mitotic slippage, because cells slip out of mitosis

without satisfying the SAC (17, 19). Notably, mitoticslippage or adaptation violates the temporal sequence ofevents needed for proper chromosome segregation becausecells enter G1 of the next cell cycle without satisfying theSAC or adequately activating APC/C. Cancer cells with CINare no more likely to continue cycling following mitoticslippage than are diploid cells (16), indicating that thisalone does not selectively kill tumor cells. Nonetheless, it isproposed that substantial DNA damage conferred by che-motherapeutic agents or mutation of DNA damageresponse genes can promote cell death during mitosis ormitotic slippage (collectively known as mitotic catastrophein ref. 20) of cancer cells. Cells that enter anaphase withmultipolar spindles abide by the appropriate temporalsequence of biochemical events for mitotic exit and onlyinitiate chromosome separation after all chromosomes areattached to the spindle. This event is mechanistically dis-tinct from mitotic slippage or adaptation, which is why wetermed it anaphase catastrophe.

Clinical-Translational Advances

An established paradigm for cancer therapy involvestargeting and killing dividing cells. Many chemotherapeu-tic agents target dividing cells during mitosis, which is asensitive window in the cell cycle during which chromo-somes align and separate to form genetically identicaldaughter cells. Taxanes and vinca alkaloids successfully killtumor cells during mitosis by targeting microtubules anddisrupting normal chromosome movement. However,these drugs are not specific to cancer cells and disruptmicrotubules in all cells, leading to side effects such asneutropenia and neurotoxicity (15). Nevertheless, thesedrug effects establish that mitotic disruption is engagedby chemotherapy treatments. On the basis of this finding,compounds are being developed that inhibit proteins thatonly function during mitosis and would not target non-dividing cells. Clinical trials are underway to explore theefficacy of inhibitingmolecularmotors required for bipolarspindle organization (21) and of inhibitors of the essentialAurora and Polo-like kinases (22, 23). Yet, these drugstarget actively dividing cells and do not necessarily exploitkey differences between malignant and benign cells, whichmight spare normal cells. For example, cells in most solidtumors are aneuploid with chromosome numbers thatdeviate from a multiple of the haploid genome. The roleof aneuploidy in tumorigenesis is under study throughexperiments conducted in clinically relevant animal mod-els (24, 25). To date, those efforts have not identifiedspecific treatment strategies that would selectively targetaneuploid cells and spare diploid cells. However, the recentinsights into the causes of CIN (3, 26), described above,revealed that the proapoptotic mechanism of anaphasecatastrophe can be pharmacologically targeted to selec-tively kill tumor cells.

We recently showed that targeted depletion of Cdk2, butnot Cdk1, induced anaphase catastrophe in lung cancercells; current work is elucidating Cdk2 targets that mediate

© 2011 American Association for Cancer Research

A B

C

Survival

Centrosome

clustering

Cdk2 inhibitor

HSET inhibitor

ILK inhibitor Multipolar

spindle

formation

Supernumerary

centrosomes

Centrosome

overduplication

Plk4 inhibitor

merican Associ tiation for Cancer Resea

Centrosome

Death

Figure 2. Inhibition of Cdk2 and other pathways triggers anaphasecatastrophe in lung cancer cells. Mouse lung cancer cells overexpressingcyclin E were treated with control siRNA, A, or Cdk2 targeting siRNA, B, for24 hours. Cells were stained for microtubules in red and DNA in blue. Arepresentative bipolar anaphase, A, and a representative multipolaranaphase leading to anaphase catastrophe, B, are shown. Scale bar, 10mm. C, pharmacologic or genetic inhibition of numerous pathways affectsmitotic fidelity and can trigger anaphase catastrophe leading to cell death.Centrosome amplification leads to supernumerary centrosomes, whichcan be prevented with a Plk4 inhibitor. Centrosome clustering enablescancer cells to survive through mitosis in the presence of supernumerarycentrosomes. Inhibition of Cdk2, HSET, or ILK inhibits centrosomeclustering, resulting in multipolar spindle formation; this causes multipolaranaphases that trigger anaphase catastrophe.

Galimberti et al.

Clin Cancer Res; 17(6) March 15, 2011 Clinical Cancer Research1220

on February 14, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2011; DOI: 10.1158/1078-0432.CCR-10-1178

this effect (Fig. 2B; ref. 1). Cdk inhibitors exist, and somesuch as flavopiridol and UCN-01 exert some clinical anti-tumor activity (27). Another inhibitor (seliciclib, CYC202,R-roscovitine) can preferentially inhibit Cdk2 at low con-centrations, whereas at higher concentrations inhibition ofCdk1, Cdk7, or Cdk9 is observed (28). Anaphase cata-strophe is induced when cells with extra centrosomes areexposed to seliciclib at dosages that should only targetCdk2 (1). Similar concentrations had minimal mitoticeffects on immortalized pulmonary epithelial cells, imply-ing differential activity against cancer cells (1). Even tran-sient exposures (4 hours) to a Cdk2 inhibitor increased thenumber of cells undergoing anaphase catastrophe, suggest-ing that the drug acts by inhibiting cyclin-dependent kinaseactivity during mitosis and not during centrosome duplica-tion in S phase (1). These and other findings provide aproof of principle for anaphase catastrophe induction aftertreatment with specific drugs. Future work should elucidatewhether mechanisms seen in vitro can be observed in vivovia proof-of-principle clinical trials.Consistent with this concept was the finding that integ-

rin-linked kinase (ILK) activity is essential for centrosomeclustering in cancer cells (29). ILK regulates actin and celladhesion at focal adhesion sites as well as microtubule-associated components during mitosis, likely through reg-ulation of Aurora A/TACC3/TOG activity (30). Small mole-cule inhibitors of ILK can induce anaphase catastrophewithin breast cancer cells with extra centrosomes, but notwithin normal cells or cancer cells without extra centro-somes (31). Therapeutic strategies that inhibit key targetenzymes or pathways responsible for centrosome cluster-ing are being uncovered that will selectively kill cancer cellswith extra centrosomes, while sparing normal cells. Animportant future direction for study is to identify down-stream targets of Cdk2 and ILK that are essential forcentrosome clustering. That search would likely revealother candidate targets that induce anaphase catastrophe.Intriguingly, drugs that induce anaphase catastrophe

seem to cooperate with taxanes. Taxanes are routinelyused in the treatment of breast, bladder, ovarian, headand neck, and lung cancer (15). These agents specificallytarget microtubules and disrupt the normal timing ofmitosis by delaying the satisfaction of the SAC. Treatmentof cells with combinations of either the Cdk2 inhibitor

seliciclib and taxanes or the ILK inhibitor QLT-0267 andtaxanes showed significant increases in cancer cell death(1, 31). In the case of combining seliciclib and taxanes,this increase was linked to augmented anaphase cata-strophe (1). These combinations likely force cells toundergo catastrophic anaphase more efficiently than witheither treatment alone. Combining an agent that inducesanaphase catastrophe with a microtubule-targeting drugis an attractive regimen to consider in future clinical trialsfor appropriate cancers.

In this regard, pharmacogenomic analysis revealed thatlung cancer cells with k-ras mutations are especially sensi-tive to Cdk2 inhibition (1). K-ras mutation typically pre-dicts resistance to an epidermal growth factor receptor–tyrosine kinase inhibitor (EGFR-TKI; ref. 32). This muta-tion is found in most pancreatic cancers, in about 30% oflung cancers, and in many other cancers (33, 34). Thisfinding suggests that a regimen that augments anaphasecatastrophe is appealing to consider for treating cancersthat harbor ras mutations.

In conclusion, recent findings reveal anaphase in mitoticcells with multipolar spindles is a lethal event that ispharmacologically conferred. Anaphase catastrophe wouldselectively kill cancer cells with extra centrosomes and likelyspare normal cells. This unique pathway, identified inmitosis, could discriminate between cancerous and normalcells. Thus, anaphase catastrophe is a novel anti-neoplasticmechanism to engage for cancer therapy and prevention.

Disclosure of Potential Conflicts of Interest

E. Dmitrovsky previously received a research grant from Cyclacel. Theother authors disclosed no potential conflicts of interest.

Grant Support

This work was supported by NIH and National Cancer Institute (NCI) grantsRO1-CA087546 (E. Dmitrovsky) and RO1-CA111422 (E. Dmitrovsky); RO1-GM51542 (D. A. Compton); T32-GM008704 (S. L. Thompson); and a SamuelWaxman Cancer Research Foundation Award (E. Dmitrovsky and D.A. Comp-ton). E. Dmitrovsky is an American Cancer Society Clinical Research Professorsupported by a generous gift from the F. M. Kirby Foundation.

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

Received September 23, 2010; revised November 23, 2010; acceptedDecember 1, 2010; published OnlineFirst February 2, 2011.

References1. Galimberti F, Thompson SL, Liu X, Li H, Memoli V, Green SR, et al.

Targeting the cyclin E-Cdk-2 complex represses lung cancer growthby triggering anaphase catastrophe. Clin Cancer Res 2010;16:109–20.

2. Doxsey S. Re-evaluating centrosome function. Nat Rev Mol Cell Biol2001;2:688–98.

3. Ganem NJ, Godinho SA, Pellman D. A mechanism linking extracentrosomes to chromosomal instability. Nature 2009;460:278–82.

4. KwonM, Godinho SA, Chandhok NS, GanemNJ, Azioune A, Thery M,et al. Mechanisms to suppress multipolar divisions in cancer cells withextra centrosomes. Genes Dev 2008;22:2189–203.

5. Thompson SL, Compton DA. Examining the link between chromo-somal instability and aneuploidy in human cells. J Cell Biol2008;180:665–72.

6. Bakhoum SF, Thompson SL, Manning AL, Compton DA. Genomestability is ensured by temporal control of kinetochore-microtubuledynamics. Nat Cell Biol 2009;11:27–35.

7. Bakhoum SF, Genovese G, Compton DA. Deviant kinetochore micro-tubule dynamics underlie chromosomal instability. Curr Biol2009;19:1937–42.

8. Quintyne NJ, Reing JE, Hoffelder DR, Gollin SM, Saunders WS.Spindle multipolarity is prevented by centrosomal clustering. Science2005;307:127–9.

9. Mountain V, Simerly C, Howard L, Ando A, Schatten G, Compton DA.The kinesin-related protein, HSET, opposes the activity of Eg5 andcross-links microtubules in the mammalian mitotic spindle. J Cell Biol1999;147:351–66.

Anaphase Catastrophe Is a Target for Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(6) March 15, 2011 1221

on February 14, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2011; DOI: 10.1158/1078-0432.CCR-10-1178

10. Lonardo F, Rusch V, Langenfeld J, Dmitrovsky E, Klimstra DS. Over-expression of cyclins D1 and E is frequent in bronchial preneoplasiaand precedes squamous cell carcinoma development. Cancer Res1999;59:2470–6.

11. Musacchio A, Salmon ED. The spindle-assembly checkpoint in spaceand time. Nat Rev Mol Cell Biol 2007;8:379–93.

12. Rieder CL, Cole RW, Khodjakov A, Sluder G. The checkpoint delayinganaphase in response to chromosome monoorientation is mediatedby an inhibitory signal produced by unattached kinetochores. J CellBiol 1995;130:941–8.

13. Rieder CL, Schultz A, Cole R, Sluder G. Anaphase onset in vertebratesomatic cells is controlled by a checkpoint that monitors sisterkinetochore attachment to the spindle. J Cell Biol 1994;127:1301–10.

14. Peters JM. The anaphase-promoting complex: proteolysis in mitosisand beyond. Mol Cell 2002;9:931–43.

15. Zelnak AB. Clinical pharmacology and use of microtubule-targetingagents in cancer therapy. Methods Mol Med 2007;137:209–34.

16. Gascoigne KE, Taylor SS. Cancer cells display profound intra- andinterline variation following prolonged exposure to antimitotic drugs.Cancer Cell 2008;14:111–22.

17. Brito DA, Rieder CL. Mitotic checkpoint slippage in humans occurs viacyclin B destruction in the presence of an active checkpoint. Curr Biol2006;16:1194–200.

18. Lanni JS, Jacks T. Characterization of the p53-dependent postmitoticcheckpoint following spindle disruption. Mol Cell Biol 1998;18:1055–64.

19. Rieder CL, Maiato H. Stuck in division or passing through: whathappens when cells cannot satisfy the spindle assembly checkpoint.Dev Cell 2004;7:637–51.

20. Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, KroemerG. Cell death bymitotic catastrophe: amolecular definition. Oncogene2004;23:2825–37.

21. Sakowicz R, Finer JT, Beraud C, Crompton A, Lewis E, Fritsch A, et al.Antitumor activity of a kinesin inhibitor. Cancer Res 2004;64:3276–80.

22. Warner SL, Stephens BJ, Von Hoff DD. Tubulin-associated proteins:Aurora and Polo-like kinases as therapeutic targets in cancer. CurrOncol Rep 2008;10:122–9.

23. Schvartzman JM, Sotillo R, Benezra R. Mitotic chromosomal instabil-ity and cancer: mouse modelling of the human disease. Nat RevCancer 2010;10:102–15.

24. Holland AJ, Cleveland DW. Boveri revisited: chromosomal instability,aneuploidy and tumorigenesis. Nat RevMol Cell Biol 2009;10:478–87.

25. Foijer F, Draviam VM, Sorger PK. Studying chromosome instability inthe mouse. Biochim Biophys Acta 2008;1786:73–82.

26. Thompson SL, Bakhoum SF, Compton DA. Mechanisms of chromo-somal instability. Curr Biol 2010;20:R285–95.

27. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changingparadigm. Nat Rev Cancer 2009;9:153–66.

28. Collins I, Garrett MD. Targeting the cell division cycle in cancer: CDKand cell cycle checkpoint kinase inhibitors. Curr Opin Pharmacol2005;5:366–73.

29. Fielding AB, Lim S, Montgomery K, Dobreva I, Dedhar S. A critical roleof integrin-linked kinase, ch-TOG and TACC3 in centrosome cluster-ing in cancer cells. Oncogene 2010. Epub 2010 Sep 13.

30. Fielding AB, Dobreva I, McDonald PC, Foster LJ, Dedhar S. Integrin-linked kinase localizes to the centrosome and regulates mitoticspindle organization. J Cell Biol 2008;180:681–9.

31. Kalra J, Warburton C, Fang K, Edwards L, Daynard T, Waterhouse D,et al. QLT0267, a small molecule inhibitor targeting integrin-linkedkinase (ILK), and docetaxel can combine to produce synergisticinteractions linked to enhanced cytotoxicity, reductions in P-AKTlevels, altered F-actin architecture and improved treatment outcomesin an orthotopic breast cancer model. Breast Cancer Res 2009;11:R25.

32. Massarelli E, Varella-Garcia M, Tang X, Xavier AC, Ozburn NC, Liu DD,et al. KRASmutation is an important predictor of resistance to therapywith epidermal growth factor receptor tyrosine kinase inhibitors innon-small-cell lung cancer. Clin Cancer Res 2007;13:2890–6.

33. Aviel-Ronen S, Blackhall FH, Shepherd FA, Tsao MS. K-ras mutationsin non-small-cell lung carcinoma: a review. Clin Lung Cancer2006;8:30–8.

34. Quinlan MP, Settleman J. Explaining the preponderance of Krasmutations in human cancer: An isoform-specific function in stem cellexpansion. Cell Cycle 2008;7:1332–5.

Galimberti et al.

Clin Cancer Res; 17(6) March 15, 2011 Clinical Cancer Research1222

on February 14, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2011; DOI: 10.1158/1078-0432.CCR-10-1178

2011;17:1218-1222. Published OnlineFirst February 2, 2011.Clin Cancer Res   Fabrizio Galimberti, Sarah L. Thompson, Saranya Ravi, et al.   Anaphase Catastrophe Is a Target for Cancer Therapy

  Updated version

  10.1158/1078-0432.CCR-10-1178doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/17/6/1218.full#ref-list-1

This article cites 33 articles, 12 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/17/6/1218.full#related-urls

This article has been cited by 7 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://clincancerres.aacrjournals.org/content/17/6/1218To request permission to re-use all or part of this article, use this link

on February 14, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2011; DOI: 10.1158/1078-0432.CCR-10-1178