Review

Aurora kinase A inhibitors: promising agents in antitumoral therapy 1.

Aurora A: more than a mitotic kinase

2.



Aurora A is a cancer-associated gene

3.

Consequences of Aurora A depletion/inhibition in vitro and in vivo

4.

Preclinical and clinical studies using Aurora inhibitors

5.

Conclusions

6.

Expert opinion

Marcos Malumbres & Ignacio Perez de Castro† Centro Nacional de Investigaciones Oncolo´gicas (CNIO), Cell Division and Cancer Group, Madrid, Spain

Introduction: Aurora proteins are serine/threonine kinases with critical functions during mitosis. Aurora A, one of the members of this family, participates in crucial processes including mitotic entry, DNA damage checkpoint recovery and centrosome and spindle maturation. Aurora A is frequently overexpressed in human cancers and, when inhibited, impairs cell proliferation. Areas covered: Here, we review the preclinical studies that support the use of Aurora A inhibitors in antitumoral strategies. We also discuss past or current clinical trials using Aurora A inhibitors in multiple tumor types. We pay special attention to Alisertib, a potent and selective Aurora A inhibitor currently in Phase III. Expert opinion: The potential of Aurora A inhibitors in the treatment of cancer depends on many factors, mainly related with the molecular status of tumor cells. Yet, we still need to find proper biomarkers to select those patients that better react to Aurora A inhibitors. Furthermore, their effect could significantly improve when used in combination with other drugs. Although some clinical trials are already testing the cooperative effect of different antitumoral drugs, additional preclinical studies are necessary to establish the best combinations. Here, we discuss some possibilities that could be explored in future studies. Keywords: Alisertib, Aurora A, cancer, clinical trials, kinase inhibitors, mitosis, MLN8237 Expert Opin. Ther. Targets (2014) 18(12):1377-1393

1.

Aurora A: more than a mitotic kinase

Among the different stages of the cell cycle, mitosis has attracted significant attention, not only because many mitotic regulators have been linked to tumorigenesis [1] but also because the success of microtubule (MT) poisons in cancer therapy [2,3]. Aurora family kinases are of special relevance during mitosis because they play essential roles in centrosome maturation and chromosome segregation [4]. The increasing trend of scientific publications (> 1600), patents (> 400) and clinical trials (> 90) on Aurora kinases during the last 16 years (Figure 1) demonstrates the importance of this family of mitotic kinases. The founding member of the Aurora family, Ipl1 (Increase in ploidy 1), was described in 1993 in a screen for Saccharomyces cerevisiae mutants that failed to undergo normal chromosome segregation [5]. Two Ipl1-like proteins (Aurora kinases A and B) have been found in Drosophila melanogaster, Caenorhabditis elegans and Xenopus laevis. Three members of this family of serine-threonine kinases, Aurora A, B and C are encoded in humans by the genes AURKA, AURKB and AURKC. Although they are highly conserved at the protein level, Aurora kinases have very distinct expression and localization patterns as well as functions [4]. Aurora A is upregulated at G2 and localizes to the centrosomes during interphase and to both spindle poles and spindle MTs during early mitosis. As expected by its expression and subcellular localization, Aurora 10.1517/14728222.2014.956085 © 2014 Informa UK, Ltd. ISSN 1472-8222, e-ISSN 1744-7631 All rights reserved: reproduction in whole or in part not permitted

1377

rez de Castro M. Malumbres & I. Pe

Article highlights. . . .

.

Aurora kinase A is a key mitotic protein frequently amplified and over-expressed in human cancers. Aurora kinase inhibitors have shown promising results in clinical trials. A total of forty clinical trials have been initiated to test Alisertib in monotherapy or in combination with other drugs. The identification of good biomarkers and effective combination therapies will improve the clinical use of Aurora inhibitors.

This box summarizes key points contained in the article.

Publications Patents

No. of publications/patents/trials

260

Clinical trials

200 140 80 80 60 40 20 20 10

13

11

20

09

20

07

20

05

20

03

20

01

20

20

19

99

0

Year

Figure 1. Number of scientific publications, clinical trials and patents related to Aurora kinases. Data were obtained from 1998 to 2013 using ‘Aurora kinase’ as the searching text. P u b M e d ( h t t p : / / w w w. n c b i . n l m . n i h . g o v / p u b m e d ? cmd=search), from the US National Library of Medicine, the U. National Cancer Institute and EU Clinical Trials Register web pages (http://www.clinicaltrials.gov and https://www.clinicaltrialsregister.eu/ctr-search/search) and PATENTSCOPE (http://patentscope.wipo.int/search/en/ search.jsf), from the World International Property Organization, were used as searching tools.

A regulates mitotic entry, centrosome maturation and spindle formation. Aurora B is expressed in proliferating cells during G2 and mitosis, shows a chromosomal-passenger localization (centromeric in early mitosis, central spindle in anaphase and mid-body during cytokinesis) and plays critical roles in chromosome condensation and cohesion, chromosome biorientation and cytokinesis. The importance of their functions make Auroras A and B required for mouse embryo development [6-9]. Aurora C is mainly expressed in testis, has 1378

a localization pattern similar to Aurora B and is required for spermatogenesis and the first divisions of mouse embryogenesis [9,10]. Aurora kinase A (AURKA) has many aliases, including serine/threonine kinase 15, serine/threonine kinase 6, breast tumor amplified kinase, Aurora-related kinase 1, Homo sapiens Aurora/IPL1-related kinase, Eg2 and Ipl- and Aurora-related kinase 1. Originally described in a screening for mutations that affect the centrosome cycle in Drosophila [11], Aurora A has been mainly associated with cell cycle regulation, specifically in the G2/M phases (Figure 2). The regulation of Aurora A activity is complex and is mainly associated with the phosphorylation of a threonine residue within the activation loop (Thr288 in human Aurora A) [12,13] and its proteolytic degradation. Although different molecules might function as AURKA-positive regulators (Figure 2) [14-22], the best-known activator is TPX2 [23]. Binding to TPX2 induces a conformational change that promotes autophosphorylation and prevents the action of its inhibitor, the phosphatase PP1 [24]. Recently, it has been shown in vitro that Aurora A autophosphorylation occurs in a long-lived dimer and that binding to TPX2 induces the activation of Aurora A molecules un-phosphorylated at Thr288 [25]. Aurora A is a target of the E3-ubiquitin ligase anaphase-promoting complex/ cyclosome, which in conjunction with its specificity cofactor Cdh1 targets this protein for destruction during mitotic exit [26]. Other molecules negatively regulate AURKA activity by dephosphorylation, SUMOylation or induction of its degradation (Figure 2) [27-34]. Several functions have also been reported for Aurora A in addition to its typical mitotic roles (Figure 2). Aurora A is essential for DNA damage-induced checkpoint recovery [35,36]. Aurora A has been involved in the regulation of p53 because it phosphorylates p53 at Ser315, leading to its Mdm2-mediated ubiquitination and subsequent proteolysis [37]. Another important non-mitotic function of Aurora A is the regulation of translation of RNAs that contain cytoplasmic polyadenylation elements at their 3¢ untranslated region, which has been shown in the regulation of meiosis [38,39]. Aurora A also modulates the function of alphaCamKII synthesis at synapses [40], calcium signaling in nonproliferative cells [41], Akt [42] and the cell cycle regulators Myc, cyclin B1 and Cdk1 [43,44]. Different reports have pointed out a role of Aurora A in development and pluripotency through the regulation of cell polarity and p53 activity. More specifically, Aurora A regulates cell polarity by determining mitotic spindle orientation through the Par complex [45-47] and p53 by inhibiting its transcriptional activity in mouse embryonic stem cells through direct phosphorylation at Ser212 [48]. Aurora A is also a major regulator of Nanog activity in epithelial tissues [49]. Other studies have pointed out to possible roles of Aurora A in angiogenesis [50], inflammation [51] and pro-oncogenic signaling pathways [42]. Finally, Aurora A has also been connected to the regulation of neurite elongation [52], cilia disassembly [22], cell motility [53], DNA replication [54] and NF-kB signaling [55].

Expert Opin. Ther. Targets (2014) 18(12)

Aurora kinase A inhibitors

Regulators PP1 / PP6 APCCdh1 P53 GADD45A

GSK3β FBXW7 USP2A AIP

TPX2 CDK1 AJUBA BORA CEP192 PUM2

SUMO1 HEF1 PAK1 MYC NPM CaM

Aurora A

Mitotic functions • Mitotic entry (BRCA1, PLK1, CDC25B) • Centrosome function (NDEL1, CENTRIN, LATS2, CENPA, CENPE, CEP192, SKI, LIMK1/2)

Non-mitotic functions • DNA damage/repair (PLK1, BRCA1) • DNA replication (GEMININ) • P53/P73 activity • RNA Poly-adenylation (CPEB) • Pluripotency (P53) • Cell polarity (PAR3, PAR6) • Cilium stability (HDAC6)

• Spindle dynamics

• NF-κB signaling (IκBα)

(KIF2A, TACC3, MCAK, HURP, TPX2, EG5, P150GLUED)

• Calcium signaling (PC2) • Cell migration/motility (RAL)

Figure 2. Regulation and cellular functions of Aurora A. Negative and positive modulators of AURKA activity are shown in red and blue, respectively. Effectors for the AURKA-dependent mitotic and non-mitotic functions are indicated. AURKA: Aurora kinase A.

2.

Aurora A is a cancer-associated gene

Aurora A is amplified and/or overexpressed in primary tumors of the breast, ovary, colon, pancreas, prostate, as well as in neuroblastoma among others (Table 1). High levels of Aurora A are associated with poor prognosis (Table 1) and Aurora A overexpression may also promote resistance to gefitinib, taxol and cisplatin in cancer cells [56-58]. Different Aurora A mutations are associated with cancer. The most frequently reported mutations consists on a single-nucleotide polymorphism T91A (Phe31Ile) that was originally proposed as a low-penetrance susceptibility allele in multiple human cancers [59]. However, a massive study performed with > 32,000 patients discarded the association between the T91A polymorphism and breast cancer [60]. Different studies demonstrated a determinant role of Aurora A in tumor development. First, overexpression of Aurora-A-induced malignant transformation in focus formation and xenograft assays [61,62], and elevated levels of Aurora A cooperated with oncogenic RAS in the

transformation of mouse fibroblasts [63]. Furthermore, transgenic mouse models for the overexpression of Aurora A showed hyperplasia in mammary glands and tumor development [64,65]. Importantly, Aurora A haploinsufficient mice are also prone to spontaneous tumor development [7], which points out a tumor suppressor role of this kinase. This antitumoral property of Aurora A has also been shown in Drosophila [66]. Therefore, both the overexpression and the downregulation of Aurora A are causally related with tumorigenesis. What are the mechanisms by which an abnormal expression of Aurora A triggers the malignant phenotype? Among the different ideas proposed during the last years, two hypotheses can be highlighted. The first one is based on the induction of chromosome instability and the generation of aneuploidy, which in turn, have been recognized as tumor drivers. The aneuploid phenotype induced by Aurora A abnormal expression has been associated to centrosome amplification, mitotic abnormalities and cytokinesis failure [58,62] and Aurora A has been included in the top-70 list of genes of the cancer-associated

Expert Opin. Ther. Targets (2014) 18(12)

1379

rez de Castro M. Malumbres & I. Pe

Table 1. Aurora A amplification/over-expression in human cancers. Tumor type

Amplification/overexpression*

Bad prognosis

Breast Colorectal Gastric Head and neck Leukemias, lymphomas Lung Neuronal Ovarian and endometrial Pancreatic Prostate Skin Urothelial and bladder

DNA, mRNA and protein DNA, mRNA and protein DNA, mRNA and protein mRNA and protein mRNA and protein mRNA and protein DNA, mRNA and protein DNA, mRNA and protein DNA, mRNA and protein mRNA and protein Protein mRNA and protein

Yes Yes Yes Yes Yes Yes Yes Yes n.r. Yes n.r. Yes

Ref. [147-150] [61,62,151,152] [153,154] [155-157] [20,158-160] [161-163] [119,164-166] [167-171] [172,173] [174,175] [176] [177-179]

*Amplification: DNA; Overexpression: mRNA and or protein. n.r.: Not reported.

Table 2. Main phenotypes observed in cells upon Aurora A depletion or inhibition. Phenotype

Inactivation mechanism

Organism/system

Ref.

Pupal lethality; mitotic arrest, monopolar spindle Lack of centrosome maturation, embryo lethality Delayed mitotic entry, lack of bipolar spindle Embryo lethality; lack of proliferation; mitotic defects; DNA damage; apoptosis; senescence

Genetic inactive mutants

D. melanogaster

[11,180]

RNAi and lack of function mutants mAb

C. elegans

[77,79,181]

X. laevis

[76,78,182]

RNAi, specific inhibitors, conditional KO

Mammalian cell lines; mouse embryos/tissues

[6-8,16,80-82,113,183],

RNAi: RNA interference.

chromosome instability signature [67]. The second possibility is based in the Aurora A-p53 crosstalk described during the last years [33,37]. Interestingly, induction of aneuploidy by Aurora A is p53-dependent [68,69], which connects the two hypotheses described above. In summary: i) Aurora A overexpression induces p53 degradation; ii) loss of p53 eliminates the checkpoints that arrest the polyploid and aneuploid cells generated upon Aurora A overexpression; and iii) the resulting cells are prone to become transformed. The mitotic regulator TPX2, which binds and stabilizes Aurora A [70], is also overexpressed in cancer, and the possible relevance of the Aurora A-TPX2 holoenzyme in cancer has been previously reviewed [71,72]. Both mitotic and nonmitotic, Aurora A partners could play a role in the AuroraA-induced malignant phenotype. The fact that Aurora A is mainly located at the nucleus of non-tumoral, interphasic cells, whereas overexpressed, tumor-associated Aurora A can also be found at cytoplasm [73], makes more complicated the characterization of the mechanisms by which Aurora A participates in tumorigenesis. So far, the relevance of Aurora A in the regulation of other major oncogenic partners such as Akt, Ras or Myc [44,57,74,75] is not well established, despite 1380

the interest of these interactions in the identification of possible biomarkers of clinical use.

Consequences of Aurora A depletion/ inhibition in vitro and in vivo

3.

Interfering with Aurora A expression or activity by siRNA expression, immunodepletion or specific inhibitors induces mitotic alterations that impair cell cycle progression (Table 2). The first functional assays demonstrated that its disruption in D. melanogaster, C. elegans and X. laevis causes defects in mitotic entry, centrosome maturation and spindle formation [11,76-79]. Similar results were obtained in mammalian cell lines when Aurora A expression was reduced using RNA interference technology [16,80,81]. Monopolar spindles, prometaphase arrest, promotion of tetraploid cells are the most commonly features cited in these works. Recently, three different groups have shown that Aurora A is necessary for mouse embryo development [6-8]. Specifically, its genetic ablation results in lethality at the morula stage mainly due to spindle assembly defects, pronounced cell proliferation failure and mitotic arrest. Aurora A depletion in adult tissues

Expert Opin. Ther. Targets (2014) 18(12)

Aurora kinase A inhibitors

A.

AurkaΔ/Δ

Aurka+/+ I

I

II

II

B.

C.

III

III

IV

IV

Aurka+/+

AurkaΔ/Δ

Aurka+/+

AurkaΔ/Δ

D. Aurka+/+

AurkaΔ/Δ

Figure 3. Cellular defects induced by Aurora A depletion. A. Upper panels show representative pictures of the mitotic progression (I to IV) in wild type (Aurka+/+) and Aurora A null (AurkaD/D) mouse fibroblasts that express EGFP-tagged Histone H2B. Arrows indicate abnormal chromosome segregation and DNA bridges. Mitotic abnormalities are also common in Aurora A null tissues. Lower panels show esophagous of wild type and Aurora A defective mice. Normal anaphase in the wild type tissue and abnormal monopolar spindles in Aurora A null cells are highlighted. B. The defects in chromosome segregation lead, with time, to the accumulation of giant nuclei, micronuclei and lobulated nuclei. Pictures are mouse fibroblasts stained with DAPI to visualize DNA. C. Senescence induced by Aurora A depletion. Images show representative cases for mouse fibroblasts (upper panels) and mouse splenocytes (lower panels). D. Aurora A null proliferative tissues show severe defects. Images are representative of wild type and Aurora A null mouse skin. Aurora A null mice show a thinner characterized by hair follicles at the catagen--telogen stage.

is characterized by a significant increase in mitotic abnormalities and DNA damage markers and with the presence of aneuploid cells, eventually resulting in impaired proliferation and senescence (Figure 3) [82]. Conditional deletion of Aurora A in adult tissues induces premature aging and prevents tumor growth in vivo. In normal or tumor cells, lack or inhibition of Aurora A results in defective chromosome segregation and the generation of tetraploid or aneuploid cells, which is subsequently accompanied by a stress response characterized by DNA damage and the induction of antiproliferative proteins such as p53 and p21Cip1. As a final consequence, cells arrest and enter into apoptosis or senescence.

Preclinical and clinical studies using Aurora inhibitors

4.

A significant number of compounds and procedures related with the use of Aurora kinase inhibitors have been patented during the last decade (Table S1). Different reviews summarizing the information about Aurora kinase inhibitors have been published in the last five years [83-86]. The number of clinical trials performed with Aurora kinase inhibitors has kept constant, and the results published from these preclinical and clinical studies have significantly increased (Figure 1).

Expert Opin. Ther. Targets (2014) 18(12)

1381

rez de Castro M. Malumbres & I. Pe

Pan or A/B 32%

A. AURKA 53%

AURKB 15%

B. Aurora A

Number of clinical trials

20 15

Phase I Phase II Phase III Aurora B

10 Pan or dual A/B 4 3 2 1

D

To

sa

ze

rti

b

(V

X-

K 68 W an 0/ -24 M 4 us K- 9 er 04 tib (P X 57) H L A- 22 73 8 93 AS 70 A 58 35 T9 ) 69 28 /R 3 -7 C YC 63 -1 1 PF SN 6 S -0 -3 38 14 14 AM 73 5 M E GK- NM 90 51 D 0 08 -2 /V 07 Al X 6 is er M -68 tib L 9 (M N-8 LN 05 -8 4 BI 237 81 ) 1 AZ 28 3 G SK D11 10 52 70 91 6

0

Figure 4. Clinical trials testing agents with anti-Aurora A activity. A. Chart shows the percentage of trials for Pan-Aurora kinase or Aurora kinase A/B dual inhibitors, Aurora kinase B selective inhibitors and Aurora-kinase--A-specific inhibitors. B. Bar graph shows the number of clinical trials started for up to 17 drugs with Pan-Aurora kinase activity (10), Aurora A selectivity (4) and specific for Aurora kinase B (3).

Most Aurora kinase inhibitors are small molecule compounds designed to bind to the ATP-binding pocket in a competitive and reversible manner. Around 10 pan-Aurora inhibitors without a clear preference for a single family member have been reported in clinical trials or in preclinical stages for the treatment of cancer (Supplementary Table S2 [184-191]). Although Aurora-B-specific compounds or Pan-Aurora inhibitors have been used in 15 and 34% of the clinical trials testing Aurora kinase inhibitors, most clinical trials (53%) have focused to Aurora A selective agents (Figure 4). In the next sections, we will describe the preclinical and clinical studies reported for the four developed AURKAspecific inhibitors used in clinical trials (Table 3). ENMD-2076 Originally developed by Miikana in 2006, this EntreMed product is the L(+) tartrate salt of ENMD-981693. 4.1

1382

ENMD-2076 selectively inhibits Aurora A (IC50 = 14 nM) instead of Aurora-B (IC50 = 700 nM) and has also potent activity (ranging 0.04 -- 21 µM IC50 in some cases) inhibiting other kinases (FGFR3, PDGFR, VEGFR1 and FLT3). A number of preclinical studies have shown the antitumoral potential of ENMD-2076. ENMD-2076 has shown significant cytotoxicity against multiple myeloma cell lines and primary cells, with minimal cytotoxicity to hematopoietic progenitors [87]. Similar results have been described in cell line-derived human colorectal cancer xenografts [88]. Interestingly, its activity against tumor cells both in vitro and in vivo takes places through several pathways and molecules (Aurora kinases A and B, PI3K/AKT and FGFR3) [87,89]. The results from one Phase I clinical trial in patients with advance solid tumors [90] showed that ENMD-2076 is well tolerated, has a linear pharmacokinetic profile and has as the most common drug-related adverse events hypertension, nausea/vomiting

Expert Opin. Ther. Targets (2014) 18(12)

Aurora kinase A inhibitors

Table 3. Aurora-A-kinase-specific inhibitors in clinical trials. Inhibitor (source) Preclinical data

Specificity (IC50 in nM) Clinical trial information (status and identifier)*

ENMD-2076 (EntreMed)

Aurora-A-specific inhibitor AurkA = 14; AurkB = 700; AurkC = NA [88]

Phase I Relapsed or refractory hematological malignancies and multiple myeloma (completed: NCT00904787) Advanced cancer (active: NCT00658671 [90]) Phase II Advanced or metastatic triple negative breast cancers (recruiting: NCT01639248 and NCT00806065) Advanced/metastatic soft tissue sarcoma (recruiting; NCT01719744) Ovarian clear cell cancers (recruiting; NCT01914510 [91]) MK-5108/ VX-689 (Merk/Vertex)

Aurora-A-specific inhibitor AurkA = 0.064; AurkB = 14; AurkC = 12 [94]

Phase I Alone or in combination with docetaxel in patients with advanced solid tumors (completed; NCT00543387 [100]) MLN-8054 (Millennium Pharmaceuticals)

Aurora-A-specific inhibitor AurkA = 0.064; AurkB = 14; AurkC = 12 [101]

Phase I Advance malignancies, breast, colon, pancreatic and bladder neoplasms (terminated: NCT00249301 and NCT00652158) [107,108] Alisertib (MLN-8237) (Millennium Pharmaceuticals)

Aurora-A-specific inhibitor AurkA = 1; AurkB > 200; AurkC = NA [113]

Phase I Advanced malignancies, solid tumors and lymphomas (completed: NCT01512758, NCT00962091, NCT01154816 [133], NCT00651664 [132], NCT00697346 [134], NCT00500903 [131], NCT01714947 and 2008-006981-27; recruiting: NCT01898078) In combination with: Esomeprazole and Rifampicin in advanced solid tumors and lymphomas (recruiting: NCT01844583) Vorinostat in treating patients with relapsed or recurrent Hodgkin lymphoma, B-cell non-Hodgkin lymphoma or peripheral T-cell lymphoma (recruiting: NCT01567709) Rituximab and Bortezomib in treating patients with relapsed or refractory mantle cell lymphoma or B-cell low grade non-Hodgkin lymphoma (recruiting: NCT01695941) Cytarabine and Idarubicin in acute myelogenous leukemia (recruiting: NCT01779843) Gemcitabine hydrochloride in treating patients with solid tumors or advanced pancreatic cancer (recruiting: NCT01924260) Romidepsin in patients with relapsed or refractory B-cell or T-cell lymphomas (recruiting: NCT01897012) Irinotecan hydrochloride in treating patients with advanced solid tumors or colorectal cancer (recruiting: NCT01923337) Pazopanib in patients with advanced, previously treated non-hematologic solid tumors (recruiting: NCT01639911) Paclitaxel albumin-stabilized nanoparticle formulation (nab-paclitaxel) in treating patients with solid malignancies that are metastatic or cannot be removed by surgery (recruiting: NCT01677559) Added to routine radiation therapy and Cetuximab in patients with head and neck cancer (recruiting: NCT01540682) Docetaxel as a treatment for advanced solid tumors (active: NCT01094288) Phase II In patients with unresectable stage III-IV melanoma (recruiting: NCT01316692), advanced nonhematological malignancies (active: NCT01045421; terminated: 2008-006981-27), relapsed or refractory non-Hodgkin’s lymphoma (completed: NCT00807495) [135], acute myeloid leukemia or myelodysplastic syndrome (completed: NCT00830518; ongoing: 2008-006977-34), with platinumrefractory or platinum-resistant epithelial ovarian, fallopian tube or primary peritoneal carcinomas (completed: NCT00853307; 2008-006979-72 [136]; ongoing: 2009-011428-79), advanced or metastatic sarcoma (recruiting: NCT01653028), relapsed or refractory solid tumors or leukemia (active: NCT01154816), relapsed or refractory peripheral T-cell non-Hodgkin lymphoma (recruiting: NCT01466881), metastatic neuroendocrine prostate cancer (recruiting: NCT01799278) In combination with: Paclitaxel in patients with breast and ovarian cancers (active: NCT01091428) and with small cell lung cancer who have relapsed or did not respond to first line standard therapy (not yet open for recruitment: NCT02038647) Rituximab for relapsed or refractory B-cell non-Hodgkin lymphomas (recruiting: NCT01812005) Erlotinib hydrochloride in patients with advanced or metastatic non-small cell lung cancer (recruiting: NCT01471964) Abiraterone or Prednisone in prostate cancer (recruiting: NCT01848067) Bortezomib in treating patients with relapsed or refractory multiple myeloma (recruiting: NCT01034553) *From National Cancer Institute and EU Clinical Trials Register web pages: http://www.clinicaltrials.gov and https://www.clinicaltrialsregister.eu/ctr-search/search NA: Not available.

Expert Opin. Ther. Targets (2014) 18(12)

1383

rez de Castro M. Malumbres & I. Pe

Table 3. Aurora-A-kinase-specific inhibitors in clinical trials (continued). Inhibitor (source) Preclinical data

Specificity (IC50 in nM) Clinical trial information (status and identifier)*

Rituximab and Vincristine in patients with relapsed or refractory diffuse large B-cell lymphoma or transformed follicular lymphoma (active: NCT01397825; 2011-000609-32) Phase III In combination with Pralatrexate, Gemcitabine or Romidepsin in patients with relapsed or refractory peripheral T-cell lymphoma (recruiting: NCT01482962) *From National Cancer Institute and EU Clinical Trials Register web pages: http://www.clinicaltrials.gov and https://www.clinicaltrialsregister.eu/ctr-search/search NA: Not available.

and fatigue. Partial response was detected in 2 out of the 57 patients that were evaluated. Both of them were ovarian cancer cases. At least three different Phase II trials have been started (Table 3). One of them has shown a progression-free survival rate at 6 months of 22% with a median time to progression of 3.6 months [91]. This effect of ENMD-2076 in platinum-resistant ovarian cancers was not associated to any of the biomarkers analyzed in this study. However, some predictive biomarkers of sensitivity have been described in preclinical breast cancer models [92]. ENMD-2076 showed more robust activity against cell lines lacking estrogen receptor expression and those without increased HER2 expression. Within the triple-negative breast cancer subset, cell lines with a p53 mutation and increased p53 expression were more sensitive to the cytotoxic and pro-apoptotic effects of ENMD2076 exposure than those with decreased p53 expression. Finally, ENMD-2076 has also radiosensitizing properties in canine mast cell tumors in vitro [93], which highlights the potential of this inhibitor in combination with other cancer therapies. MK-5108 This agent, also known as VX-689, has been developed by Merck and Vertex. MK-5108 specifically inhibits AURKA with an IC50 of 0.064 nM. The first preclinical studies showed a potent antitumoral activity of MK-5108 alone or in combination with Docetaxel in cell lines from different tumor types [94]. Later, it was reported that MK-5108 was able to decrease the proliferation of epithelial ovarian cancer stem cells, the ones that are able to resist chemotherapy [95]. In uterine leiomyosarcomas, MK-5108 also inhibits proliferation and induces apoptosis both in vitro and in vivo, but it does not synergize with gemcitabine or docetaxel [96]. By contrast, a cooperative effect was detected in lymphoma cells treated with the histone deacetylase inhibitor vorinostat, neuroblastoma cells treated with the anti-GD2 ganglioside (GD2) 14G2a mouse mAb and lung cancer cell lines treated with cisplatin or docetaxel [97-99]. One Phase I trial has been completed for MK-5108 alone or in combination with docetaxel in patients with advanced solid tumors [100]. No dose-limiting toxicity was observed in the monotherapy arm; disease stabilization was seen in 32% of the patients treated 4.2

1384

with MK-5108 alone or in combination. Although partial response was observed in 12% of the patients in the combination arm, none was seen in the monotherapy arm. MLN8054 and MLN8237 (Alisertib) These two compounds from Millennium Pharmaceuticals are ATP-competitive and reversible inhibitors with a potent activity against AURKA (IC50 = 4 nM for MLN8054 and IC50 = 1 nM for MLN8237). MLN8054, which was patented in 2004, is a benzazepine-fused ring compound with potent antitumoral properties as it has been demonstrated in several preclinical studies. Specifically, MLN8054 inhibits the growth of colon, breast, lung, ovary, neuroblastoma and prostate cancer cell lines and that of colon cancer human xenografts [101-103]. Cellular defects induced by MLN-8054, spindle pole and chromosome congression/segregation abnormalities, were consisting with AURKA-specific inhibition [103,104]. Moreover, it has been shown that MLN-8054 can induce apoptosis in p53-negative cells in a p73-dependent manner [105]. Using this Aurora A inhibitor, it was suggested for the first time, both in vitro and in vivo, that the main cellular phenotype induced by Aurora A inhibition is senescence rather than apoptosis [106]. Two Phase I clinical trials initiated in advance malignancies and solid tumors showed somnolence and liver toxicity as the major dose-limiting toxicities for MLN-8054 (maximum once-daily administration dose was 30 mg/day) [107,108]. The formerly was a consequence of the structural and functional similarities that this agent has with benzodiazepines. Due to these adverse effects this oraladministrated drug could not be used in the doses that allowed a proper Aurora A inhibition. Therefore, MLN8054 development was abandoned. MLN8237, also named Alisertib, is by far the most frequently used AURKA inhibitor (Figure 4B). Patented in 2008, MLN8237 is another benzazepine-fused pyrimidine ring compound that inhibits Aurora A better than MLN8054 (IC50 = 1 nM) and has decreased tendency to cause somnolence. Several preclinical studies have shown, in vitro and in vivo, the antitumoral properties of MLN8237 in cancer cell lines from different tumor types including lymphomas [109,110], leukemias [111,112], multiple myeloma [113], breast, ovarian [114], bladder [115] and malignant peripheral nerve sheath [116] tumors. 4.3

Expert Opin. Ther. Targets (2014) 18(12)

Aurora kinase A inhibitors

Different works have demonstrated that MLN8237 induces the defects expected for a bona fide AURKA inhibitor [117] and inhibits cell proliferation mainly through the induction of senescence [118,119]. To note, is the fact that loss of p53 sensitizes cells to MLN-8237 treatment [120] and that Ki67 could be a good biomarker for the anti-proliferative effects of this AURKA inhibitor [121]. Several preclinical studies have shown that Alisertib cooperates with many compounds and cancer treatments (Table 3). Specifically, MLN8237 induces additive effects with: dexamethasone, doxorubicin or bortezomib in multiple myeloma cells [113]; tyrosine kinase inhibitor nilotinib or the DNA synthesis inhibitor cytarabine in myeloid leukemias [122,123]; the MT stabilizer docetaxel in mantle cell lymphomas and gastrointestinal adenocarcinomas [124,125]; radiation in atypical teratoid rhabdoid tumor cells [126]; the mTOR inhibitor rapamycin in uterine leiomyosarcoma [127]; the MT assembly inhibitor vincristine and the mAb against the protein CD20 rituximab in aggressive B-cell non-Hodgkin lymphoma [128]; the DNA crosslinker cisplatin in esophageal adenocarcinomas [129]; the histone deacetylase inhibitor vorinostat in pediatric neuroblastoma, medulloblastoma and leukemia cell lines [130] and the MT inhibitor paclitaxel in ovarian cancer cells [114]. Eight Phase I clinical trials have been completed for Alisertib in advanced solid tumors and lymphomas. In one of them (NCT00500903), the safety, pharmacokinetics, pharmacodynamics and the bioavailability of two oral formulations was evaluated [131]. Adverse effects included fatigue, nausea and neutropenia. Plasma exposures increased dose proportionally (5 -- 150 mg/day) and were similar for the two formulations (enteric-coated tablet and the original powder-in-capsule). The terminal half-life was 23 h. Partial response and stable disease was detected in 1 and 23% of the patients, respectively. In another Phase I trial (NCT00651664), patients with advanced solid tumors were selected and similar results were obtained [132]. Neutropenia and stomatitis were the most common dose-limiting toxicities. The maximum tolerated dose for the 7-day and 21-day schedules were 50 mg twice daily and 50 mg once daily, respectively. The mean terminal half-life was ~ 19 h. At steady state, pharmacodynamic effects were shown by accumulation of mitotic and apoptotic cells in skin, and mitotic abnormalities in tumor specimens, which corroborates with the Aurora A inhibition by MLN8237. Stable disease was observed in ~ 10% of the patients, without notable cumulative toxicity. In a third Phase I trial (NCT011 54816), pharmacokinetic properties of MLN8237 were analyzed in 37 children with advanced/refractory tumors [133]. Myelosuppression, mood alteration and mucositis were the main dose-limiting effects. There was one partial response (3%) and six (18%) with prolonged stable disease among 33 evaluable subjects. Finally, a Phase I trial (NCT006 97346) testing MLN8237 in relapsed/refractory multiple myeloma, non-Hodgkin lymphoma and chronic lymphocytic leukemia has been recently published [134]. The results were similar to the ones obtained in other Phase I trials for MLN 8237. Neutropenia (45%), thrombocytopenia (28%), anemia

(19%) and leukopenia (19%) were the main adverse effects found among the 58 enrolled patients. The maximum tolerated dose on the 7-day schedule was 50 mg twice daily and the terminal half-life was ~ 19 h. Six (13%) patients achieved partial responses and 13 (28%) stable disease. Up to 13 Phase II studies have been initiated in the US and in Europe to test Alisertib in a broad range of human tumors. The results from two of these studies have recently been published. In one of them (NCT00807495), 48 patients suffering from relapsed and refractory, aggressive B-cell and T-cell non-Hodgkin lymphomas were treated with Alisertib administered orally at 50 mg twice daily for 7 days in 21-day cycles [135]. The adverse events were similar to the ones detected in Phase I trials: low numbers of blood cells, stomatitis and fatigue. The overall response rate was 27%. The other, published Phase II study was conducted in the US and in Europe (NCT00853307; 2008-006979-72) in patients with platinum-resistant or platinum-refractory epithelial ovarian, fallopian tube or primary peritoneal carcinoma [136]. Responses (of 6.9 -- 11.1 months duration) and stable disease (mean duration of 2.86 months) were observed in 10 and 52% of the patients, respectively. This modest antitumor activity in patients with advanced ovarian cancers was observed together with the common Alisertib-associated adverse effects (neutropenia, leukopenia, stomatitis, thrombocytopenia and febrile neutropenia). At least 11 Phase I and 8 Phase II trials are recruiting or currently testing Alisertib in combination with other drugs or treatments (Table 3). Among the drugs/treatments, there are inhibitors for DNA synthesis, MT dynamics, angiogenesis, the immune system, proton pumps, antibiotics and different molecules such as EGFR, HDAC, CD20 and CYP17 (Table S3). As a consequence of the promising results obtained in Phase I and II studies with solid and non-solid tumors, confirmatory mono-therapy and combination studies have been initiated (Table 3). Furthermore, a Phase III trial (NCT0148 2962) is currently recruiting patients with refractory peripheral T-cell lymphoma to test Alisertib alone or in combination with pralatrexate, gemcitabine or romidepsin. 5.

Conclusions

AURKA has a two-sided importance in oncology. It is amplified and over-expressed in many human cancers and has been causally associated with tumor development and progression. On the other hand, its depletion or inhibition impairs cell proliferation of tumor cells, reason why it has been considered a therapeutic target for cancer treatment. Several Aurora kinase inhibitors have been developed during the last decade, and most pre-clinical studies showed the potent antiproliferative effect of these compounds. Approximately 20 Aurora kinase inhibitors have been or are going to be tested in almost one hundred clinical trials, and half of them use AURKA-specific inhibitors. Alisertib (MLN8237) is the most frequently tested Aurora A inhibitor (40 clinical trials

Expert Opin. Ther. Targets (2014) 18(12)

1385

rez de Castro M. Malumbres & I. Pe

completed, recruiting or active), and Phase I and Phase II trials have shown promising results in the treatment of different human cancers. The results obtained in monotherapies or combination trials with Aurora A kinase inhibitors during the next years will be critical to define the potential of this agent in anticancer therapy. 6.

Expert opinion

Given the essential role of Aurora A in mitosis, its inhibition could in principle impair the proliferation of any tumor. This makes Aurora A inhibitors potential universal drugs for the treatment of oncologic disorders. However, their inclusion in routine antitumor therapies is still far to be achieved, and a number of issues need to be addressed to improve the efficacy of AURKA inhibition for the treatment of cancer diseases. New and/or improved ways to inactivate AURKA Although very potent and specific Aurora A inhibitors have been developed, drug resistance has already been reported in tumor cells [137,138]. It will be therefore important to have compounds with different chemical structures or mechanisms of action. Although most Aurora inhibitors are ATP competitors, Tripolin A, which has a modest affinity for AURKA (IC50 = 1.5 µM) [139], shows a non-ATP competitive mode of action. Other strategies to prevent resistance are based on the mechanisms that allow cancer cells to resist the action of Aurora A inhibitors. That is the case of resistance to VX680 in breast cancer cells due to the induction of autophagy. Thus, repression of autophagy by depletion of either LC3 or ATG5 sensitizes cancer cells to VX-680-induced apoptosis [138]. Another example has been the identification of proteins that contribute resistance to other two Aurora inhibitors, CYC116 and ZM447439 [137]. Specifically, platelet-activating factor acetylhydrolase and GTP-binding nuclear protein Ran contribute to the development of resistance to ZM447439, whereas serine hydroxymethyltransferase, serpin B5 and calretinin help to overcome the effects of CYC116. Further studies in this area will provide clinicians with potent tools to monitor patients and avoid drug resistance to Aurora inhibitors. 6.1

Biomarkers Different markers have been used to predict or monitor the efficacy of Aurora inhibitors, including mitotic index, spindle bipolarity and chromosome alignment [104], nuclear volume [82] and percentage of aligned spindles [140]. The crosstalk between Aurora A and p53 [69] makes this protein a potential marker for a more potent effect of AURKA inhibitors. As Aurora A inhibits p53 [37], it can be postulated that a wild type, functional p53 pathway could be reactivated upon Aurora A inhibition and, therefore, sensitize cells to this treatment. In fact, depletion of Aurora A in Zebrafish causes p53-dependent cell death [141]. However, it has also been reported the opposite, the lack of p53-sensitizing cells to inhibition of Alisertib [120] and also that breast cancer cell 6.2

1386

lines with a p53 mutation are more sensitive to ENMD2076 [92]. Because AURKA inhibition leads to apoptosis in p53-deficient cancer cells in a p73-dependent manner [105], this last protein could also be studied to confirm its utility as a biomarker for AURKA inhibitors efficacy. High proliferative rates have also been associated with tumors that better respond to Aurora A inhibitors, since proliferation requires Aurora A activity. Aurora A and its regulator TPX2 are among the different markers that could be used to test proliferation status. Moreover, both TPX2 and Aurora A are co-overexpressed in many human tumors, which has made others to postulate the hypothesis that both Aurora A and TPX2 work as an oncogenic holoenzyme [72]. Interestingly, it has been shown that TPX2 can alter the binding mode with Aurora of the inhibitor VX-680 [142]. However, Aurora A amplification/overexpression has been analyzed, with negative correlation with clinical response, within a Phase II trial conducted in advanced lymphomas [135]. Other potential biomarkers for Aurora A inhibition activity are NEDD9, whose depletion in tumor cells increases sensitivity to Aurora A inhibitors [143]; p21, whose deficiency enhances the activity of BPRIK0609S1 [144]; and estrogen receptor and HER2, whose expression in breast cancer cells have been related with the antiproliferative activity of ENMD2076 [92]. All these proteins should be further analyzed to confirm their potential. Besides all these candidate-based approaches, genomic strategies such as deep sequencing, transcriptomics or the analysis of the methylome and metabolome must be used to identify new biomarkers. Combination therapies: looking for synergisms One of the most interesting uses of Aurora A inhibitors is their ability to potentiate the responses induced by other agents. Several Phase I and II trials have been initiated to test the effect of Aurora A inhibitors in combination with other drugs that already approved for cancer treatment (Table S3). Some of those trials have been completed offering promising results. We believe that further combinations could be explored. The newly discovered roles of Aurora A in DNA damage, senescence, aneuploidy and differentiation offer new possibilities [82,112,119]. Therefore, DNA repair inhibitors or agents that selectively kill aneuploid cells can be interesting candidates for combination therapies. Among them, ATR inhibitors, the energy stress-inducing agent AICAR, the protein-folding inhibitor 17-AAG and the autophagy inhibitor chloroquine have already shown antitumoral properties in preclinical studies [145,146]. In parallel, massive screenings must be considered for converting the senescence or differentiation phenotype induced by Aurora A inhibitors into cell death. Further research on the physiological relevance of Aurora A in different cell types may be required to provide a rationale for these new approaches in the future. In conclusion, the use of AURKA inhibitors have shown promising results during the last years. We anticipate that additional discoveries on the function of Aurora A kinase 6.3

Expert Opin. Ther. Targets (2014) 18(12)

Aurora kinase A inhibitors

will also significantly impact the field improving the clinical use of Aurora A kinase inhibitors.

Acknowledgments We want to apologize for all those works that have not been cited in this review due to space constrains.

Declaration of interest We want to thank David Partida for his technical assistance. This work was supported by grants from the Ministerio de Bibliography Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

Perez de Castro I, de Carcer G, Malumbres M. A census of mitotic cancer genes: new insights into tumor cell biology and cancer therapy. Carcinogenesis 2007;28(5):899-912

2.

Chan KS, Koh CG, Li HY. Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis 2012;3:e411

3.

Domenech E, Malumbres M. Mitosis-targeting therapies: a troubleshooting guide. Curr Opin Pharmacol 2013;13(4):519-28

4.

Carmena M, Earnshaw WC. The cellular geography of aurora kinases. Nat Rev Mol Cell Biol 2003;4(11):842-54

5.

Chan CS, Botstein D. Isolation and characterization of chromosome-gain and increase-in-ploidy mutants in yeast. Genetics 1993;135(3):677-91

6.

.

7.

.

8.

Cowley DO, Rivera-Perez JA, Schliekelman M, et al. Aurora-A kinase is essential for bipolar spindle formation and early development. Mol Cell Biol 2009;29(4):1059-71 Describe the phenotypes of the knock out (KO) mouse models of Aurora kinases A, B and C. Lu LY, Wood JL, Ye L, et al. Aurora A is essential for early embryonic development and tumor suppression. J Biol Chem 2008;283(46):31785-90 Describe the phenotypes of the KO mouse models of Aurora kinases A, B and C. Sasai K, Parant JM, Brandt ME, et al. Targeted disruption of Aurora A causes abnormal mitotic spindle assembly,

.

9.

.

10.

.

Economı´a y Competitividad (MINECO; SAF2010-19710 to I Perez de Castro; and SAF2012-38215 to M Malumbres), Fundacio´n Ramo´n Areces, the OncoCycle Programme (S2010/BMD-2470) from the Comunidad de Madrid, the OncoBIO Consolider-Ingenio 2010 Programme (CSD2007-00017) from the MINECO, and the European Union Seventh Framework Programme (MitoSys project; HEALTH-F5-2010-241548). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

chromosome misalignment and embryonic lethality. Oncogene 2008;27(29):4122-7 Describe the phenotypes of the KO mouse models of Aurora kinases A, B and C. Ferna´ndez-Miranda G, Trakala M, Martı´n J, et al. Genetic disruption of aurora B uncovers an essential role for aurora C during early mammalian development. Development 2011;138(13):2661-72 Describe the phenotypes of the KO mouse models of Aurora kinases A, B and C. Dieterich K, Soto Rifo R, Faure AK, et al. Homozygous mutation of AURKC yields large-headed polyploid spermatozoa and causes male infertility. Nat Genet 2007;39(5):661-5 Describe the phenotypes of the KO mouse models of Aurora kinases A, B and C.

11.

Glover DM, Leibowitz MH, McLean DA, Parry H. Mutations in aurora prevent centrosome separation leading to the formation of monopolar spindles. Cell 1995;81(1):95-105

12.

Eyers PA, Erikson E, Chen LG, Maller JL. A novel mechanism for activation of the protein kinase Aurora A. Curr Biol 2003;13(8):691-7

13.

Tsai MY, Wiese C, Cao K, et al. A Ran signalling pathway mediated by the mitotic kinase Aurora A in spindle assembly. Nat Cell Biol 2003;5(3):242-8

14.

15.

Zhao ZS, Lim JP, Ng YW, et al. The GIT-associated kinase PAK targets to the centrosome and regulates Aurora-A. Mol Cell 2005;20(2):237-49 Van Horn RD, Chu S, Fan L, et al. Cdk1 activity is required for mitotic activation of aurora A during G2/M Expert Opin. Ther. Targets (2014) 18(12)

transition of human cells. J Biol Chem 2010;285(28):21849-57 16.

Hirota T, Kunitoku N, Sasayama T, et al. Aurora-A and an interacting activator, the LIM protein Ajuba, are required for mitotic commitment in human cells. Cell 2003;114(5):585-98

17.

Huang YH, Wu CC, Chou CK, Huang CY. A translational regulator, PUM2, promotes both protein stability and kinase activity of Aurora-A. PLoS One 2011;6(5):e19718

18.

Hutterer A, Berdnik D, Wirtz-Peitz F, et al. Mitotic activation of the kinase Aurora-A requires its binding partner Bora. Dev Cell 2006;11(2):147-57

19.

Joukov V, De Nicolo A, Rodriguez A, et al. Centrosomal protein of 192 kDa (Cep192) promotes centrosome-driven spindle assembly by engaging in organelle-specific Aurora A activation. Proc Natl Acad Sci USA 2010;107(49):21022-7

20.

den Hollander J, Rimpi S, Doherty JR, et al. Aurora kinases A and B are upregulated by Myc and are essential for maintenance of the malignant state. Blood 2010;116(9):1498-505

21.

Reboutier D, Troadec MB, Cremet JY, et al. Nucleophosmin/B23 activates Aurora A at the centrosome through phosphorylation of serine 89. J Cell Biol 2012;197(1):19-26

22.

Pugacheva EN, Jablonski SA, Hartman TR, et al. HEF1-dependent Aurora A activation induces disassembly of the primary cilium. Cell 2007;129(7):1351-63

23.

Kufer TA, Sillje HH, K€orner R, et al. Human TPX2 is required for targeting Aurora-A kinase to the spindle. J Cell Biol 2002;158(4):617-23

1387

rez de Castro M. Malumbres & I. Pe

24.

25.

26.

27.

28.

29.

Bayliss R, Sardon T, Vernos I, Conti E. Structural basis of Aurora-A activation by TPX2 at the mitotic spindle. Mol Cell 2003;12(4):851-62 Zorba A, Buosi V, Kutter S, et al. Molecular mechanism of Aurora A kinase autophosphorylation and its allosteric activation by TPX2. Elife 2014;3:e02667 Littlepage LE, Ruderman JV. Identification of a new APC/C recognition domain, the A box, which is required for the Cdh1-dependent destruction of the kinase Aurora-A during mitotic exit. Genes Dev 2002;16(17):2274-85 Shao S, Wang Y, Jin S, et al. Gadd45a interacts with aurora-A and inhibits its kinase activity. J Biol Chem 2006;281(39):28943-50 Zeng K, Bastos RN, Barr FA, Gruneberg U. Protein phosphatase 6 regulates mitotic spindle formation by controlling the T-loop phosphorylation state of Aurora A bound to its activator TPX2. J Cell Biol 2010;191(7):1315-32 Kiat LS, Hui KM, Gopalan G. Aurora-A kinase interacting protein (AIP), a novel negative regulator of human Aurora-A kinase. J Biol Chem 2002;277(47):45558-65

30.

Fumoto K, Lee PC, Saya H, Kikuchi A. AIP regulates stability of Aurora-A at early mitotic phase coordinately with GSK-3beta. Oncogene 2008;27(32):4478-87

31.

Kwon YW, Kim IJ, Wu D, et al. Pten regulates Aurora-A and cooperates with Fbxw7 in modulating radiation-induced tumor development. Mol Cancer Res 2012;10(6):834-44

32.

33.

34.

35.

1388

A to promote checkpoint recovery. Nature 2008;455(7209):119-23 36.

37.

.

38.

39.

40.

41.

42.

Shi Y, Solomon LR, Pereda-Lopez A, et al. Ubiquitin-specific cysteine protease 2a (USP2a) regulates the stability of Aurora-A. J Biol Chem 2011;286(45):38960-8 Chen SS, Chang PC, Cheng YW, et al. Suppression of the STK15 oncogenic activity requires a transactivationindependent p53 function. EMBO J 2002;21(17):4491-9 Perez de Castro I, Aguirre-Portoles C, Martin B, et al. A SUMOylation motif in Aurora-A: implications for spindle dynamics and oncogenesis. Front Oncol 2011;1:50 Macu˚rek L, Lindqvist A, Lim D, et al. Polo-like kinase-1 is activated by aurora

43.

44.

45.

Wirtz-Peitz F, Nishimura T, Knoblich JA. Linking cell cycle to asymmetric division: Aurora-A phosphorylates the Par complex to regulate Numb localization. Cell 2008;135(1):161-73

46.

Regan JL, Sourisseau T, Soady K, et al. Aurora A kinase regulates mammary epithelial cell fate by determining mitotic spindle orientation in a Notch-dependent manner. Cell Rep 2013;4(1):110-23

47.

Lee CY, Andersen RO, Cabernard C, et al. Drosophila Aurora-A kinase inhibits neuroblast self-renewal by regulating aPKC/Numb cortical polarity and spindle orientation. Genes Dev 2006;20(24):3464-74

48.

Lee DF, Su J, Ang YS, et al. Regulation of embryonic and induced pluripotency by aurora kinase-p53 signaling. Cell Stem Cell 2012;11(2):179-94

49.

Piazzolla D, Palla AR, Pantoja C, et al. Lineage-restricted function of the pluripotency factor NANOG in stratified epithelia. Nat Commun 2014;5:4226

50.

Huang YS, Jung MY, Sarkissian M, Richter JD. N-methyl-D-aspartate receptor signaling results in Aurora kinase-catalyzed CPEB phosphorylation and alpha CaMKII mRNA polyadenylation at synapses. EMBO J 2002;21(9):2139-48

Romain C, Paul P, Kim KW, et al. Targeting Aurora kinase-A downregulates cell proliferation and angiogenesis in neuroblastoma. J Pediatr Surg 2014;49(1):159-65

51.

Plotnikova OV, Pugacheva EN, Golemis EA. Aurora A kinase activity influences calcium signaling in kidney cells. J Cell Biol 2011;193(6):1021-32

Katsha A, Soutto M, Sehdev V, et al. Aurora kinase A promotes inflammation and tumorigenesis in mice and human gastric neoplasia. Gastroenterology 2013;145(6):1312-22; e1-8

52.

Yao JE, Yan M, Guan Z, et al. Aurora-A down-regulates IkappaBalpha via Akt activation and interacts with insulin-like growth factor-1 induced phosphatidylinositol 3-kinase pathway for cancer cell survival. Mol Cancer 2009;8:95

Mori D, Yamada M, Mimori-Kiyosue Y, et al. An essential role of the aPKCAurora A-NDEL1 pathway in neurite elongation by modulation of microtubule dynamics. Nat Cell Biol 2009;11(9):1057-68

53.

Wu JC, Chen TY, Yu CT, et al. Identification of V23RalA-Ser194 as a critical mediator for Aurora-A-induced cellular motility and transformation by small pool expression screening. J Biol Chem 2005;280(10):9013-22

54.

Tsunematsu T, Takihara Y, Ishimaru N, et al. Aurora-A controls pre-replicative complex assembly and DNA replication by stabilizing geminin in mitosis. Nat Commun 2013;4:1885

55.

Briassouli P, Chan F, Savage K, et al. Aurora-A regulation of nuclear factorkappaB signaling by phosphorylation of IkappaBalpha. Cancer Res 2007;67(4):1689-95

Seki A, Coppinger JA, Jang CY, et al. Bora and the kinase Aurora a cooperatively activate the kinase Plk1 and control mitotic entry. Science 2008;320(5883):1655-8 Katayama H, Sasai K, Kawai H, et al. Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53. Nat Genet 2004;36(1):55-62 In this study it is reported for the first time the relationship between Aurora kinase A (AURKA) and p53. Mendez R, Murthy KG, Ryan K, et al. Phosphorylation of CPEB by Eg2 mediates the recruitment of CPSF into an active cytoplasmic polyadenylation complex. Mol Cell 2000;6(5):1253-9 Sarkissian M, Mendez R, Richter JD. Progesterone and insulin stimulation of CPEB-dependent polyadenylation is regulated by Aurora A and glycogen synthase kinase-3. Genes Dev 2004;18(1):48-61

Sasayama T, Marumoto T, Kunitoku N, et al. Over-expression of Aurora-A targets cytoplasmic polyadenylation element binding protein and promotes mRNA polyadenylation of Cdk1 and cyclin B1. Genes Cells 2005;10(7):627-38 Otto T, Horn S, Brockmann M, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 2009;15(1):67-78

Expert Opin. Ther. Targets (2014) 18(12)

Aurora kinase A inhibitors

56.

Wu CC, Yu CT, Chang GC, et al. Aurora-A promotes gefitinib resistance via a NF-kappaB signaling pathway in p53 knockdown lung cancer cells. Biochem Biophys Res Commun 2011;405(2):168-72

65.

Zhang D, Hirota T, Marumoto T, et al. Cre-loxP-controlled periodic Aurora-A overexpression induces mitotic abnormalities and hyperplasia in mammary glands of mouse models. Oncogene 2004;23(54):8720-30

57.

Yang H, He L, Kruk P, et al. Aurora-A induces cell survival and chemoresistance by activation of Akt through a p53-dependent manner in ovarian cancer cells. Int J Cancer 2006;119(10):2304-12

66.

Wang H, Somers GW, Bashirullah A, et al. Aurora-A acts as a tumor suppressor and regulates self-renewal of Drosophila neuroblasts. Genes Dev 2006;20(24):3453-63

67.

Carter SL, Eklund AC, Kohane IS, et al. A signature of chromosomal instability inferred from gene expression profiles predicts clinical outcome in multiple human cancers. Nat Genet 2006;38(9):1043-8

68.

Meraldi P, Honda R, Nigg EA. Aurora-A overexpression reveals tetraploidization as a major route to centrosome amplification in p53-/- cells. EMBO J 2002;21(4):483-92

69.

Mao JH, Wu D, Perez-Losada J, et al. Crosstalk between Aurora-A and p53: frequent deletion or downregulation of Aurora-A in tumors from p53 null mice. Cancer Cell 2007;11(2):161-73

58.

Anand S, Penrhyn-Lowe S, Venkitaraman AR. AURORA-A amplification overrides the mitotic spindle assembly checkpoint, inducing resistance to Taxol. Cancer Cell 2003;3(1):51-62

59.

Miao X, Sun T, Wang Y, et al. Functional STK15 Phe31Ile polymorphism is associated with the occurrence and advanced disease status of esophageal squamous cell carcinoma. Cancer Res 2004;64(8):2680-3

60.

61.

..

62.

..

63.

64.

Sun H, Bai J, Chen F, et al. Lack of an association between AURKA T91A polymorphisms and breast cancer: a meta-analysis involving 32,141 subjects. Breast Cancer Res Treat 2011;125(1):175-9 Bischoff JR, Anderson L, Zhu Y, et al. A homologue of Drosophila aurora kinase is oncogenic and amplified in human colorectal cancers. EMBO J 1998;17(11):3052-65 Demonstrated for the first time the transforming activity of the over-expression of Aurora kinase A. Zhou H, Kuang J, Zhong L, et al. Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation. Nat Genet 1998;20(2):189-93 Demonstrated for the first time the transforming activity of the over-expression of Aurora kinase A. Tatsuka M, Sato S, Kitajima S, et al. Overexpression of Aurora-A potentiates HRAS-mediated oncogenic transformation and is implicated in oral carcinogenesis. Oncogene 2005;24(6):1122-7 Wang X, Zhou YX, Qiao W, et al. Overexpression of aurora kinase A in mouse mammary epithelium induces genetic instability preceding mammary tumor formation. Oncogene 2006;25(54):7148-58

70.

Giubettini M, Asteriti IA, Scrofani J, et al. Control of Aurora-A stability through interaction with TPX2. J Cell Sci 2011;124(Pt 1):113-22

71.

Perez de Castro I, Malumbres M. Mitotic stress and chromosomal instability in cancer: the case for TPX2. Genes Cancer 2012;3(11-12):721-30

72.

Asteriti IA, Rensen WM, Lindon C, et al. The Aurora-A/TPX2 complex: a novel oncogenic holoenzyme? Biochim Biophys Acta 2010;1806(2):230-9

73.

74.

75.

Burum-Auensen E, De Angelis PM, Schjølberg AR, et al. Subcellular localization of the spindle proteins Aurora A, Mad2, and BUBR1 assessed by immunohistochemistry. J Histochem Cytochem 2007;55(5):477-86 Lim KH, Brady DC, Kashatus DF, et al. Aurora-A phosphorylates, activates, and relocalizes the small GTPase RalA. Mol Cell Biol 2010;30(2):508-23 Dimova I, Raitcheva S, Dimitrov R, et al. Correlations between c-myc gene copy-number and clinicopathological parameters of ovarian tumours. Eur J Cancer 2006;42(5):674-9

Expert Opin. Ther. Targets (2014) 18(12)

76.

Giet R, Prigent C. The Xenopus laevis aurora/Ip11p-related kinase pEg2 participates in the stability of the bipolar mitotic spindle. Exp Cell Res 2000;258(1):145-51

77.

Hannak E, Kirkham M, Hyman AA, Oegema K. Aurora-A kinase is required for centrosome maturation in Caenorhabditis elegans. J Cell Biol 2001;155(7):1109-16

78.

Roghi C, Giet R, Uzbekov R, et al. The Xenopus protein kinase pEg2 associates with the centrosome in a cell cycledependent manner, binds to the spindle microtubules and is involved in bipolar mitotic spindle assembly. J Cell Sci 1998;111(Pt 5):557-72

79.

Schumacher JM, Ashcroft N, Donovan PJ, Golden A. A highly conserved centrosomal kinase, AIR-1, is required for accurate cell cycle progression and segregation of developmental factors in Caenorhabditis elegans embryos. Development 1998;125(22):4391-402

80.

Du J, Hannon GJ. Suppression of p160ROCK bypasses cell cycle arrest after Aurora-A/STK15 depletion. Proc Natl Acad Sci USA 2004;101(24):8975-80

81.

Kunitoku N, Sasayama T, Marumoto T, et al. CENP-A phosphorylation by Aurora-A in prophase is required for enrichment of Aurora-B at inner centromeres and for kinetochore function. Dev Cell 2003;5(6):853-64

82.

Perez de Castro I, Aguirre-Portoles C, Fernandez-Miranda G, et al. Requirements for Aurora-A in tissue regeneration and tumor development in adult mammals. Cancer Res 2013;73(23):6804-15 This work describes the effect that Aurora A genetic elimination has in adult tissues and in tumor growth.

.

83.

Cheung CH, Coumar MS, Chang JY, Hsieh HP. Aurora kinase inhibitor patents and agents in clinical testing: an update (2009-10). Expert Opin Ther Pat 2011;21(6):857-84

84.

Coumar MS, Cheung CH, Chang JY, Hsieh HP. Advances in Aurora kinase inhibitor patents. Expert Opin Ther Pat 2009;19(3):321-56

85.

Cheung CH, Coumar MS, Hsieh HP, Chang JY. Aurora kinase inhibitors in preclinical and clinical testing.

1389

rez de Castro M. Malumbres & I. Pe

Expert Opin Investig Drugs 2009;18(4):379-98 86.

95.

Green MR, Woolery JE, Mahadevan D. Update on Aurora kinase targeted therapeutics in oncology. Expert Opin Drug Discov 2011;6(3):291-307

Chefetz I, Holmberg JC, Alvero AB, et al. Inhibition of Aurora-A kinase induces cell cycle arrest in epithelial ovarian cancer stem cells by affecting NFkB pathway. Cell Cycle 2011;10(13):2206-14

Wang X, Sinn AL, Pollok K, et al. Preclinical activity of a novel multiple tyrosine kinase and aurora kinase inhibitor, ENMD-2076, against multiple myeloma. Br J Haematol 2010;150(3):313-25

96.

Tentler JJ, Bradshaw-Pierce EL, Serkova NJ, et al. Assessment of the in vivo antitumor effects of ENMD2076, a novel multitargeted kinase inhibitor, against primary and cell linederived human colorectal cancer xenograft models. Clin Cancer Res 2010;16(11):2989-98

97.

Fletcher GC, Brokx RD, Denny TA, et al. ENMD-2076 is an orally active kinase inhibitor with antiangiogenic and antiproliferative mechanisms of action. Mol Cancer Ther 2011;10(1):126-37

98.

Diamond JR, Bastos BR, Hansen RJ, et al. Phase I safety, pharmacokinetic, and pharmacodynamic study of ENMD2076, a novel angiogenic and Aurora kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res 2011;17(4):849-60

99.

91.

Matulonis UA, Lee J, Lasonde B, et al. ENMD-2076, an oral inhibitor of angiogenic and proliferation kinases, has activity in recurrent, platinum resistant ovarian cancer. Eur J Cancer 2013;49(1):121-31

92.

Diamond JR, Eckhardt SG, Tan AC, et al. Predictive biomarkers of sensitivity to the aurora and angiogenic kinase inhibitor ENMD-2076 in preclinical breast cancer models. Clin Cancer Res 2013;19(1):291-303

100. Minton S, LoRusso P, Lockhart A, et al. A phase I study of MK-5108, an oral aurora A kinase inhibitor, in both monotherapy and in combination with docetaxel in patients with advanced solid tumors. J Clin Oncol 2010;28(15_Suppl):abstract e13026

87.

88.

89.

90.

93.

94.

1390

Shiomitsu K, Sajo E, Rubin C, Sehgal I. The radiosensitizing effect of the aurora kinase inhibitors, ENMD-2076, on canine mast cell tumours in vitro. Vet Comp Oncol 2013. [Epub ahead of print] Shimomura T, Hasako S, Nakatsuru Y, et al. MK-5108, a highly selective Aurora-A kinase inhibitor, shows antitumor activity alone and in combination with docetaxel. Mol Cancer Ther 2010;9(1):157-66

Shan W, Akinfenwa PY, Savannah KB, et al. A small-molecule inhibitor targeting the mitotic spindle checkpoint impairs the growth of uterine leiomyosarcoma. Clin Cancer Res 2012;18(12):3352-65 Chinn DC, Holland WS, Mack PC. Anticancer activity of the Aurora A kinase inhibitor MK-5108 in non-small-cell lung cancer (NSCLC) in vitro as monotherapy and in combination with chemotherapies. J Cancer Res Clin Oncol 2014;140(7):1137-49 Horwacik I, Durbas M, Boratyn E, et al. Targeting GD2 ganglioside and aurora A kinase as a dual strategy leading to cell death in cultures of human neuroblastoma cells. Cancer Lett 2013;341(2):248-64 Kretzner L, Scuto A, Dino PM, et al. Combining histone deacetylase inhibitor vorinostat with aurora kinase inhibitors enhances lymphoma cell killing with repression of c-Myc, hTERT, and microRNA levels. Cancer Res 2011;71(11):3912-20

101. Manfredi MG, Ecsedy JA, Meetze KA, et al. Antitumor activity of MLN8054, an orally active small-molecule inhibitor of Aurora A kinase. Proc Natl Acad Sci USA 2007;104(10):4106-11 102. Shang X, Burlingame SM, Okcu MF, et al. Aurora A is a negative prognostic factor and a new therapeutic target in human neuroblastoma. Mol Cancer Ther 2009;8(8):2461-9 103. Hoar K, Chakravarty A, Rabino C, et al. MLN8054, a small-molecule inhibitor of Aurora A, causes spindle pole and chromosome congression defects leading to aneuploidy. Mol Cell Biol 2007;27(12):4513-25 104. Chakravarty A, Shinde V, Tabernero J, et al. Phase I assessment of new Expert Opin. Ther. Targets (2014) 18(12)

mechanism-based pharmacodynamic biomarkers for MLN8054, a smallmolecule inhibitor of Aurora A kinase. Cancer Res 2011;71(3):675-85 105. Dar AA, Belkhiri A, Ecsedy J, et al. Aurora kinase A inhibition leads to p73dependent apoptosis in p53-deficient cancer cells. Cancer Res 2008;68(21):8998-9004 106. Huck JJ, Zhang M, McDonald A, et al. MLN8054, an inhibitor of Aurora A kinase, induces senescence in human tumor cells both in vitro and in vivo. Mol Cancer Res 2010;8(3):373-84 . Reports for the first time how the inhibition Aurora kinase A induces senescence. 107. Macarulla T, Cervantes A, Elez E, et al. Phase I study of the selective Aurora A kinase inhibitor MLN8054 in patients with advanced solid tumors: safety, pharmacokinetics, and pharmacodynamics. Mol Cancer Ther 2010;9(10):2844-52 108. Dees EC, Infante JR, Cohen RB, et al. Phase 1 study of MLN8054, a selective inhibitor of Aurora A kinase in patients with advanced solid tumors. Cancer Chemother Pharmacol 2011;67(4):945-54 109. Manfredi MG, Ecsedy JA, Chakravarty A, et al. Characterization of Alisertib (MLN8237), an investigational smallmolecule inhibitor of aurora A kinase using novel in vivo pharmacodynamic assays. Clin Cancer Res 2011;17(24):7614-24 . Is a very good pre-clinical characterization of the Aurora kinase A inhibitor MLN8237. 110. Qi W, Spier C, Liu X, et al. Alisertib (MLN8237) an investigational agent suppresses Aurora A and B activity, inhibits proliferation, promotes endoreduplication and induces apoptosis in T-NHL cell lines supporting its importance in PTCL treatment. Leuk Res 2013;37(4):434-9 111. Tomita M, Mori N. Aurora A selective inhibitor MLN8237 suppresses the growth and survival of HTLV-1-infected T-cells in vitro. Cancer Sci 2010;101(5):1204-11 112. Wen Q, Goldenson B, Silver SJ, et al. Identification of regulators of polyploidization presents therapeutic targets for treatment of AMKL. Cell 2012;150(3):575-89

Aurora kinase A inhibitors

113.

114.

G€orgu¨n G, Calabrese E, Hideshima T, et al. A novel Aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cellcycle arrest in multiple myeloma. Blood 2010;115(25):5202-13

with the investigational agent alisertib increases the efficacy of cytarabine through a FOXO-dependent mechanism. Int J Cancer 2012;131(11):2693-703 124.

Do TV, Xiao F, Bickel LE, et al. Aurora kinase A mediates epithelial ovarian cancer cell migration and adhesion. Oncogene 2014;33(5):539-49

115.

Zhou N, Singh K, Mir MC, et al. The investigational Aurora kinase A inhibitor MLN8237 induces defects in cell viability and cell-cycle progression in malignant bladder cancer cells in vitro and in vivo. Clin Cancer Res 2013;19(7):1717-28

116.

Patel AV, Eaves D, Jessen WJ, et al. Ras-driven transcriptome analysis identifies aurora kinase A as a potential malignant peripheral nerve sheath tumor therapeutic target. Clin Cancer Res 2012;18(18):5020-30

125.

Qi W, Cooke LS, Liu X, et al. Aurora inhibitor MLN8237 in combination with docetaxel enhances apoptosis and antitumor activity in mantle cell lymphoma. Biochem Pharmacol 2011;81(7):881-90 Sehdev V, Katsha A, Ecsedy J, et al. The combination of alisertib, an investigational Aurora kinase A inhibitor, and docetaxel promotes cell death and reduces tumor growth in preclinical cell models of upper gastrointestinal adenocarcinomas. Cancer 2013;119(4):904-14

126.

Venkataraman S, Alimova I, Tello T, et al. Targeting Aurora Kinase A enhances radiation sensitivity of atypical teratoid rhabdoid tumor cells. J Neurooncol 2012;107(3):517-26

Asteriti IA, Giubettini M, Lavia P, Guarguaglini G. Aurora-A inactivation causes mitotic spindle pole fragmentation by unbalancing microtubule-generated forces. Mol Cancer 2011;10:131

127.

Brewer Savannah KJ, Demicco EG, Lusby K, et al. Dual targeting of mTOR and aurora-A kinase for the treatment of uterine Leiomyosarcoma. Clin Cancer Res 2012;18(17):4633-45

118.

Liu Y, Hawkins OE, Su Y, et al. Targeting aurora kinases limits tumour growth through DNA damage-mediated senescence and blockade of NF-kappaB impairs this drug-induced senescence. EMBO Mol Med 2013;5(1):149-66

128.

119.

Lehman NL, O’Donnell JP, Whiteley LJ, et al. Aurora A is differentially expressed in gliomas, is associated with patient survival in glioblastoma and is a potential chemotherapeutic target in gliomas. Cell Cycle 2012;11(3):489-502

Mahadevan D, Stejskal A, Cooke LS, et al. Aurora A inhibitor (MLN8237) plus vincristine plus rituximab is synthetic lethal and a potential curative therapy in aggressive B-cell non-Hodgkin lymphoma. Clin Cancer Res 2012;18(8):2210-19

129.

Sehdev V, Peng D, Soutto M, et al. The aurora kinase A inhibitor MLN8237 enhances cisplatin-induced cell death in esophageal adenocarcinoma cells. Mol Cancer Ther 2012;11(3):763-74

130.

Muscal JA, Scorsone KA, Zhang L, et al. Additive effects of vorinostat and MLN8237 in pediatric leukemia, medulloblastoma, and neuroblastoma cell lines. Invest New Drugs 2013;31(1):39-45

117.

120.

121.

122.

123.

Marxer M, Ma HT, Man WY, Poon RY. p53 deficiency enhances mitotic arrest and slippage induced by pharmacological inhibition of Aurora kinases. Oncogene 2014;33(27):3550-60 Neel NF, Stratford JK, Shinde V, et al. Response to MLN8237 in pancreatic cancer is not dependent on RalA phosphorylation. Mol Cancer Ther 2014;13(1):122-33 Kelly KR, Ecsedy J, Medina E, et al. The novel Aurora A kinase inhibitor MLN8237 is active in resistant chronic myeloid leukaemia and significantly increases the efficacy of nilotinib. J Cell Mol Med 2011;15(10):2057-70 Kelly KR, Nawrocki ST, Espitia CM, et al. Targeting Aurora A kinase activity

131.

.

132.

Dees EC, Cohen RB, von Mehren M, et al. Phase I study of aurora A kinase inhibitor MLN8237 in advanced solid tumors: safety, pharmacokinetics, pharmacodynamics, and bioavailability of two oral formulations. Clin Cancer Res 2012;18(17):4775-84 Describe the monotherapy phase I clinical trials performed for Alisertib. Cervantes A, Elez E, Roda D, et al. Phase I pharmacokinetic/ pharmacodynamic study of MLN8237, Expert Opin. Ther. Targets (2014) 18(12)

.

an investigational, oral, selective aurora a kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res 2012;18(17):4764-74 Describe the monotherapy phase I clinical trials performed for Alisertib.

133. Mosse YP, Lipsitz E, Fox E, et al. Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective smallmolecule inhibitor of Aurora kinase A: a Children’s Oncology Group Phase I Consortium study. Clin Cancer Res 2012;18(21):6058-64 . Describe the monotherapy phase I clinical trials performed for Alisertib. 134. Kelly KR, Shea TC, Goy A, et al. Phase I study of MLN8237investigational Aurora A kinase inhibitorin relapsed/refractory multiple myeloma, Non-Hodgkin lymphoma and chronic lymphocytic leukemia. Invest New Drugs 2014;32(3):489-99 . Describe the monotherapy phase I clinical trials performed for Alisertib. 135. Friedberg JW, Mahadevan D, Cebula E, et al. Phase II study of alisertib, a selective Aurora A kinase inhibitor, in relapsed and refractory aggressive B- and T-cell non-Hodgkin lymphomas. J Clin Oncol 2014;32(1):44-50 .. Correspond to the two phase II clinical trials reported for Alisertib in advanced malignancies. 136. Matulonis UA, Sharma S, Ghamande S, et al. Phase II study of MLN8237 (alisertib), an investigational Aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol 2012;127(1):63-9 .. Correspond to the two phase II clinical trials reported for Alisertib in advanced malignancies. 137. Hrabakova R, Kollareddy M, Tyleckova J, et al. Cancer cell resistance to aurora kinase inhibitors: identification of novel targets for cancer therapy. J Proteome Res 2013;12(1):455-69 138. Zou Z, Yuan Z, Zhang Q, et al. Aurora kinase A inhibition-induced autophagy triggers drug resistance in breast cancer cells. Autophagy 2012;8(12):1798-810 139. Kesisova IA, Nakos KC, Tsolou A, et al. Tripolin A, a novel small-molecule inhibitor of aurora A kinase, reveals new regulation of HURP’s distribution on

1391

rez de Castro M. Malumbres & I. Pe

microtubules. PLoS One 2013;8(3):e58485 140. Palani S, Patel M, Huck J, et al. Preclinical pharmacokinetic/ pharmacodynamic/efficacy relationships for alisertib, an investigational smallmolecule inhibitor of Aurora A kinase. Cancer Chemother Pharmacol 2013;72(6):1255-64 141. Jeon HY, Lee H. Depletion of Aurora-A in zebrafish causes growth retardation due to mitotic delay and p53-dependent cell death. FEBS J 2013;280(6):1518-30 142. Zhao B, Smallwood A, Yang J, et al. Modulation of kinase-inhibitor interactions by auxiliary protein binding: crystallography studies on Aurora A interactions with VX-680 and with TPX2. Protein Sci 2008;17(10):1791-7 143. Ice RJ, McLaughlin SL, Livengood RH, et al. NEDD9 depletion destabilizes Aurora A kinase and heightens the efficacy of Aurora A inhibitors: implications for treatment of metastatic solid tumors. Cancer Res 2013;73(10):3168-80 144. Shionome Y, Lin WH, Shiao HY, et al. A novel aurora-A inhibitor, BPR1K0609S1, sensitizes colorectal tumor cells to 5-fluorofracil (5-FU) treatment. Int J Biol Sci 2013;9(4):403-11 145. Tang YC, Williams BR, Siegel JJ, Amon A. Identification of aneuploidyselective antiproliferation compounds. Cell 2011;144(4):499-512 146. Toledo LI, Murga M, Zur R, et al. A cell-based screen identifies ATR inhibitors with synthetic lethal properties for cancer-associated mutations. Nat Struct Mol Biol 2011;18(6):721-7 147. Siggelkow W, Boehm D, Gebhard S, et al. Expression of aurora kinase A is associated with metastasis-free survival in node-negative breast cancer patients. BMC Cancer 2012;12:562 148. Nadler Y, Camp RL, Schwartz C, et al. Expression of Aurora A (but not Aurora B) is predictive of survival in breast cancer. Clin Cancer Res 2008;14(14):4455-62 149. Staff S, Isola J, Jumppanen M, Tanner M. Aurora-A gene is frequently amplified in basal-like breast cancer. Oncol Rep 2010;23(2):307-12 150. Miyoshi Y, Iwao K, Egawa C, Noguchi S. Association of centrosomal

1392

kinase STK15/BTAK mRNA expression with chromosomal instability in human breast cancers. Int J Cancer 2001;92(3):370-3 151. Goos JA, Coupe VM, Diosdado B, et al. Aurora kinase A (AURKA) expression in colorectal cancer liver metastasis is associated with poor prognosis. Br J Cancer 2013;109(9):2445-52

count in patients with acute myeloid leukemia. Leuk Res 2011;35(2):260-4 161. Xu HT, Ma L, Qi FJ, et al. Expression of serine threonine kinase 15 is associated with poor differentiation in lung squamous cell carcinoma and adenocarcinoma. Pathol Int 2006;56(7):375-80

152. Dotan E, Meropol NJ, Zhu F, et al. Relationship of increased aurora kinase A gene copy number, prognosis and response to chemotherapy in patients with metastatic colorectal cancer. Br J Cancer 2012;106(4):748-55

162. Ogawa E, Takenaka K, Katakura H, et al. Perimembrane Aurora-A expression is a significant prognostic factor in correlation with proliferative activity in non-small-cell lung cancer (NSCLC). Ann Surg Oncol 2008;15(2):547-54

153. Sakakura C, Hagiwara A, Yasuoka R, et al. Tumour-amplified kinase BTAK is amplified and overexpressed in gastric cancers with possible involvement in aneuploid formation. Br J Cancer 2001;84(6):824-31

163. Lo Iacono M, Monica V, Saviozzi S, et al. Aurora Kinase A expression is associated with lung cancer histologicalsubtypes and with tumor de-differentiation. J Transl Med 2011;9:100

154. Yen CC, Yeh CN, Cheng CT, et al. Integrating bioinformatics and clinicopathological research of gastrointestinal stromal tumors: identification of aurora kinase A as a poor risk marker. Ann Surg Oncol 2012;19(11):3491-9

164. Neben K, Korshunov A, Benner A, et al. Microarray-based screening for molecular markers in medulloblastoma revealed STK15 as independent predictor for survival. Cancer Res 2004;64(9):3103-11

155. Reiter R, Gais P, Ju¨tting U, et al. Aurora kinase A messenger RNA overexpression is correlated with tumor progression and shortened survival in head and neck squamous cell carcinoma. Clin Cancer Res 2006;12(17):5136-41 156. Tatsuka M, Sato S, Kanda A, et al. Oncogenic role of nuclear accumulated Aurora-A. Mol Carcinog 2009;48(9):810-20

165. Reichardt W, Jung V, Brunner C, et al. The putative serine/threonine kinase gene STK15 on chromosome 20q13.2 is amplified in human gliomas. Oncol Rep 10(5):1275-9 166. Klein A, Reichardt W, Jung V, et al. Overexpression and amplification of STK15 in human gliomas. Int J Oncol 2004;25(6):1789-94

157. Kitajima S, Kudo Y, Ogawa I, et al. Constitutive phosphorylation of aurora-a on ser51 induces its stabilization and consequent overexpression in cancer. PLoS One 2007;2(9):e944

167. Moreno-Bueno G, Sa´nchez-Estevez C, Cassia R, et al. Differential gene expression profile in endometrioid and nonendometrioid endometrial carcinoma: STK15 is frequently overexpressed and amplified in nonendometrioid carcinomas. Cancer Res 2003;63(18):5697-702

158. Camacho E, Bea S, Salaverrı´a I, et al. Analysis of Aurora-A and hMPS1 mitotic kinases in mantle cell lymphoma. Int J Cancer 2006;118(2):357-63

168. Gritsko TM, Coppola D, Paciga JE, et al. Activation and overexpression of centrosome kinase BTAK/Aurora-A in human ovarian cancer. Clin Cancer Res 2003;9(4):1420-6

159. Inamdar KV, O’Brien S, Sen S, et al. Aurora-A kinase nuclear expression in chronic lymphocytic leukemia. Mod Pathol 2008;21(12):1428-35

169. Landen CN, Lin YG, Immaneni A, et al. Overexpression of the centrosomal protein Aurora-A kinase is associated with poor prognosis in epithelial ovarian cancer patients. Clin Cancer Res 2007;13(14):4098-104

160. Lucena-Araujo AR, de Oliveira FM, Leite-Cueva SD, et al. High expression of AURKA and AURKB is associated with unfavorable cytogenetic abnormalities and high white blood cell

Expert Opin. Ther. Targets (2014) 18(12)

170. Lassus H, Staff S, Leminen A, et al. Aurora-A overexpression and aneuploidy predict poor outcome in serous ovarian

Aurora kinase A inhibitors

correlates with clinical and pathological parameters in bladder neoplasms. Histol Histopathol 2010;25(11):1371-7

carcinoma. Gynecol Oncol 2011;120(1):11-17 171.

172.

173.

174.

175.

176.

Mendiola M, Barriuso J, Marin˜o-Enrı´quez A, et al. Aurora kinases as prognostic biomarkers in ovarian carcinoma. Hum Pathol 2009;40(5):631-8 Li D, Zhu J, Firozi PF, et al. Overexpression of oncogenic STK15/ BTAK/Aurora A kinase in human pancreatic cancer. Clin Cancer Res 2003;9(3):991-7 Zhu J, Abbruzzese JL, Izzo J, et al. AURKA amplification, chromosome instability, and centrosome abnormality in human pancreatic carcinoma cells. Cancer Genet Cytogenet 2005;159(1):10-17 Bar-Shira A, Pinthus JH, Rozovsky U, et al. Multiple genes in human 20q13 chromosomal region are involved in an advanced prostate cancer xenograft. Cancer Res 2002;62(23):6803-7 Buschhorn HM, Klein RR, Chambers SM, et al. Aurora-A overexpression in high-grade PIN lesions and prostate cancer. Prostate 2005;64(4):341-6 Torchia EC, Chen Y, Sheng H, et al. A genetic variant of Aurora kinase A promotes genomic instability leading to highly malignant skin tumors. Cancer Res 2009;69(18):7207-15

177.

Sen S, Zhou H, Zhang RD, et al. Amplification/overexpression of a mitotic kinase gene in human bladder cancer. J Natl Cancer Inst 2002;94(17):1320-9

178.

Lei Y, Yan S, Ming-De L, et al. Prognostic significance of Aurora-A expression in human bladder cancer. Acta Histochem 2011;113(5):514-18

179.

Bufo P, Sanguedolce F, Tortorella S, et al. Expression of mitotic kinases phospho-aurora A and aurora B

180.

Giet R, McLean D, Descamps S, et al. Drosophila Aurora A kinase is required to localize D-TACC to centrosomes and to regulate astral microtubules. J Cell Biol 2002;156(3):437-51

181.

Furuta T, Baillie DL, Schumacher JM. Caenorhabditis elegans Aurora A kinase AIR-1 is required for postembryonic cell divisions and germline development. Genesis 2002;34(4):244-50

182.

Liu Q, Ruderman JV. Aurora A, mitotic entry, and spindle bipolarity. Proc Natl Acad Sci USA 2006;103(15):5811-16

183.

Yang H, Burke T, Dempsey J, et al. Mitotic requirement for aurora A kinase is bypassed in the absence of aurora B kinase. FEBS Lett 2005;579(16):3385-91

184.

Pratz KW, Cortes J, Roboz GJ, et al. A pharmacodynamic study of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical response. Blood 2009;113(17):3938-46

185.

Traynor AM, Hewitt M, Liu G, et al. Phase I dose escalation study of MK0457, a novel Aurora kinase inhibitor, in adult patients with advanced solid tumors. Cancer Chemother Pharmacol 2011;67(2):305-14

186.

Giles FJ, Swords RT, Nagler A, et al. MK-0457, an Aurora kinase and BCRABL inhibitor, is active in patients with BCR-ABL T315I leukemia. Leukemia 2013;27(1):113-17

187.

Meulenbeld HJ, Bleuse JP, Vinci EM, et al. Randomized phase II study of danusertib in patients with metastatic castration-resistant prostate cancer after docetaxel failure. BJU Int 2013;111(1):44-52

188.

Dent SF, Gelmon KA, Chi KN, et al. NCIC CTG IND.181: phase I study of

AT9283 given as a weekly 24 hour infusion in advanced malignancies. Invest New Drugs 2013;31(6):1522-9 189. Foran J, Ravandi F, Wierda W, et al. A phase I and pharmacodynamic study of AT9283, a small-molecule inhibitor of aurora kinases in patients with relapsed/ refractory leukemia or myelofibrosis. Clin Lymphoma Myeloma Leuk 2014;14(3):223-30 190. Arkenau HT, Plummer R, Molife LR, et al. A phase I dose escalation study of AT9283, a small molecule inhibitor of aurora kinases, in patients with advanced solid malignancies. Ann Oncol 2012;23(5):1307-13 191. Sch€offski P, Jones SF, Dumez H, et al. Phase I, open-label, multicentre, doseescalation, pharmacokinetic and pharmacodynamic trial of the oral aurora kinase inhibitor PF-03814735 in advanced solid tumours. Eur J Cancer 2011;47(15):2256-64

Affiliation Marcos Malumbres*1 PhD & Ignacio Perez de Castro†2 PhD †,* Authors for correspondence 1 Group Leader, Spanish National Cancer Research Centre (CNIO), Cell Division and Cancer Group, Melchor Ferna´ndez Almagro 3, E-28029 Madrid, Spain Tel: +34 91 224 6900; Fax: +34 91 732 8033; E-mail: [email protected] 2 Staff Scientist, Spanish National Cancer Research Centre (CNIO), Cell Division and Cancer Group, Melchor Ferna´ndez Almagro 3, E-28029 Madrid, Spain Tel: +34 91 224 6900; Fax: +34 91 732 8033; E-mail: [email protected]

Supplementary materials available online Tables S1, S2 and S3.

Expert Opin. Ther. Targets (2014) 18(12)

1393

Aurora kinase A inhibitors: promising agents in antitumoral therapy.

Aurora proteins are serine/threonine kinases with critical functions during mitosis. Aurora A, one of the members of this family, participates in cruc...
505KB Sizes 1 Downloads 4 Views