inhibitors targeting mitosis: tales of how great drugs ...transport kinesins, which function in...

13
Inhibitors Targeting Mitosis: Tales of How Great Drugs against a Promising Target Were Brought Down by a Flawed Rationale Edina Komlodi-Pasztor 1 , Dan L. Sackett 2 , and Antonio Tito Fojo 1 Abstract Although they have been advocated with an understandable enthusiasm, mitosis-specific agents such as inhibitors of mitotic kinases and kinesin spindle protein have not been successful clinically. These drugs were developed as agents that would build on the success of microtubule-targeting agents while avoiding the neurotoxicity that encumbers drugs such as taxanes and vinca alkaloids. The rationale for using mitosis- specific agents was based on the thesis that the clinical efficacy of microtubule-targeting agents could be ascribed to the induction of mitotic arrest. However, the latter concept, which has long been accepted as dogma, is likely important only in cell culture and rapidly growing preclinical models, and irrelevant in patient tumors, where interference with intracellular trafficking on microtubules is likely the principal mechanism of action. Here we review the preclinical and clinical data for a diverse group of inhibitors that target mitosis and identify the reasons why these highly specific, myelosuppressive compounds have failed to deliver on their promise. Clin Cancer Res; 18(1); 51–63. Ó2012 AACR. Introduction On the premise that tumors harbor a (much) larger fraction of actively dividing cells than do normal tissues and should therefore be more vulnerable to agents aimed at cell division, researchers have developed drugs to target numerous components of this intricate process as che- motherapeutics. Chief among these are agents that target the processes of mitosis and the accompanying cytokinesis, with inhibitors of mitotic kinases being among the most recent entries (1). Although the clinical efficacy of agents that target the process of cell division [e.g., microtubule- targeting agents (MTA)] provided support for this rationale, accumulating clinical and preclinical evidence is encourag- ing a reassessment of how MTAs act (2). Our increasing understanding of the working of cells in general and cancer cells in particular, together with greater knowledge about the diversity of targets of our chemotherapeutic agents, suggests that a more complex approach than only targeting cell division may be warranted. Among the components of cell division, mitosis, the process whereby a eukaryotic cell separates its replicated chromosomes into identical sets in 2 offspring, has long been viewed as an attractive target for chemotherapeutic agents. Together, mitosis and the accom- panying cytokinesis that divides the nucleus, cytoplasmic organelles, and cell membrane into 2 cells define the mitotic or M-phase of the cell cycle. In most cells, this accounts for less (and often substantially less) than 10% of the cell cycle. Emerging knowledge is revealing the complexity of the mitotic process and allowing the identification of a diverse group of proteins whose activity is precisely orchestrated during mitosis. Mitosis requires many other players in addition to tubulin and microtubules (MT). Many of these other proteins are primarily functional only during mitosis and are responsible for controlling different steps in the assembly and function of the complex machinery of the mitotic spindle. Among these proteins, the Aurora kinases (AK), Polo-like kinases (PLK), and kinesin spindle protein (KSP) have emerged as targets for cancer therapeutics (3, 4). Mitosis: The Role of MTs and Mitotic Kinases As shown in Fig. 1, ordered arrays of MTs play essential roles in both interphase (G 1 -, S-, and G 2 -phases) and mitosis. In animal cells, centrosomes act as MT-organizing centers (i.e., the site of MT nucleation), the structures from which MTs emerge (5). Centrosomal nucleation of polar- ized arrays of MTs is essential for mitosis and for cellular organization during the large portion of the cell cycle that is not mitosis. In interphase, the MT-organizing center orga- nizes the array of MTs that provides polarity to the cyto- plasm. This array is an essential structure for the cellular trafficking of a myriad of proteins, including many Authors' Afliations: 1 Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, and 2 Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Antonio T. Fojo, National Cancer Institute, 10 Center Drive, MSC 1906, Bldg. 10 Rm. 12N226, Bethesda, MD 20892- 1906. Phone: 301-402-1357; Fax: 301-402-1680; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-0999 Ó2012 American Association for Cancer Research. CCR FOCUS www.aacrjournals.org 51 Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association for https://bloodcancerdiscov.aacrjournals.org Downloaded from

Upload: others

Post on 15-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

Inhibitors Targeting Mitosis: Tales of How Great Drugsagainst a Promising Target Were Brought Down by a FlawedRationale

Edina Komlodi-Pasztor1, Dan L. Sackett2, and Antonio Tito Fojo1

AbstractAlthough they have been advocated with an understandable enthusiasm, mitosis-specific agents such as

inhibitors of mitotic kinases and kinesin spindle protein have not been successful clinically. These drugs

were developed as agents thatwould build on the success ofmicrotubule-targeting agentswhile avoiding the

neurotoxicity that encumbers drugs such as taxanes and vinca alkaloids. The rationale for using mitosis-

specific agents was based on the thesis that the clinical efficacy of microtubule-targeting agents could be

ascribed to the induction of mitotic arrest. However, the latter concept, which has long been accepted as

dogma, is likely important only in cell culture and rapidly growing preclinical models, and irrelevant in

patient tumors, where interference with intracellular trafficking on microtubules is likely the principal

mechanism of action. Here we review the preclinical and clinical data for a diverse group of inhibitors that

target mitosis and identify the reasons why these highly specific, myelosuppressive compounds have failed

to deliver on their promise. Clin Cancer Res; 18(1); 51–63. �2012 AACR.

Introduction

On the premise that tumors harbor a (much) largerfraction of actively dividing cells than do normal tissuesand should therefore bemore vulnerable to agents aimed atcell division, researchers have developed drugs to targetnumerous components of this intricate process as che-motherapeutics. Chief among these are agents that targetthe processes of mitosis and the accompanying cytokinesis,with inhibitors of mitotic kinases being among the mostrecent entries (1). Although the clinical efficacy of agentsthat target the process of cell division [e.g., microtubule-targeting agents (MTA)] provided support for this rationale,accumulating clinical and preclinical evidence is encourag-ing a reassessment of how MTAs act (2). Our increasingunderstanding of the working of cells in general and cancercells in particular, together with greater knowledge aboutthe diversity of targets of our chemotherapeutic agents,suggests that a more complex approach than only targetingcell division may be warranted. Among the components of

cell division, mitosis, the process whereby a eukaryotic cellseparates its replicated chromosomes into identical sets in 2offspring, has long been viewed as an attractive target forchemotherapeutic agents. Together, mitosis and the accom-panying cytokinesis that divides the nucleus, cytoplasmicorganelles, and cellmembrane into 2 cells define themitoticor M-phase of the cell cycle. In most cells, this accounts forless (and often substantially less) than 10%of the cell cycle.Emerging knowledge is revealing the complexity of themitotic process and allowing the identification of a diversegroup of proteins whose activity is precisely orchestratedduring mitosis. Mitosis requires many other players inaddition to tubulin and microtubules (MT). Many of theseother proteins are primarily functional only during mitosisand are responsible for controlling different steps in theassembly and function of the complex machinery of themitotic spindle. Among these proteins, the Aurora kinases(AK), Polo-like kinases (PLK), and kinesin spindle protein(KSP) have emerged as targets for cancer therapeutics (3, 4).

Mitosis: The Role of MTs and Mitotic Kinases

As shown in Fig. 1, ordered arrays of MTs play essentialroles in both interphase (G1-, S-, and G2-phases) andmitosis. In animal cells, centrosomes act as MT-organizingcenters (i.e., the site of MT nucleation), the structures fromwhich MTs emerge (5). Centrosomal nucleation of polar-ized arrays of MTs is essential for mitosis and for cellularorganization during the large portion of the cell cycle that isnot mitosis. In interphase, the MT-organizing center orga-nizes the array of MTs that provides polarity to the cyto-plasm. This array is an essential structure for the cellulartrafficking of a myriad of proteins, including many

Authors' Affiliations: 1Medical Oncology Branch, Center for CancerResearch, National Cancer Institute, and 2Program in Physical Biology,Eunice Kennedy Shriver National Institute of Child Health and HumanDevelopment, NIH, Bethesda, Maryland

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Author: Antonio T. Fojo, National Cancer Institute, 10Center Drive, MSC 1906, Bldg. 10 Rm. 12N226, Bethesda, MD 20892-1906. Phone: 301-402-1357; Fax: 301-402-1680; E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-11-0999

�2012 American Association for Cancer Research.

CCRFOCUS

www.aacrjournals.org 51

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 2: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

important oncoproteins. Polarized arrays of MTs growoutward from near the nucleus in growing epithelial cells,and fromapical to basal in polarized cells (6). Their polarityis recognized by motor proteins of the dynein and kinesinsuperfamilies, allowing for directed movement of cargo onMTs (7). Dyneins move toward the minus (nuclear) end ofMTs, whereas kinesinsmostlymove to the plus (peripheral)end of MTs. In cells undergoing cell division, a majorfunction of the centrosome is to organize the 2 opposingarrays that form the mitotic spindle apparatus, which isrequired for separation of chromosomes during celldivision.

MTs provide the structure and machinery for chromo-some segregationduringmitosis, a process that also requiresan array of other proteins, including essential serine/thre-onine kinases. Figure 2 depicts the principal localization ofthe mitosis-associated proteins that have been targets ofdrugs in clinical trials (8). Figure 3 shows the proteins thatmodulate the activity of the mitotic kinases and thosethrough which the effects of these mitosis-associated pro-teins aremediated. Although themitotic kinases often act inconcert, they also perform unique functions without over-lap. During S-phase, Aurora kinase A (AKA) localizes to thecentrosome. It then translocates to themitotic poles and theadjacent spindle MTs as the cell progresses through pro-phase, metaphase, and anaphase, eventually locating in themidbody during telophase and cytokinesis (9). Duringtranslocation from the centrosome, the level of AKA

increases. This indicates that expression is largely restrictedto mitosis and that agents designed to target AKA would beinactive in cells in other phases of the cell cycle (this point isdiscussed further below). Aurora kinase B (AKB) localizes tobundles of specialized MTs termed K-fibers and helps con-nect the kinetochore, a protein structure that assembles onthe centromere of chromosomes, to spindle fibers duringprometaphase and metaphase. It then relocalizes to themidzone or central spindle at the metaphase–anaphasetransition, influencing chromosome separation and cyto-kinesis. Like AKB, Aurora kinase C (AKC) is a chromosomepassenger protein that localizes first to the inner centro-mere, then to the central spindle, andfinally to themidbodyofmitotic cells as the cell cycle progresses (10). Aswith AKA,the mRNA and protein expressions of AKB and AKC aremaximally elevated during the G2/M-phase of the cell cycleand decrease rapidly as the cells enter G1 (10).

PLK1–4 localize to the centrosomeduring interphase andprophase, move to the spindle poles after prophase, andrelocate to the midbody for telophase and cytokinesis (11).

Like the mitotic kinases, KSP is mitosis-specific and thushas attracted attention as a target for anticancer agentstargeting mitosis (12). KSP is a homotetrameric kinesinmotor protein and a member of the kinesin superfamily ofproteins that binds and hydrolyzes ATP, coupling the ener-gy released from hydrolysis with force production thatallows for unidirectional movement along MTs. Unliketransport kinesins, which function in cytosolic movement

Figure 1. Organization of MTsduring the cell cycle and theexpression of proteins involvedin mitosis. During the G1-, S-,and G2-phases, MTs areassembled in parallel, polarizedarrays with their plus (þ) endpointing outward from the cellcenter. The minus (�) end of thearrays is nucleated (anchoredand initiated) by the centrosome(also known as the MT-organizing center). Enteringmitosis, the centrosomes moveto the 2 opposite sides of thecell, forming the mitotic spindleapparatus that separateschromosomes during celldivision. Theexpression levels ofspecialized proteins in mitosis,such as AKA (red), AKB (yellow),and PLK (green), increase ascells traverse the G2 portion ofthe cell cycle and reach theirmaximum level during mitosis.

CCRFOCUS

Clin Cancer Res; 18(1) January 1, 2012 Clinical Cancer Research52

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 3: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

and the localization of organelles and vesicles, mitotickinesins such as KSP participate in the assembly, mainte-nance, and elongation of the mitotic spindle, chromosomealignment and segregation, and MT depolymerization.KSP’s antiparallel MT-sliding function is required for sep-

aration of the 2 centrosomes in early mitosis, an essentialevent for bipolar spindle formation. Consequently, inhibi-tion of KSP results in mitotic arrest because the failure ofcentrosomal separation results in anMT arraywith only onepole.

Figure 2. Localization of AKA,AKB, and PLK during mitosis.During mitosis, AKA (red)translocates from the centrosometo the mitotic poles and theadjacent spindle MTs, AKB(yellow) localizes to MTs near thekinetochores, and PLK (green)moves from the centrosome to thespindle poles. By telophase andcytokinesis, all 3 kinases relocateto the midbody.

Tales of How Great Drugs Were Brought Down by a Flawed Rationale

www.aacrjournals.org Clin Cancer Res; 18(1) January 1, 2012 53

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 4: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

Numerous studies have shown that AK, PLK, and KSP areexpressed primarily in theM-phase of the cell cycle and, to alesser extent, in G2 (8). During G1-, G0-, and S-phases,expression either does not occur or occurs only at very lowlevels. Consequently, drugs aimed at these proteins willonly find their target and have an effect on cells that arerapidly cycling (dividing) and likely to be in or passingthrough the G2- or M-phase while the drug is present. Thishighly restricted, cycle-specific expression likely explainswhy these agents have failed to affect tumor growth inpatients.

What happens when a cell is exposed to an inhibitorthat targets mitosis?

Inhibitors of mitotic proteins all cause disruption ofnormal mitotic function, as do MTAs, which additionallydisrupt interphase functions. The mechanism wherebymitosis is disrupted differs among these agents. MTAs andinhibitors of KSP and PLK interfere with proper assembly ofthemitotic spindle and lead to arrest inmid-mitosis (4, 13).Inhibitors of AKA cause transient arrest followed by mitoticexit with misaligned chromosomes, and inhibitors of AKBcause premature mitotic exit with major defects in chromo-some attachment (13). Although the mechanistic detailsdiffer, all of these compounds, including MTAs, disrupt thenormal process of mitosis, but of course, they only do sowhen mitosis occurs. None of these agents produces orpromotes the initiation of mitosis, and hence will onlycause damage to cells when mitosis happens to occur. Ifmitosis occurs rarely, agents whose action is restricted to

mitosis will rarely cause damage. However, MTs are presentthroughout the cell cycle, andMTarrays aremost vulnerableduring mitosis due to the increased rate of MT turnover.Hence, MTAs potently target mitosis when it occurs, butthey can also target interphase cells.

Duration of mitotic arrestStudies involving a number of cell types and a variety of

methods, including the use of mitosis inhibitors, showedthat mitotic arrest can only be sustained for �1–2 days inhuman cells with constant drug exposure, and less than thatin rodent cells (14). Although 1–2 days is a significantduration, given that the normal duration of mitosis is only1–2 hours inmany different cell types and tissues, it is by nomeans indefinite (15). Mitotic arrest causes many differenttypes of stress. For example, condensed chromosomes can-not be transcribed, which makes it impossible to replenishneeded transcripts—a state that cannot be sustained. Con-sequently, this period is limited by mitotic slippage, espe-cially in normal cells, or cell death, especially in cancer cells(14). A further complication is that cells display significantintra- and interline variability in the duration of mitoticarrest, and great variation among different drugs and themode of exit from arrest (16). The latter presumably hap-pens when the slow degradation of cyclin B1, which con-tinues during arrest, is reduced below the level that isrequired to maintain the mitotic state. At this point somecells die; some exit mitosis (termed mitotic slippage),remain tetraploid, and possibly die later; and some showother fates (reviewed in ref. 17). Whatever the details, the

Figure 3. Binding partners ofAKA, AKB, and PLK duringmitosis. After phosphorylationby Lats2, Ajuba interacts withAKA. AKA phosphorylatesBRCA1 (101), Cdc25b (102),CENP-A (103), p53 (104), Eg5(105), TACC3 (106), and Tpx2(107). PLK phosphorylatesAPC/C (108), cyclin B (109),NudC (110), and cohesinsubunit SCC1 (111). AKBphosphorylates INCENP (112),Dam1 (113), CENP-A (114),histone 3 (115), Ncd80 (116),myosin II (117), Nlp (118), desmin(119), andseptin (120), andbindsto survivin (121) and vimentin(122). APC/C, anaphase-promoting complex/cyclosome;CENPA, centromere protein A;Eg5, kinesin-like protein; H3,histone 3; Nlp, ninein-likeprotein; NudC, nucleardistribution gene C.

CCRFOCUS

Clin Cancer Res; 18(1) January 1, 2012 Clinical Cancer Research54

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 5: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

data show that mitotic arrest is not maintained for anyextendedperiod. Furthermore, it is unclear howany of theseresults would change with pulsed or varying drug concen-trations. Very few data are available regarding mitotic dura-tion in patients. In one study, mitotic counts that wereobtained before and after treatment with an MTA (pacli-taxel) showed great variability from patient to patient in theextent and duration of mitotic arrest (18).

Doubling times of human tumorsCancer is often mistakenly thought of as a mass of

abnormal cells growing rapidly in an uncontrolledmanner.This is misleading for at least 2 reasons. First, the data showthat cell division is a very precisely regulated process thatfollows similar pathways in both normal and cancer cellsand involves highly specialized proteins. Second, onemightthink that frequent cell division is a hallmark of tumor cells.This misconception is encouraged by the rapid rate of celldivision of cancer cells observed in vitro and in xenograftmodels. Although it can be challenging to measure tumordoubling times in humans, studies have shown that tumorsdonot double as rapidly as onemight think. As summarizedin Table 1, data obtained by a variety of radiologic imagingmodalities show median doubling times for many humantumors of >100 days, which is much longer than thedoubling times observed in preclinical animal models(2). This means that at any one time, only a very smallpercentage of tumor cells are undergoing mitosis. Indeed,the meanmitotic index in a variety of tumor types has beenshown to be <1% (19, 20). Additionally, the mean labelingindex measured by radiolabeled imaging [e.g., tritiatedthymidine ([3H]-TdR)] was only a few percent of the tumor(21). Together, these data show that in contrast to in vitroand xenograft models, human tumors have very long dou-bling times. This makes such tumors indifferent to drugsthat target cell proliferation or proteins whose expression ishighly restricted to one phase of the cell cycle.

Preclinical in vivo dataTable 2 and Fig. 4 show preclinical in vivo data obtained

for mitotic kinase and KSP inhibitors, and they provideinsight into why this class of agents has not been successfulclinically. Also shown in Table 2 are the doubling timesof the preclinical models used in various studies (see

also Table 1). As noted above, this rate of doubling ismarkedly faster than that found in any solid tumor inhumans, and this difference may be significant for targetgenes/proteins whose expression is highly restricted duringthe cell cycle, such as occurs with mitosis-specific proteins.Furthermore, in most models the agents were administeredfrequently (often daily and even twice daily). Frequentadministration is essential for agents that target proteinswhose expression is restricted to a very short part of the cellcycle (22). The importance of frequent drug administrationis underscored by the results depicted in Fig. 4, which showa decline in activity when drug administration was changedfrom twice per week to once per week. Finally, we note, assummarized in Table 2, that complete responses wereachieved inonly 2 of the 53models evaluated, underscoringthe fact that despite a frequent schedule of administration torapidly growing tumors, complete tumor disappearancecould not be achieved. From these data, it is clear that inthe models with rapid doubling times, cells that are not inG2/M-phase are refractory to therapy and can repopulate therapidly dividing tumormass unless frequent administrationis used (23).

Clinical dataFew (if any) agents have shown as much activity as

paclitaxel did during its development, in terms of bothbreadth and magnitude. A review in 1995 of early evalua-tions of the drug’s activity showed single-agent responserates of 17% to 62%, 20% to 48%, and 21% to 41% inbreast, ovarian, and lung cancers, respectively (24). Activitywas subsequently reported in other cancers, includingKaposi sarcoma and esophageal, urothelial, and head andneck cancers. In our own tabulation of 29 studies of single-agent paclitaxel in these seven cancers, we recordedresponse rates ranging from 7% to 71%, with a median of37% and an overall response rate of 28%, in a group of2,271 patients (data not shown; see references in ref. 25).With this as background, we summarize in Table 3 andSupplementary Tables S1A and S1B the clinical results of 46studies conductedwith20different agents targeting theAKs,PLK, and KSP. The range and median values for all patientgroups are presented at the bottomof each table. The overallresponse rate of 1.6% for all studies shows unequivocallythat these agents lack activity against solid tumors [787

Table 1. Tumor doubling time in days: a comparison of preclinical in vivo models and patient data

Preclinical models Patient data

Mean Median Reference Mean Median Reference

Breast cancer 5.6 4.5 (34) 152 137 (35–46)Colon cancer 3.4 3.4 (34) 391 334 (47–51)Lung cancer 4.4 3.8 (34) 114 100 (52–60)Prostate cancer 3.4 3.4 (34) 219 126 (61)Melanoma 5.4 5.7 (34) 147 78 (62–67)

Tales of How Great Drugs Were Brought Down by a Flawed Rationale

www.aacrjournals.org Clin Cancer Res; 18(1) January 1, 2012 55

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 6: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

Table 2. Mitotic kinase inhibitors in xenograft models

Drug Cell lineTumororigin

In vivoDT,daysa Hostb

Treatmentschedule(dose peradministration)

Tumorreduction(comparedwith) Reference

AK inhibitorsMK-0457/VX-680 HL-60 AML 3.3 Nude i.p., bid � 13d (75 mg/kg) 98% (C) (68)

MIA PaCa-2 Pancreas — Nude i.p., bid (50 mg/kg) 22% (ITS)HCT-116 Colon 2.6 Nude rats i.v., 3d/w (1 mg/kg/h) 56% (ITS)

AZD1152(barasertib)

SW-620 Colon 2.4 AT s.c., 48-h infusion(150 mg/kg/d)

87% (C) (69)

Colo 205 Colon 4.3 AT s.c., 48-h infusion(150 mg/kg/d)

94% (C)

HCT116 Colon 2.6 AT s.c., 48-h infusion(150 mg/kg/d)

74% (C)

A549 Lung 8.4 AT s.c., 48-h infusion(150 mg/kg/d)

69% (C)

Calu-6 Lung — AT s.c., 48-h infusion(150 mg/kg/d)

55% (C)

HL-60 AML 3.3 AT s.c., 48-h infusion(150 mg/kg/d)

>100% (C)

PHA739358(danusertib)

Huh-7 Liver — NOD/SCID i.p., bid (30 mg/kg/d) 75% (C) (70)HepG2 Liver — NOD/SCID i.p., bid (30 mg/kg/d) 88% (C)

MLN8054 HCT-116 Colon 2.6 Nude p.o., bid � 21d (30 mg/kg) 81% (C) (71)MLN8237 MM1.S MM — SCID p.o., bid � 21d (40mg/kg) 80% (C) (72)R763 MIA PaCa-2 Pancreas — SCID p.o. 3d/w � 4w (15 mg/kg) 25% (C) (73)AT9283 HCT-116 Colon 2.6 AT nu/nu q9d � 3 27% (C) (74)SNS-314 A2780 Ovarian nu/nu i.p., qw � 3w 57% (C) (75)

nu/nu i.p., biw � 3w (170 mg/kg) 54% (C)A375 Melanoma — nu/nu i.p., qw � 3w (170 mg/kg) 35% (C)

nu/nu i.p., biw � 3w (170 mg/kg) 65% (C)H1299 Lung — nu/nu i.p., qw � 3w (170 mg/kg) 18% (C)

nu/nu i.p., biw � 3w (170 mg/kg) 69% (C)MDA-MB-231 Breast 4.4 nu/nu i.p., qw � 3w (170 mg/kg) 49% (C)

nu/nu i.p., biw � 3w (170 mg/kg) 74% (C)PC-3 Prostate 2.4 nu/nu i.p., qw � 3w (170 mg/kg) 56% (C)

nu/nu i.p., biw � 3w (170 mg/kg) 68% (C)Calu-6 Lung — nu/nu i.p., qw � 3w (170 mg/kg) 35% (C)

nu/nu i.p., biw � 3w (170 mg/kg) 91% (C)ENMD-2076 HCT-116 Colon 2.6 Nude or SCID p.o., bid (200 mg/kg) 89% (C) (76)

HT-29 Colon 5.1 Nude or SCID p.o., qd (100 mg/kg) 62% (C)CT-26 Mouse colon — Nude or SCID p.o., qd (100 mg/kg) 21% (C)A375 Melanoma — Nude or SCID p.o., qd (151 mg/kg) 81% (C)MDA-MB-231 Breast 4.4 Nude or SCID p.o., qd (100 mg/kg) 94% (C)H929 MM — Nude or SCID p.o., qd (150 mg/kg) 88% (C)OPM-2 MM — Nude or SCID p.o., qd (75 mg/kg) 56% (C)MV4 AML — Nude or SCID p.o., bid (50 mg/kg) 99% (C)HL-60 AML 3.3 Nude or SCID p.o., qd (150 mg/kg) 83% (C)

PLK inhibitorsBI2536 Caki-1 Kidney 2.1 nu/nu i.v., qd � 2d, 5d off

(50 mg/kg)Minor effect (77)

SN12C Kidney 5.6 nu/nu i.v., qd � 2d, 5d off(50 mg/kg)

Minor effect

(Continued on following page)

CCRFOCUS

Clin Cancer Res; 18(1) January 1, 2012 Clinical Cancer Research56

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 7: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

(AKs, Table 3) þ 345 (PLK, Table S1A) þ 267 (KSP, TableS1B) ¼ 1,399 patients total; objective response rate: 22/1399¼ 1.6%]. Such a response rate might even be recordedwith a placebo (26, 27). Some may wonder whether lowbioavailability can explain why these drugs were inactive inpatients. We think this unlikely given that poor results wereseen even in cases where adequate biomarkers for mitoticarrest (mitotic index or histone H3 phosphorylation mostoften in skin biopsies) showed target engagement (28, 29).Moreover, wewould argue that the neutropenia observed inmany of the treated patients represents a biomarker orsurrogate that clearly proves the drug hit the target andinhibited mitosis (even if it was not in the tumor). Inaddition, we emphasize our belief that a stable disease rateof �20% can only be interpreted as a measure of theinherent biology of the tumors being treated. As reviewedabove,mitotic arrest canonly be sustained for�1–2days, soany assumption that this class of agents (or any other) canarrest cells in mitosis for any period that might be scoredclinically as stable disease is not scientifically defensible.Similarly, given the restricted expression of the targets in

mitosis, only a small percentage of cells would provevulnerable to inhibitors targetingmitosis. This would argueagainst a situation inwhich the fraction of killed cells equalsthe quantity that has been newly added to the tumor bydivision.

Not surprisingly, we note that in the subset of patientswith hematologic malignancies, the response rate washigher, but still low, at 8.2%. Given that hematologiccancers often have faster doubling times than solid tumors,one could envision a potential role for agents that targetmitosis in these malignancies. However, the neutropeniathat will invariably occur might limit treatment such thatboth the amount and frequency of drug administrationwould be inadequate.

Finally, although infrequent responseshavebeenobservedclinically, occasionally an off-target effect inhibiting a kinaseother than a mitotic kinase has resulted in activity. Forexample, activity has been reported with an AK inhibitor,tozasertib (MK-0457/VX680), in patients with imatinib-resistant chronic myeloid leukemia (CML) harboring theT315Imutation (30).However, these effects are independent

Table 2. Mitotic kinase inhibitors in xenograft models (Cont'd )

Drug Cell lineTumororigin

In vivoDT,daysa Hostb

Treatmentschedule(dose peradministration)

Tumorreduction(comparedwith) Reference

786–0 Kidney 6.7 nu/nu i.v., qd � 2d, 5d off(50 mg/kg)

No effect

nu/nu i.t., qd � 2d, 5d off(50 mg/kg)

No effect

nu/nu i.t., bid � 2d, 5d off(100 mg/kg)

Significantregression

A498 Kidney 3.4 nu/nu i.v., qd � 2 d, 5d off(50 mg/kg)

No effect

nu/nu i.t., bid � 2d, 5d off(100 mg/kg)

Significantregression

HMN-214 PC-3 Prostate 2.4 Nude p.o., qd � 28d(20 mg/kg)

79% (C) (78)

WiDr Colon — Nude p.o., qd � 28d(20 mg/kg)

73% (C)

A549 Lung 8.4 Nude p.o., qd � 28d(20 mg/kg)

75% (C)

KSP inhibitorsIspinesib(SB-715992)

MCF-7 Breast 4.5 nu/nu i.p., q4d � 3 (10 mg/kg) 92% (C) (79)

HCC-1954 Breast — nu/nu i.p., q4d � 3 (10 mg/kg) 95% (C)KLP4 Breast — nu/nu i.p., q4d � 3 (10 mg/kg) 94% (C)BT-474 Breast NA SCID i.p., q4d � 3 (8 mg/kg) 61% (C)MDA-MB-468 Breast — SCID i.p., q4d � 3 (8 mg/kg) 100% (C)

Abbreviations: AT, athymic; bid, twice per day; biw, twice per week; C, control; d, day; DT, doubling time; i.p., intraperitoneal; i.t.,intratumoral; ITS, initial tumor size; MM, multiple myeloma; NOD, nonobese diabetic; nu, nude; p.o., orally; SCID, severe combinedimmunodeficient; qw, once per week; qd, once per day; w, week.aPlowman et al. (34).bMouse, unless otherwise indicated.

Tales of How Great Drugs Were Brought Down by a Flawed Rationale

www.aacrjournals.org Clin Cancer Res; 18(1) January 1, 2012 57

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 8: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

of the cell cycle anddonot contradict the reasonsgivenhereinto explain why these drugs have failed to benefit the over-whelming majority of patients with solid tumors.

Conclusions

Mitotic kinase inhibitors were developed as nonneuro-toxic alternatives to MTAs; however, the rationale behindthe development of these agents, i.e., the thesis that MTAskill cells inhuman tumors onlyby inhibitingmitosis,meantthat if unrecognized mechanisms proved to be important,such agents would fail when they reached the clinic. In an invitro or a xenograft model with a doubling time of a fewdays, a large fraction of cells will prove vulnerable to atherapy that targets a protein that is crucial for mitosis.Therefore, it is not surprising that in a patient with a tumordoubling time of�30–60 days and an S-phase fraction of atmost a few percent, only a small, insignificant fraction ofcells will be vulnerable to a drug aimed at a target that isexpressed only transiently in most tumor (and normal)cells. Inhibitors that target mitosis could prove effective inrapidly growing (i.e., rapidly dividing) leukemias and lym-phomas (i.e., Burkitt lymphoma), and their value in suchmalignancies should be further explored. However, theirlack of activity against a majority of cancers highlightsseveral aspects of cancer drug development in the 21stcentury and informs our understanding of the mechanismsof drug action, as summarized below.

First, the dose-limiting toxicity of mitotic kinase inhibi-tors—reversible neutropenia—stands as a testament to theprowess of pharmaceutical drug developers, especially thechemists who synthesized these agents, and the biologists

who identified and characterized the targets and developedthe models used in their validation. Although the majorityof human tumors do not divide rapidly enough to besusceptible to these mitotic poisons, the same cannot besaid of vulnerable marrow elements. The doubling time ofgranulocyte precursors is very short [17 hours for myelo-blasts, 63 hours for promyelocytes, and 55 hours for mye-locytes (31)]. Thus, reversible neutropenia would beexpected of an agent targeting a mitotic kinase or KSP,because at any one time �25% of marrow neutrophils areundergoing mitosis. Indeed, we would argue that the neu-tropenia observed nearly uniformly in clinical trials withagents that inhibit mitosis can be viewed as a biomarker orsurrogate of activity.

Second, although this pharmaceutical prowess has pos-itive attributes, it led to theoverdevelopment of drugs aimedat targets that have not yet been validated (e.g., AKs, PLKs,and KSP). To date, clinical data have been reported for atleast 20 different mitosis-specific poisons, and there arelikely more yet to be reported. This has involved a consid-erable financial outlay and patient recruitment effort with-out a return on the investment (the response rate to aninhibitor targeting mitosis in clinical trials involving 1,399patients with solid tumors was 1.6%; see Fig. 4 and Sup-plementary Tables S1A and S1B). Although a certainamount of redundancy represents attempts to design abetter agent, the clinical evidence and the nearly uniformmyelosuppression suggest that these drugs were remarkablysimilar and potent.

Third, the results indicate yet again the deficiencies ofthe preclinical models used in drug development.Although these preclinical models are far from ideal (butat some level valuable), it is clear that the rapid doublingtimes in such models compared with human tumorsallow for an accelerated drug-development timeline.However, this precludes their use when the doublingtime is a critical factor in a drug’s activity. The rapiddoubling times of the preclinical models explains whyagents targeting mitosis proved active in these models butwere ineffective against patient tumors.

Fourth, the results highlight the increasing use/abuse of"stable disease" as a measure of drug efficacy. As discussedabove, scoring stable disease as a measure of activity cannotbe defended scientifically for this class of compounds,arguing against its use as a measure of drug activity. Asnoted above, studies have shown that inhibitors targetingmitosis cannot be expected to stabilize tumor growth byarresting cells in mitosis for prolonged periods of time.Indeed, one can persuasively argue that these studies havecollectively defined �20% as the stable disease rate dueto the inherent tumor biology of patients harboringadvanced solid tumors who enroll in similar phase I/IItrials, because in effect, these patients received a drugthat had no disease-stabilizing activity. We note that thisestimate of 20% as the stable disease rate due to inherenttumor biology is not excessive, because stable disease ratesof 55% to 67% have been obtained with placebos in renalcell and hepatocellular carcinomas (26, 27). Finally, the

© 2012 American Association for Cancer Research

0

A2780

A375

H1299

MDA-M

B-231

PC-3

Calu-6

20

SNS-314

% t

umo

r re

duc

tion

com

par

ed t

o c

ont

rol

Once weeklyTwice weekly

40

60

80

100

Figure 4. Frequent administration of SNS-314 leads to greater antitumoreffect. SNS-314, a selective inhibitor of AKA, AKB, and AKC, wasadministered once or twice a week in mouse xenograft models. Acomparison with the control tumor size shows that the twice-weeklyschedule resulted in significantly greater tumor reduction than the once-weekly treatment schedule. Data from Arbitrario et al. (75).

CCRFOCUS

Clin Cancer Res; 18(1) January 1, 2012 Clinical Cancer Research58

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 9: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

Table 3. AK inhibitors in clinical trials

Drug (company) Disease TreatmentPatients,N

Cyclesmedian(range) Response DLT/AE Reference

MK-0457/VX-680(Vertex/Merck)

CML/ALL 5-d CIV q2–3 wk 3 ND 1 PHR 1 CHR #BM (30)ST 5-d CIV q28d 16 2 (1–6) 3 SD #ANC, F/N, allergic reaction (80)ST 24-h CIV q21d 27 Total: 8 1 SD #ANC,N/V/diarrhea, fatigue (81)

AZD1152/barasertib(AstraZeneca)

AML 7-d CIV q21d 16 Total: 28 2 CR, 1 PR,6 SD

#ANC, F/N, #WBC, #Plt,fatigue, pneumonia,ARDS, sepsis

(82)

PHA 739358/danusertib(Nerviano)

ST 6-h CIV 42 ND 7 SD #ANC, HTN, fatigue,anorexia, N/diarrhea

(83)

ST 24-h CIV q14d �G-CSF

56 3 (1–20) 1 PR; 18 SD,4 PSD

F/N, fatigue, anorexia, N/V/D, mucositis, "LFTs, #Kþ,HTN, fever

(84)

ST 3-wk CIV q28d 50 2 (1–28) 14 SD #ANC, #WBC, #Hgb N/diarrhea anorexia, fatigue

(85)

MLN8054(Millenium)

ST p.o. d1–5 þ d8–12q28d or p.o. QIDd1–14 q28d

43 1 (1–10) 3 SD #MS, "LFTs, mucositis (86)

ST p.o. � 7d q21d; p.o.� 14d q28d; p.o.�21d q35d

61 2 (1–14) 9 SD #MS, N/V, confusion,cognitive disorder,hallucination, fatigue

(87)

MLN8237(Millenium)

ST p.o. � 7d q21d; p.o.� 14d q28d; p.o.�21d q35d

65 ND 1 PR, 8 SD #ANC, #Plt, sepsis,mucositis, N/D, fatigue,alopecia

(28)

NHM p.o. bid � 7d q21d 17 2 (1–8) ND F/N, #ANC, fatigue (88)R763 (Merck/Serono)

ST p.o. d1, 8 q21d; p.o.d1–3 q21d

15 ND 2 PD ND (89)

AT9283 (Astex) Leuk 72h CIV q3wk 29 ND 2 PHR; 1 PCR #ANC, #BM, "LFTs;alopecia

(90)

ST 72h CIV q3wk 22 2 (2–7) 3 SD F/N (91)ST 72h CIV q3wk 33 2 (1–12) 1 PR, 4 SD F/N, GI, fatigue (92)

SNS-314 (Sunesis) ST 3-h CIV d1, 8and 15 q28d

32 2 6 SD N/V, fatigue, constipation,pain

(93)

SU6668 (Sugen/Pfizer)

ST p.o. bid, 28d 35 ND 4 SD #Plt, pericarditis, pleuritis,fatigue

(94)

ST p.o. bid, 28d 19 ND 3 SD GI, fatigue, pleuritis, SOB,pericardial effusion

(95)

ENMD-2076(EntreMed)

ST p.o. qd, 28d 14 3 (< 1–9) 4 SD HTN, fatigue, proteinuria,diarrhea

(96)

ST p.o. qd � 21d,q28d

67 3 (1–24) 2 PR, 49 SD #ANC, HTN, N/V/D, fatigue,"LFTs

(97)

OvCa p.o. 64 2 3 PR, 27 SD #BM, fatigue, HTN,mucositis, "LFTs, HFS,thromboembolic event,subarachnoidhemorrhage, #LVfunction, PRES

(98)

BI 811283(BoehringerIngelheim)

ST 24h CIV q21d 57 Mean ¼ 3 33% SD #ANC, F/N, #WBC (99)

ST 24h CIV q2wk 52 Mean ¼ 3 29% SD #ANC, #WBC (100)Tumor type No. of patients No. of treatments:

range/medianNo. of responders(percentage of all)

(Continued on following page)

Tales of How Great Drugs Were Brought Down by a Flawed Rationale

www.aacrjournals.org Clin Cancer Res; 18(1) January 1, 2012 59

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 10: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

disappointing clinical results ratify the paradigm thatMTAs do not kill cancer cells in humans primarily byinhibiting mitosis, even though both MTAs and inhibitorsthat target mitosis are lethal to bone marrow elements inthis way. We previously proposed the inhibition of traffick-ing on MTs as the principal mechanism of action of MTAs(2).One such example forwhich clear evidencenowexists isthe androgen receptor, which plays a crucial role in thegrowth of prostate cancer (32, 33). The evidence that MTdisruption affects androgen-receptor trafficking helpsexplain the activity of both docetaxel and cabazitaxel in adisease that is often so indolent that questions about the

need for therapy remain unresolved. Therefore, when study-ing cells in which an MTA or a combination of MTAs isactive, one should seek to determine which of the criticalproteins that traffic on MTs have been affected by thetherapy in question.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Received July 28, 2011; revised October 19, 2011; accepted November 7,2011; published online January 3, 2012.

References1. Harrison MR, Holen KD, Liu G. Beyond taxanes: a review of novel

agents that target mitotic tubulin and microtubules, kinases, andkinesins. Clin Adv Hematol Oncol 2009;7:54–64.

2. Komlodi-Pasztor E, Sackett D, Wilkerson J, Fojo T. Mitosis is not akey target ofmicrotubule agents in patient tumors. NatRevClinOncol2011;8:244–50.

3. Taylor S, Peters JM. Polo and Aurora kinases: lessons derived fromchemical biology. Curr Opin Cell Biol 2008;20:77–84.

4. Sarli V, Giannis A. Targeting the kinesin spindle protein: basic prin-ciples and clinical implications. Clin Cancer Res 2008;14:7583–7.

5. Brinkley BR. Microtubule organizing centers. Annu Rev Cell Biol1985;1:145–72.

6. Bergen LG, Kuriyama R, Borisy GG. Polarity of microtubules nucle-ated by centrosomes and chromosomes of Chinese hamster ovarycells in vitro. J Cell Biol 1980;84:151–9.

7. Terada S, Hirokawa N. Moving on to the cargo problem of microtu-bule-dependent motors in neurons. Curr Opin Neurobiol 2000;10:566–73.

8. Jackson JR, Patrick DR, Dar MM, Huang PS. Targeted anti-mitotictherapies: can we improve on tubulin agents? Nat Rev Cancer2007;7:107–17.

9. Dutertre S, Descamps S, Prigent C. On the role of Aurora-A incentrosome function. Oncogene 2002;21:6175–83.

10. Sasai K, Katayama H, Stenoien DL, Fujii S, Honda R, Kimura M, et al.Aurora-C kinase is a novel chromosomal passenger protein that cancomplement Aurora-B kinase function in mitotic cells. Cell MotilCytoskeleton 2004;59:249–63.

11. Kishi K, van Vugt MA, Okamoto K, Hayashi Y, Yaffe MB. Functionaldynamics of Polo-like kinase 1 at the centrosome. Mol Cell Biol2009;29:3134–50.

12. Huszar D, TheoclitouME, Skolnik J, Herbst R. Kinesinmotor proteinsas targets for cancer therapy. Cancer Metastasis Rev 2009;28:197–208.

13. Lens SM, Voest EE, Medema RH. Shared and separate functions ofpolo-like kinases and aurora kinases in cancer. Nat Rev Cancer2010;10:825–41.

14. Rieder CL. Mitosis in vertebrates: the G2/M and M/A transitionsand their associated checkpoints. Chromosome Res 2011;19:291–306.

15. Baserga R. The relationship of the cell cycle to tumor growthand control of cell division: a review. Cancer Res 1965;25:581–95.

Table 3. AK inhibitors in clinical trials (Cont'd )

Drug (company) Disease TreatmentPatients,N

Cyclesmedian(range) Response DLT/AE Reference

Solid tumors 787 1–30/2 Partial/transient response:10 (1.2%)

Stable disease:203 (25.8%)

Hematologic malignancies 48 ND Partial/tumor response:8 (16.7%)

Stable disease:6 (12.5%)

Abbreviations:#ANC,decreasedabsoluteneutrophil count (neutropenia); ARDS, acute respiratory distresssyndrome; #BM,decreasedbonemarrow (pancytopenia); CHR, complete hematologic response; CIV, continuous i.v. infusion; CML/ALL, T315I abl-mutated CMLor (Ph)þ ALL; cycles, median number of cycles administered; DLT/AE, dose-limiting toxicity/adverse event; F/N, febrile neutropenia;G-CSF, granulocyte colony-stimulating factor; GI, gastrointestinal disturbance; HFS, hand-foot syndrome; #Hgb, anemia; #Kþ,hypokalemia; HTN, hypertension; Leuk, refractory leukemia; "LFTs, elevated liver function tests; #MS, reduced mental status/somnolence; NHM, nonhematologic malignancies; N/V, nausea/vomiting; OvCa, platinum-resistant ovarian cancer; PCR, partialcytogenetic response; PHR, partial hematologic response; #Plt, thrombocytopenia; p.o., orally; PR, partial response; PRES, reversibleposterior leukoencephalopathy syndrome; SD, stable disease; SOB, shortness of breath; ST, advanced solid tumors or advanced andrefractory solid tumors or refractory solid tumors; #WBC, decreased white blood cell count (leukopenia).

CCRFOCUS

Clin Cancer Res; 18(1) January 1, 2012 Clinical Cancer Research60

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 11: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

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. GascoigneKE, Taylor SS.Howdo anti-mitotic drugs kill cancer cells?J Cell Sci 2009;122:2579–85.

18. SymmansWF, VolmMD,Shapiro RL, Perkins AB, KimAY,Demaria S,et al. Paclitaxel-induced apoptosis and mitotic arrest assessed byserialfine-needle aspiration: implications for early prediction of breastcancer response to neoadjuvant treatment. Clin Cancer Res2000;6:4610–7.

19. Weidner N, Moore DH 2nd, Vartanian R. Correlation of Ki-67 antigenexpression with mitotic figure index and tumor grade in breastcarcinomas using the novel "paraffin"-reactive MIB1 antibody. HumPathol 1994;25:337–42.

20. Szende B, Romics I, Minik K, Szab�o J, Torda I, Lov�asz S, et al.Repeated biopsies in evaluation of therapeutic effects in prostatecarcinoma. Prostate 2001;49:93–100.

21. Livingston RB, Ambus U, George SL, Freireich EJ, Hart JS. In vitrodetermination of thymidine-3H labeling index in human solid tumors.Cancer Res 1974;34:1376–80.

22. Crosio C, Fimia GM, Loury R, Kimura M, Okano Y, Zhou H, et al.Mitotic phosphorylation of histone H3: spatio-temporal regulationby mammalian Aurora kinases. Mol Cell Biol 2002;22:874–85.

23. Steegmaier M, HoffmannM, Baum A, L�en�art P, Petronczki M, Krss�akM, et al. BI 2536, a potent and selective inhibitor of polo-like kinase 1,inhibits tumor growth in vivo. Curr Biol 2007;17:316–22.

24. Rowinsky EK, Donehower RC. Paclitaxel (taxol). N Engl J Med1995;332:1004–14.

25. Drugs.com [homepage on the Internet]. Paclitaxel. Available from:http://www.drugs.com/monograph/paclitaxel.html#r24.

26. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M,et al. TARGET Study Group. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007;356:125–34.

27. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al.SHARP Investigators Study Group. Sorafenib in advanced hepato-cellular carcinoma. N Engl J Med 2008;359:378–90.

28. Dees EC, Infante JR, Burris HA, Astsaturov IA, Stinchcombe T, Liu H,et al. Phase I study of the investigational drugMLN8237, an Aurora Akinase (AAK) inhibitor, in patients (pts) with solid tumors. J Clin Oncol28:15s, 2010 (suppl; abstr 3010).

29. Kristeleit R, Calvert H, Arkenau H, Olmos D, Adam J, Plummer ER,et al. A phase I study of AT9283, an aurora kinase inhibitor, in patientswith refractory solid tumors. J Clin Oncol 27:15s, 2009 (suppl; abstr2566).

30. Giles FJ, Cortes J, JonesD, BergstromD, KantarjianH, FreedmanSJ.MK-0457, a novel kinase inhibitor, is active in patients with chronicmyeloid leukemia or acute lymphocytic leukemiawith the T315I BCR-ABL mutation. Blood 2007;109:500–2.

31. Boll IT, Fuchs G. A kinetic model of granulocytopoiesis. Exp Cell Res1970;61:147–52.

32. DarshanMS, LoftusMS, Thadani-MuleroM, LevyBP, EscuinD, ZhouXK, et al. Taxane-induced blockade to nuclear accumulation of theandrogen receptor predicts clinical responses in metastatic prostatecancer. Cancer Res 2011;71:6019–29.

33. Zivi A, Massard C, De-Bono J. Changing therapeutic paradigms incastrate-resistant prostate cancer. Clin Genitourin Cancer 2010;8:17–22.

34. Plowman J, Dykes DJ, HollingsheadM, Simpson-Herren L, AlleyMC.Human tumor xenograft models in NCI drug development. In: TeicherBA, editor. Anticancer drug development guide, preclinical screen-ing, clinical trials, and approval. Totowa, NJ: Humana Press; 1997.p. 101–25.

35. Conant EF, Dillon RL, Palazzo J, Ehrlich SM, Feig SA. Imagingfindings in mucin-containing carcinomas of the breast: correlationwith pathologic features. AJR Am J Roentgenol 1994;163:821–4.

36. Peer PG, van Dijck JA, Hendriks JH, Holland R, Verbeek AL. Age-dependent growth rate of primary breast cancer. Cancer 1993;71:3547–51.

37. Arnerl€ovC, EmdinSO, LundgrenB,RoosG,S€oderstr€omJ,Bjersing L,et al. Breast carcinoma growth rate described by mammographic

doubling time and S-phase fraction. Correlations to clinical andhistopathologic factors in a screened population. Cancer 1992;70:1928–34.

38. Kuroishi T, Tominaga S, Morimoto T, Tashiro H, Itoh S, WatanabeH, et al. Tumor growth rate and prognosis of breast cancer mainlydetected by mass screening. Jpn J Cancer Res 1990;81:454–62.

39. Semiglazov VF, Moiseenko VM, Chernomordikova MF. [Growthkinetics of breast cancer]. Vopr Onkol 1989;35:288–93.

40. Spratt JS, Greenberg RA, Heuser LS. Geometry, growth rates, andduration of cancer and carcinoma in situ of the breast before detec-tion by screening. Cancer Res 1986;46:970–4.

41. KoscielnyS, TubianaM,ValleronAJ. Asimulationmodel of the naturalhistory of human breast cancer. Br J Cancer 1985;52:515–24.

42. von Fournier D, Weber E, Hoeffken W, Bauer M, Kubli F, Barth V.Growth rate of 147mammary carcinomas.Cancer 1980;45:2198–207.

43. Heuser L, Spratt JS, Polk HC Jr. Growth rates of primary breastcancers. Cancer 1979;43:1888–94.

44. Spratt JS Jr, KaltenbachML, Spratt JA.Cytokinetic definition of acuteand chronic breast cancer. Cancer Res 1977;37:226–30.

45. Lundgren B. Observations on growth rate of breast carcinomas andits possible implications for lead time. Cancer 1977;40:1722–5.

46. Tilanus-Linthorst MM, Kriege M, Boetes C, Hop WC, Obdeijn IM,Oosterwijk JC, et al. Hereditary breast cancer growth rates and itsimpact on screening policy. Eur J Cancer 2005;41:1610–7.

47. Umetani N, Masaki T, Watanabe T, Sasaki S, Matsuda K, Muto T.Retrospective radiographic analysis of nonpedunculated colorectalcarcinomas with special reference to tumor doubling time and mor-phological change. Am J Gastroenterol 2000;95:1794–9.

48. Matsui T, Yao T, Iwashita A. Natural history of early colorectal cancer.World J Surg 2000;24:1022–8.

49. Finlay IG, Meek D, Brunton F, McArdle CS. Growth rate of hepaticmetastases in colorectal carcinoma. Br J Surg 1988;75:641–4.

50. TadaM,Misaki F, Kawai K. Growth rates of colorectal carcinoma andadenoma by roentgenologic follow-up observations. GastroenterolJpn 1984;19:550–5.

51. Bolin S, Nilsson E, Sj€odahl R. Carcinoma of the colon and rectum—

growth rate. Ann Surg 1983;198:151–8.52. Quint LE, Cheng J, Schipper M, Chang AC, Kalemkerian G. Lung

lesion doubling times: values and variability based on method ofvolume determination. Clin Radiol 2008;63:41–8.

53. El Sharouni SY, Kal HB, Battermann JJ. Accelerated regrowth of non-small-cell lung tumours after induction chemotherapy. Br J Cancer2003;89:2184–9.

54. Winer-Muram HT, Jennings SG, Tarver RD, Aisen AM, Tann M,Conces DJ, et al. Volumetric growth rate of stage I lung cancer priorto treatment: serial CT scanning. Radiology 2002;223:798–805.

55. Sato G, Saijo Y, Uchiyama B, Kumano N, Sugawara S, Fujimura S,et al. Prognostic value of nucleolar protein p120 in patients withresected lung adenocarcinoma. J Clin Oncol 1999;17:2721–7.

56. Arai T, Kuroishi T, Saito Y, Kurita Y, Naruke T, Kaneko M JapaneseLung Cancer Screening Research Group. Tumor doubling time andprognosis in lung cancer patients: evaluation from chest films andclinical follow-up study. Jpn J Clin Oncol 1994;24:199–204.

57. Usuda K, Saito Y, Sagawa M, Sato M, Kanma K, Takahashi S, et al.Tumor doubling time and prognostic assessment of patients withprimary lung cancer. Cancer 1994;74:2239–44.

58. Filderman AE, Shaw C, Matthay RA. Lung cancer. Part I: Etiology,pathology, natural history, manifestations, and diagnostic techni-ques. Invest Radiol 1986;21:80–90.

59. Geddes DM. The natural history of lung cancer: a review based onrates of tumour growth. Br J Dis Chest 1979;73:1–17.

60. Fujimura S, Suda S, Yamauchi A, Sato H, Sohara Y, Kondo T, et al.Tumor doubling time and PPD skin test reactivity in resectable lungcancer. J Jpn Lung Cancer Soc 1979;19:135–42.

61. Berges RR, Vukanovic J, Epstein JI, CarMichel M, Cisek L, JohnsonDE, et al. Implication of cell kinetic changes during the progression ofhuman prostatic cancer. Clin Cancer Res 1995;1:473–80.

62. Lee JH, Gulec SA, Kyshtoobayeva A, SimMS, Morton DL. Biologicalfactors, tumor growth kinetics, and survival after metastasectomy forpulmonary melanoma. Ann Surg Oncol 2009;16:2834–9.

Tales of How Great Drugs Were Brought Down by a Flawed Rationale

www.aacrjournals.org Clin Cancer Res; 18(1) January 1, 2012 61

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 12: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

63. Carlson JA. Tumor doubling time of cutaneous melanoma and itsmetastasis. Am J Dermatopathol 2003;25:291–9.

64. Eskelin S, Pyrh€onen S, Summanen P, Hahka-KemppinenM, Kivel€a T.Tumor doubling times inmetastaticmalignantmelanoma of the uvea:tumor progression before and after treatment. Ophthalmology2000;107:1443–9.

65. Char DH, Kroll S, Phillips TL. Uveal melanoma. Growth rate andprognosis. Arch Ophthalmol 1997;115:1014–8.

66. Sch€otterl HD, Paul E. [Growth dynamics of malignant melanomas.Determination of tumor doubling time based on morphometric mea-surements of roentgen images of lung metastases]. Z Hautkr1988;63:481–7.

67. Augsburger JJ, Gonder JR, Amsel J, Shields JA, Donoso LA. Growthrates and doubling times of posterior uveal melanomas. Ophthal-mology 1984;91:1709–15.

68. Harrington EA, BebbingtonD,Moore J, RasmussenRK, Ajose-Adeo-gun AO, Nakayama T, et al. VX-680, a potent and selective small-molecule inhibitor of the Aurora kinases, suppresses tumor growth invivo. Nat Med 2004;10:262–7.

69. Wilkinson RW, Odedra R, Heaton SP,Wedge SR, Keen NJ, Crafter C,et al. AZD1152, a selective inhibitor of AuroraBkinase, inhibits humantumor xenograft growth by inducing apoptosis. Clin Cancer Res2007;13:3682–8.

70. Benten D, Keller G, Quaas A, Schrader J, Gontarewicz A, BalabanovS, et al. Aurora kinase inhibitor PHA-739358 suppresses growth ofhepatocellular carcinoma in vitro and in a xenograft mouse model.Neoplasia 2009;11:934–44.

71. Huck JJ, Zhang M, McDonald A, Bowman D, Hoar KM, Stringer B,et al. MLN8054, an inhibitor of Aurora A kinase, induces senescencein human tumor cells both in vitro and in vivo. Mol Cancer Res2010;8:373–84.

72. G€org€un G, Calabrese E, Hideshima T, Ecsedy J, Perrone G, Mani M,et al. A novel Aurora-A kinase inhibitorMLN8237 induces cytotoxicityand cell-cycle arrest in multiple myeloma. Blood 2010;115:5202–13.

73. McLaughlin J, Markovtsov V, Li H, Wong S, Gelman M, Zhu Y, et al.Preclinical characterization of Aurora kinase inhibitor R763/AS703569 identified through an image-based phenotypic screen.J Cancer Res Clin Oncol 2010;136:99–113.

74. Curry J, Angove H, Fazal L, Lyons J, Reule M, Thompson N, et al.Aurora B kinase inhibition in mitosis: strategies for optimising theuse of aurora kinase inhibitors such as AT9283. Cell Cycle2009;8:1921–9.

75. Arbitrario JP, Belmont BJ, Evanchik MJ, Flanagan WM, Fucini RV,Hansen SK, et al. SNS-314, a pan-Aurora kinase inhibitor, showspotent anti-tumor activity and dosing flexibility in vivo. Cancer Che-mother Pharmacol 2010;65:707–17.

76. Fletcher GC, Brokx RD, Denny TA, Hembrough TA, Plum SM, FoglerWE, et al. ENMD-2076 is an orally active kinase inhibitor with anti-angiogenic and antiproliferative mechanisms of action. Mol CancerTher 2011;10:126–37.

77. Ding Y, Huang D, Zhang Z, Smith J, Petillo D, Looyenga BD, et al.Combined gene expression profiling and RNAi screening in clear cellrenal cell carcinoma identify PLK1 and other therapeutic kinasetargets. Cancer Res 2011;71:5225–34.

78. Takagi M, Honmura T, Watanabe S, Yamaguchi R, Nogawa M,Nishimura I, et al. In vivo antitumor activity of a novel sulfonamide,HMN-214, against human tumor xenografts inmice and the spectrumof cytotoxicity of its active metabolite, HMN-176. Invest New Drugs2003;21:387–99.

79. Purcell JW, Davis J, Reddy M, Martin S, Samayoa K, Vo H, et al.Activity of the kinesin spindle protein inhibitor ispinesib (SB-715992)in models of breast cancer. Clin Cancer Res 2010;16:566–76.

80. Rubin EH,ShapiroGI, SteinMN,WatsonP,BergstromD, XiaoA, et al.A phase I clinical and pharmacokinetic (PK) trial of the aurora kinase(AK) inhibitor MK-0457 in cancer patients. J Clin Oncol 24:18s, 2006(suppl; abstr 3009).

81. Traynor AM, Hewitt M, Liu G, Flaherty KT, Clark J, FreedmanSJ, et al.Phase I dose escalation study of MK-0457, a novel Aurora kinaseinhibitor, in adult patients with advanced solid tumors. Cancer Che-mother Pharmacol 2011;67:305–14.

82. Tsuboi K, Yokozawa T, Sakura T,Watanabe T, Fujisawa S, YamauchiT, et al. A Phase I study to assess the safety, pharmacokinetics andefficacy of barasertib (AZD1152), an Aurora B kinase inhibitor, inJapanese patients with advanced acute myeloid leukemia. Leuk Res2011;35:1384–9.

83. De Jonge M, Steeghs N, Verweij J, Nortier JW, Eskens F, OuwerkerkJ, et al. Phase I study of the aurora kinases (AKs) inhibitor PHA-739358 administered as a 6 and 3-h IV infusion on days 1, 8, 15 every4 wks in patients with advanced solid tumors. J Clin Oncol 26: 2008(May 20 suppl; abstr 3507).

84. Cohen RB, Jones SF, Aggarwal C, von Mehren M, Cheng J,Spigel DR, et al. A phase I dose-escalation study of danusertib(PHA-739358) administered as a 24-hour infusion with and with-out granulocyte colony-stimulating factor in a 14-day cycle inpatients with advanced solid tumors. Clin Cancer Res 2009;15:6694–701.

85. Steeghs N, Eskens FA, Gelderblom H, Verweij J, Nortier JW, Ouwer-kerk J, et al. Phase I pharmacokinetic and pharmacodynamic study ofthe aurora kinase inhibitor danusertib in patients with advanced ormetastatic solid tumors. J Clin Oncol 2009;27:5094–101.

86. Macarulla T, Cervantes A, Elez E, Rodr��guez-Braun E, Baselga J,Rosell�oS, et al. Phase I study of the selective Aurora Akinase inhibitorMLN8054 in patients with advanced solid tumors: safety, pharma-cokinetics, and pharmacodynamics. Mol Cancer Ther 2010;9:2844–52.

87. Dees EC, Infante JR, Cohen RB, O'Neil BH, Jones S, von Mehren M,et al. Phase 1 study of MLN8054, a selective inhibitor of Aurora Akinase in patients with advanced solid tumors. Cancer ChemotherPharmacol 2011;67:945–54.

88. Sharma S, Kurzrock R, Gouw L, Hong DS, Jones K, Zhou X, et al.Phase I dose-escalation study of the investigational Aurora A kinase(AAK) inhibitor MLN8237 as an enteric-coated tablet (ECT) formula-tion in patients with nonhematologic malignancies. J Clin Oncol 29:2011 (suppl; abstr 3094).

89. Renshaw JS, Patnaik A, Gordon M, Beeram M, Fischer D, Gianella-Borradori A, et al. A phase I twoarm trial of AS703569 (R763), an orallyavailable aurora kinase inhibitor, in subjects with solid tumors: pre-liminary results. J Clin Oncol 25:18s, 2007 (suppl; abstr 14130).

90. Foran JM, Ravandi F, O'Brien SM, Borthakur G, Rios M, Boone P,et al. Phase I and pharmacodynamic trial of AT9283, an aurora kinaseinhibitor, in patients with refractory leukemia. J Clin Oncol 26: 2008(May 20 suppl; abstr 2518).

91. Plummer ER, Calvert H, Arkenau H, Mallett KH, Squires M, Smith D,et al. A dose-escalation and pharmacodynamic study of AT9283 inpatients with refractory solid tumours. J Clin Oncol 26: 2008 (May 20suppl; abstr 2519).

92. Kristeleit R, Calvert H, Arkenau H, Olmos D, Adam J, Plummer ER,et al. A phase I study of AT9283, an aurora kinase inhibitor, in patientswith refractory solid tumors. J Clin Oncol 27:15s, 2009 (suppl; abstr2566).

93. Robert F, Verschraegen CF, Hurwitz H, Uronis H, Advani R, Chen A,et al. A phase I trial of sns-314, a novel and selective pan-aurorakinase inhibitor, in advanced solid tumor patients. J Clin Oncol27:15s, 2009 (suppl; abstr 2536).

94. BrittenCD,Rosen LS, Kabbinavar F, RosenP,MulayM,Hernandez L,et al. Phase I trial of SU6668, a small molecule receptor tyrosinekinase inhibitor, given twice daily in patients with advanced cancers.Proc Am Soc Clin Oncol 2002;21:1922.

95. Brahmer JR,KelseyS,Scigalla P,Hill G,BelloC,Elza-BrownK, et al. Aphase I study of SU6668 in patients with refractory solid tumors. ProcAm Soc Clin Oncol 21: 2002 (abstr 335).

96. Bastos BR, Diamond J, Hansen R, Gustafson D, Arnott J, Bray M,et al. An open-label, dose escalation, safety, and pharmacokineticstudy of ENMD-2076 administered orally to patients with advancedcancer. J Clin Oncol 27:15s, 2009 (suppl; abstr 3520).

97. Diamond JR, Bastos BR, Hansen RJ, Gustafson DL, Eckhardt SG,Kwak EL, et al. Phase I safety, pharmacokinetic, and pharmacody-namic study of ENMD-2076, a novel angiogenic and Aurora kinaseinhibitor, in patients with advanced solid tumors. Clin Cancer Res2011;17:849–60.

CCRFOCUS

Clin Cancer Res; 18(1) January 1, 2012 Clinical Cancer Research62

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 13: Inhibitors Targeting Mitosis: Tales of How Great Drugs ...transport kinesins, which function in cytosolic movement Figure 1. Organization of MTs during the cell cycle and the expression

98. Matulonis U, Tew WP, Matei D, Behbakht K, Fleming GF, Oza AM,et al. A phase II study of ENMD-2076 in platinum-resistant ovariancancer. J Clin Oncol 29: 2011 (suppl; abstr 5021).

99. Mross KB, ScheulenME, Frost A, Scharr D, Richly H, Nokay B, et al. Aphase I dose-escalation study of BI 811283, an Aurora B inhibitor,administered every three weeks in patients with advanced solidtumors. J Clin Oncol 28:15s, 2010 (suppl; abstr 3011).

100. ScheulenME,Mross KB, Richly H, Nokay B, Frost A, Scharr D, et al. Aphase I dose-escalation study of BI 811283, an Aurora B inhibitor,administered days 1 and 15, every four weeks in patients withadvanced solid tumors. J Clin Oncol 28: 2010 (suppl; abstr e13065).

101. Ouchi M, Fujiuchi N, Sasai K, Katayama H, Minamishima YA, Ongu-saha PP, et al. BRCA1 phosphorylation by Aurora-A in the regulationof G2 to M transition. J Biol Chem 2004;279:19643–8.

102. Dutertre S, Cazales M, Quaranta M, Froment C, Trabut V, Dozier C,et al. Phosphorylation of CDC25B by Aurora-A at the centrosomecontributes to the G2-M transition. J Cell Sci 2004;117:2523–31.

103. KunitokuN, SasayamaT,Marumoto T, ZhangD,HondaS, KobayashiO, et al. CENP-Aphosphorylation byAurora-A in prophase is requiredfor enrichment of Aurora-B at inner centromeres and for kinetochorefunction. Dev Cell 2003;5:853–64.

104. Liu Q, Kaneko S, Yang L, Feldman RI, Nicosia SV, Chen J, et al.Aurora-A abrogation of p53 DNA binding and transactivationactivity by phosphorylation of serine 215. J Biol Chem 2004;279:52175–82.

105. Giet R, Uzbekov R, Cubizolles F, Le Guellec K, Prigent C. TheXenopus laevis aurora-related protein kinase pEg2 associates withand phosphorylates the kinesin-related protein XlEg5. J Biol Chem1999;274:15005–13.

106. Kinoshita K, Noetzel TL, Pelletier L, Mechtler K, Drechsel DN, Schwa-gerA, et al. Aurora Aphosphorylation of TACC3/maskin is required forcentrosome-dependent microtubule assembly in mitosis. J Cell Biol2005;170:1047–55.

107. Giet R, Petretti C, Prigent C. Aurora kinases, aneuploidy andcancer, acoincidence or a real link? Trends Cell Biol 2005;15:241–50.

108. Golan A, Yudkovsky Y, Hershko A. The cyclin-ubiquitin ligase activityof cyclosome/APC is jointly activated by protein kinases Cdk1-cyclinB and Plk. J Biol Chem 2002;277:15552–7.

109. Toyoshima-Morimoto F, Taniguchi E, Shinya N, Iwamatsu A, NishidaE. Polo-like kinase 1 phosphorylates cyclin B1 and targets it to thenucleus during prophase. Nature 2001;410:215–20.

110. Zhou T, Aumais JP, Liu X, Yu-Lee LY, Erikson RL. A role for Plk1phosphorylation of NudC in cytokinesis. Dev Cell 2003;5:127–38.

111. Sumara I, Vorlaufer E, StukenbergPT,KelmO,RedemannN,NiggEA,et al. The dissociation of cohesin from chromosomes in prophase isregulated by Polo-like kinase. Mol Cell 2002;9:515–25.

112. Bishop JD, Schumacher JM. Phosphorylation of the carboxylterminus of inner centromere protein (INCENP) by the Aurora BKinase stimulates Aurora B kinase activity. J Biol Chem 2002;277:27577–80.

113. Cheeseman IM, Anderson S, JwaM,Green EM,Kang J, Yates JR3rd,et al. Phospho-regulation of kinetochore-microtubule attachmentsby the Aurora kinase Ipl1p. Cell 2002;111:163–72.

114. Zeitlin SG, Shelby RD, Sullivan KF. CENP-A is phosphorylated byAurora B kinase and plays an unexpected role in completion ofcytokinesis. J Cell Biol 2001;155:1147–57.

115. Goto H, Yasui Y, Nigg EA, Inagaki M. Aurora-B phosphorylatesHistone H3 at serine28 with regard to the mitotic chromosomecondensation. Genes Cells 2002;7:11–7.

116. Akiyoshi B, Nelson CR, Ranish JA, Biggins S. Analysis of Ipl1-mediated phosphorylation of the Ndc80 kinetochore protein in Sac-charomyces cerevisiae. Genetics 2009;183:1591–5.

117. Murata-Hori M, Fumoto K, Fukuta Y, Iwasaki T, Kikuchi A, TatsukaM,et al.Myosin II regulatory light chain as a novel substrate for AIM-1, anaurora/Ipl1p-related kinase from rat. J Biochem 2000;128:903–7.

118. Yan J, Jin S, Li J, Zhan Q. Aurora B interaction of centrosomal Nlpregulates cytokinesis. J Biol Chem 2010;285:40230–9.

119. Kawajiri A, Yasui Y, Goto H, TatsukaM, Takahashi M, Nagata K, et al.Functional significanceof the specific sites phosphorylated in desminat cleavage furrow: Aurora-Bmay phosphorylate and regulate type IIIintermediate filaments during cytokinesis coordinatedly with Rho-kinase. Mol Biol Cell 2003;14:1489–500.

120. Qi M, Yu W, Liu S, Jia H, Tang L, Shen M, et al. Septin1, a newinteraction partner for human serine/threonine kinase aurora-B. Bio-chem Biophys Res Commun 2005;336:994–1000.

121. Bolton MA, Lan W, Powers SE, McCleland ML, Kuang J, StukenbergPT. AuroraBkinase exists in a complexwith survivin and INCENPandits kinase activity is stimulated by survivin binding and phosphory-lation. Mol Biol Cell 2002;13:3064–77.

122. Goto H, Yasui Y, Kawajiri A, Nigg EA, Terada Y, Tatsuka M, et al.Aurora-B regulates the cleavage furrow-specific vimentin phosphor-ylation in the cytokinetic process. J Biol Chem 2003;278:8526–30.

www.aacrjournals.org Clin Cancer Res; 18(1) January 1, 2012 63

Tales of How Great Drugs Were Brought Down by a Flawed Rationale

Cancer Research. by guest on August 27, 2020. Copyright 2012 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from