Open Access

The Relevance of Aurora Kinase Inhibition in Hematological Malignancies

CAIO BEZERRA MACHADO 1
EMERSON LUCENA DA SILVA 1
BEATRIZ MARIA DIAS NOGUEIRA 1
JEAN BRENO SILVEIRA DA SILVA 1
MANOEL ODORICO DE MORAES FILHO 1
RAQUEL CARVALHO MONTENEGRO 1
MARIA ELISABETE AMARAL DE MORAES 1
  &  
CAROLINE AQUINO MOREIRA-NUNES 1

Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil

Cancer Diagnosis & Prognosis Jul-Aug; 1(3): 111-126 DOI: 10.21873/cdp.10016
Received 09 April 2021 | Revised 10 December 2024 | Accepted 27 April 2021
Corresponding author
Caroline Aquino Moreira-Nunes, Federal University of Ceará, Coronel Nunes de Melo st, n 1000, Rodolfo Teófilo, CEP: 60416-000 Fortaleza, CE, Brazil. Tel: +55 8533668033 carolfam@gmail.com
pdf image icon

Abstract

Aurora kinases are a family of serine/threonine protein kinases that play a central role in eukaryotic cell division. Overexpression of aurora kinases in cancer and their role as major regulators of the cell cycle quickly inspired the idea that their inhibition might be a potential pathway when treating oncologic patients. Over the past couple of decades, the search for designing and testing of molecules capable of inhibiting aurora activities fueled many pre-clinical and clinical studies. In this study, data from the past 10 years of in vitro and in vivo investigations, as well as clinical trials, utilizing aurora kinase inhibitors as therapeutics for hematological malignancies were compiled and discussed, aiming to highlight potential uses of these inhibitors as a novel monotherapy model or alongside conventional chemotherapies. While there is still much to be elucidated, it is clear that these kinases play a key role in oncogenesis, and their manageable toxicity and potentially synergistic effects still render them a focus of interest for future investigations in combinatorial clinical trials.
Keywords: Hematological neoplasms, aurora kinases, targeted molecular therapy, review

In 2017 alone, nearly 24.5 million new cases of cancer were diagnosed worldwide, and more than 9.5 million people died from the disease. Cancer can be described as a heterogeneous set of diseases that share the main clinical characteristic of uncontrolled cell proliferation in the form of a solid tumor or as individual cells in the bloodstream (1,2).

Even though cancer is characterized as a multifactorial disease, the accumulation of genetic mutations, as well as the deregulation of the cellular cycle, are both well-established occurrences in cancerous cells. Failure in the regulation of the cell cycle is associated with malignant neoplasms since it is fundamental for genome maintenance, repairing DNA damage and, consequently, preventing the proliferation of cells with malignant potential (3,4).

As one of the main factors in cell-cycle control, the activity of protein kinases is to be highlighted due to their vital roles in several phases of cell division. In recent years, it has been demonstrated that alterations in cellular expression levels of many kinases are associated with malignant cancer phenotypes, pointing them out as possible therapeutic targets for these diseases. One such kinase family of possible clinical relevance are the aurora kinases (5,6).

Role of Aurora Kinases in Cell Division

The aurora kinases are a family of serine/threonine protein kinases that play a central role in eukaryotic cell division. Humans express three different types of aurora kinases: aurora kinase A (AURKA), aurora kinase B (AURKB) and aurora kinase C (AURKC). AURKA and AURKB are the most relevant to mitosis, being expressed in most somatic cell types, while AURKC is mostly expressed in germ cells due to its role in meiosis regulation (7,8).

From the first reports of them being essential in cell division to the recent years of thorough molecular studies, the auroras have been described as oncogenes relevant to many human malignancies (9,10). Their gene amplification and overexpression is detected particularly in solid tumors, however, their correlation with leukemia development has also been reported (10,11).

The family name as aurora was conceived when it was first discovered that aurora-mutated Drosophila cells failed to properly duplicate and separate their centrosomes during mitosis, inducing the formation of monopolar spindles that, when observed under a microscope, would bear resemblance to the aurora borealis phenomenon (12). Later studies led to the determination of the localization of these kinases (Figure 1) and their actual cellular roles in humans.

AURKA

Prior to bipolar spindle formation, from late S phase to pro-metaphase, AURKA is mainly concentrated at centrosomes (13,14) due to an increase in cyclin-dependent kinase 11 (CDK11) activity (15). AURKA is then a part of important processes for centrosome maturation such as pericentriolar matrix recruitment and microtubule nucleation and stabilization (16,17).

The role of AURKA in maturation occurs through phosphorylation loops and protein-protein interactions with many different kinases. One such pathway is AURKA-mediated phosphorylation of a large tumor suppressor 2 (LATS2) and ajuba LIM protein (AJUBA) complex which acts on an auto-phosphorylation loop that directs AURKA to the centrosomes and induces the recruitment of a centrosomal pool of γ-tubulin, an essential step for microtubule nucleation and later spindle assembly (17-20).

During metaphase and throughout mitosis, the bipolar spindle assembly begins and AURKA also localizes to spindle microtubules, as well as the centrosomes (21,22). AURKA localization to spindle microtubules is dependent on activity of TPX2 microtubule nucleation factor (TPX2) which binds to and activates AURKA at the spindles, inhibiting dephosphorylation by protein phosphatase 1 (PP1) (22-24).

Aurora-related proteins have been shown to phosphorylate kinesin family member 11 (EG5), an important microtubule-motor protein responsible for centrosome separation, in the Xenopus laevis model, thus conferring it a major role in bipolar spindle formation (25,26). However, it has been demonstrated that AURKA is also involved in EG5-independent pathways for bipolar spindle assembly through phosphorylation of another motor protein, kinesin-12 (KIF15/HKLP2), in human cell models (27,28).

Besides its role in cellular spatial organization, AURKA also exerts a series of nuclear functions related to the control of mitotic checkpoint G2/M (Figure 2), making it a key factor in initiation of mitosis (20,29).

AURKB

Along with the regulatory proteins inner centromere protein, survivin and borealin, AURKB forms the chromosomal passenger complex (CPC) and takes part in regulating crucial pathways for chromosome condensation, sister chromatid separation and cytokinesis (30).

During the G2 phase, at mitosis onset, AURKB is responsible for phosphorylation of histone H3 (Figure 2), an event that coincides with heterochromatin condensation and proper chromosome formation (31-33). In vitro experiments utilizing analogous animal models have demonstrated AURKA to be a better kinase for the phosphorylation of H3 than AURKB, however, the intracellular colocalization of AURKB and phosphorylated H3, and the knowledge that a decrease in AURKB level implicates in reduced H3 phosphorylation, leads to the understanding that in vivo models favor AURKB activity for histone phosphorylation (33,34).

After chromosome condensation, during prophase in early mitosis, the activation of phosphorylated histones H3 and H2A recruits AURKB to the centromeres (35-37). It has been suggested that recruitment of AURKB happens in a two-step process where H2A phosphorylation by BUB1 mitotic checkpoint serine/threonine protein kinase (BUB1) firstly creates a pool of kinetochore-bound AURKB and then H3 phosphorylation by haspin kinase translocates this AURKB pool to the inner centromere, where it is mainly located during prophase (38).

Until the onset of anaphase, AURKB phosphorylation of the KMN network, an association of the conserved kinetochore proteins kinetochore null protein 1 (KNL1), mis-segregation 12 (MIS12) and nuclear division cycle 80 (NDC80), takes part in regulating kinetochore-microtubule attachment stabilization (39,40). While during early mitosis the attachment turnover is high, ensuring quickly destabilization of incorrect attachments, it tends to slow down alongside mitosis progression after correct and stable attachments are formed and the spindle assembly checkpoint is satisfied, allowing for increased activity of anaphase-promoting complex/cyclosome and proper chromosomal separation in anaphase (38,40-42).

AURKB also exerts major roles in cytokinesis, from initial anaphase through telophase and the end of mitosis, being relocated from inner centromeres to the spindle midzone by the action of mitotic kinesin MKLP2 and then interacting with kinesins KIF4A and KIF2A to promote microtubule bundling and consequent central spindle formation, as well as proper cleavage furrow completion (43-45). It has been shown that after chromosomaI segregation, the activity of AURKB at the spindle midzone functions as a checkpoint for the regulation of incorrectly segregated chromatin, inhibiting the formation of tetraploid daughter cells and protecting lagging chromosomes from breakage (45,46).

AURKC

The activity and metabolic role of AURKC is still poorly understood when compared to the other two aurora kinases expressed by humans, being mostly described by its overlapping functions with AURKB (47,48). Although poorly expressed in most somatic cells, high AURKC expression in germ cells and its ability to interact with chromosomal passenger complex proteins has led to the understanding that AURKC exerts many functions that overlap with activities of AURKB in meiotic cells, being responsible for the regulation of kinetochore-microtubule attachment and proper sister chromatid segregation (48,49).

AURKC has also been observed to be highly expressed in pre-implantation embryo cells, being able to carry out normal cell division in AURKB-null mice with death occurring only after implantation takes place, making it clear that AURKC may be relevant in the division of somatic cells (50,51). While the non-overlapping functions of AURKB and AURKC are not fully elucidated, both kinases are known to form distinct complexes with chromosomal passenger complex proteins and act through distinct mechanisms, while still being able to interfere with each other’s activities, as shown by experiments of selective inhibition of one or both kinases (52).

Aurora Family as Targets in Oncohematological Therapy

The finding of overexpression of aurora kinases in cancer and their role as major regulators of the cell cycle quickly led to the idea that their inhibition might be a potential pathway when treating oncological patients. Over the past couple of decades, the quest to design and test molecules capable of inhibiting aurora activities fueled many pre-clinical and clinical studies, resulting in the development of a series of pan-aurora inhibitors as well as selective inhibitors of one of the three human kinases (53). While some aurora kinase inhibitors (AKIs) demonstrate promising results as single treatments, it is also important to perceive their potential as synergistic compounds, being able to restore sensitivity to chemotherapy agents in tumor cells and interact with other targeted therapies for increased efficacy (54).

In this scenario, hematological malignant disorders as leukemia presents overexpression of AURK, and the use of AKIs seems to be an effective pharmacological approach to treating these diseases (14). Table I presents studies published in the past 10 years describing the mechanism of action of several AKIs in different leukemia cell lines, as well as in in vivo models, and validation of AURKA and B overexpression in patient samples. The studies showed that the antileukemic effect of AKI AURKA and AURKB are consistently related to G2/M cell-cycle arrest, modulation of the expression of cell cycle regulators, and polyploidy induction resulting in apoptotic cell death.

AURKA and AURKB have an important role in cell-cycle progression, in fact, the transcription of AURK is cell cycle-regulated. AURKA mRNA is found at higher concentrations in the G2/M phase, its protein level reaching higher a little later; the same happens to AURKB, with enhanced levels of mRNA and protein expression just after those of AURKA (14). Inhibition of AURKA expression by genetic knockdown was shown to lead to errors in cell mitotic processes, once the spindle checkpoint detects a failure in chromosome alignment, it culminates in mitotic arrest and cellular death by apoptosis (71). The knockdown of AURKB leads to reduction in histone H3 phosphorylation required in chromosome condensation and cytokinesis, these errors are associated with abnormal segregation, resulting in polyploid cells (34). Otherwise, cells lacking both genes are able to finish mitosis without completing anaphase, resulting in polyploid cells with just one nucleus (72).

Most studies evaluating the mechanism of AKI action in hematologic cell lines focus on acute myeloid leukemia (AML). This neoplasia is characterized by clonal expansion of undifferentiated myeloid precursors, leading to impaired hematopoiesis and bone marrow failure (73). In in vitro studies, AKIs were evaluated alone or in combination with cytarabine, a nucleoside analog that is incorporated into DNA in cell replication (S-phase) (74). The synergism of cytarabine with pan-AKI AMG 900 enhanced the antileukemic effect and attenuated polyploidization (60). In another study, the AURKB inhibitor barasertib used in combination with cytarabine showed a greater-than-additive cytotoxic effect with evidence of apoptosis, since barasertib does not prevent DNA synthesis, enabling cytarabine incorporation into DNA (66).

Internal tandem duplication of FMS-like receptor tyrosine kinase 3 (FLT3) is one of the most common somatic mutations characterized in AML. It results in the constant activation of FLT3 kinase, which is linked to cancer resurgence and poor patient survival (75). The dual FLT3 inhibitor/AKI CCT241736 showed promising antileukemic effect in vitro and in vivo, as well as in primary samples from patients with AML, including those who were resistant to the FLT3 inhibitor quizatinib, indicating that dual inhibition can be a tool to manage therapies in patients with AML presenting internal tandem duplication of FLT3 with resistance to current chemotherapy (56).

Translocation t(8;21) is frequent in 4-8% of patients with AML and results in the chimeric protein RUNX family transcription factor 1-RUNX1 partner transcriptional co-repressor 1 (RUNX1-RUNX1T1); 40-60% of patients harboring this translocation are susceptible to relapse after complete remission, this being one of the major causes of treatment failure (76-78). Qi et al. showed that AURKB inhibitor barasertib was more successful in inhibiting proliferation of AML cell lines with t(8;21) than cytarabine and AURKA inhibitor alisertib, and combination of the two AKIs did not show a synergistic effect, indicating that AURKB inhibitor may be a more efficient inhibitor in the treatment of patients with a specific subtype of AML (58).

Acute lymphoblastic leukemia (ALL) is the most common leukemia diagnosed in adults and the second most frequent acute subtype in children worldwide, this neoplasia presents chromosomal and genetic alterations related to the differentiation and proliferation of T and B precursor cells (79,80). AURKA and AURKB are overexpressed in samples from patients with ALL (55,65,81) and some research described the mechanism of action of AKI in ALL cell lines alone or in combination with other antineoplastics agents.

Genetic alterations as t(4;11) with mixed lineage leukemia (MLL) fusion–associated gene AF4 (MLL-AF4) and t(9;22) with breakpoint cluster region–Abelson murine leukemia 1 (BCR–ABL1) are frequent in ALL and are related to treatment resistance, poor outcome, and disease relapse (65,82). Rearrangements of MLL gene represents 10% of ALL cases and MLL–AF4 fusion is present in the majority of ALL cases (57%), is highly associated with rapid cancer development, fast progression, and poor prognosis in comparison with patients without this rearrangement (83,84).

In in vitro studies evaluating the cytotoxicity of the pan-AKIs VX-680 and VE-465 against a panel of ALL cell lines, cell lines with MLL–AF4 fusion showed higher sensitivity to AKI treatment, and VE-465 gave better results when compared to VX-68. Curiously, the same study reported that these cell lines presented AURKB protein expression higher than AURKA (65). A recent study conducted by Moreira-Nunes et al. observed no difference between AURKA and AURKB gene expression in samples with MLL–AF4 fusion from patients with ALL when compared to other frequent fusions in leukemia [BCR–ABL1, transcription factor 3 (E2A immunoglobulin enhancer-binding factors E12/E47) and pre-B-cell leukemia transcription factor 1 (E2A–PBX1) translocation, stem leukemia cell interrupting locus and T-cell acute lymphocytic leukemia protein 1 (SIL–TAL1) fusion, translocation–Ets–leukemia and acute myeloid leukemia 1 protein (TEL–AML1) fusion]. In contrast, when the expression of AUKA and B were analyzed in all patients in the study, AURKB expression was higher than that of AURKA and overexpression was correlated with lower survival rates (55). Thus, due to the role of AURKB in ALL, and even the lack of evidence that correlates the expression of this kinase with outcome in those with MLL–AF4, more studies evaluating AURKB inhibitors in therapy of MLL–AF4 ALL are still necessary.

The Philadelphia chromosome (Ph+) is characterized by the translocation between chromosomes 9 and 22, generating the chimeric gene BCR–ABL1 (85,86). Although this translocation is more described in chronic myeloid leukemia (CML), the presence of Ph affects around 3-5% of children and 25% of adults with ALL, and is related to a poor prognosis (87-89). The chimeric BCR–ABL1 gene is constitutively active and overexpresses a tyrosine-kinase receptor that is involved in signaling pathways linked to cellular proliferation, de-differentiation, and apoptosis escape (90). Although tyrosine-kinase inhibitors (TKIs) such as imatinib, bosutinib, dasatinib, and nilotinib were developed, mutations in catalytic enzymatic domains as ABL1T315I result in TKI resistance, leading to risk of treatment failure in patients with CML or ALL Ph+ (67,91-93).

Due to the efficacy of AKIs in leukemia cell lines in vitro and in in vivo studies, Fei et al. evaluated the pan-AKI danusertib (PHA-739358) in human ALL cell lines with and without BCR–ABL1T315I mutation, and also in ALL Ph− cell lines. Danusertib induced cytotoxicity in all cells independently of BCR–ABL1T315I mutation and Ph presence, as well as reducing the percentage of leukemia cells (CD10+/CD19+) and enhancing the survival rates of mice transplanted with cells with BCR–ABL1T315I mutation (67). The study also attested to synergism in the cytotoxic effect of the pan-AKI danusertib in combination with farnesyltransferase inhibitor, vincristine, and with the TKI dasatinib independently of BCR–ABL1T315I mutation, suggesting danusertib as a new option for chemotherapy in patients with ALL, since it is effective independently of Ph presence or BCR–ABL1T315I mutation, and, especially for patients with BCR–ABL1T315I mutation who are resistant to actual treatment with TKIs.

CML is basically, but not only, characterized by the presence of Ph+ cells, since around 95% of cases present this translocation (94,95). Thus, the malignant potential of CML cells is related to the BCR–ABL1 chimeric gene, which is also its pharmacologic target (85,96). Resistance to TKIs in CML is widely documented and occurs in 20-40% of cases (97), furthermore, the overexpression of AURKA in several cancer types is described as one mechanism of chemoresistance (98-104).

In vitro studies monitored the antileukemia effect of pan-AKIs GW809897X and GW806742X in a non-resistant CML cell line, with promising results (55). Long et al. showed that AURKA inhibitor AKI603 suppressed cell growth and induced cell differentiation of both imatinib-resistant (BCR–ABL1T315I) and non-resistant cell lines (64). A similar study showed that AURKA inhibitor alisertib, when used alone, reduced viability of CML cell lines, both non-resistant and resistant to imatinib and nilotinib, as well as cells harboring BCR–ABL1T315I mutations. Moreover, when administered in combination with the TKIs imatinib, ponatinib or nilotinib, there was a disruption in the resistance mechanism and reduced rates of cell growth were observed in the respective resistant cell lines. Moreover, alisertib plus ponatinib showed an additive effect in reducing tumor volume and enhanced the rate of survival in mice transplanted with CML BCR–ABL1T315I+ cells (61). Inhibition of AURKA seems to reduce the profile of resistance in CML cell lines in vitro and in vivo; the utilization of AKIs in combination with conventional chemotherapy may improve the outcome of patients that are unresponsive to chemotherapy for CML.

Lymphomas are a group of malignant diseases that can be derived from constituent cells of the lymphoid tissue, lymphocytes and histiocytes. Malignant lymphomas are divided into Hodgkin’s and non-Hodgkin’s lymphoma (NHL) and are more common in the head and neck region, but NHL can also be found in extranodal regions (25% of the cases), with or without lymph node involvement. NHL is more frequent than Hodgkin’s disease and corresponds to around 90% of head and neck malignancies (105,106). Of NHL subtypes, follicular and diffuse large B-cell lymphomas are the most frequently diagnosed, corresponding to about 20% and 30%, respectively. Other subtypes account for fewer than 10% of the cases (107).

Studies have shown that AURKA is overexpressed in several NHL subtypes when compared to normal tissues and B-cells, Chowdhury et al. evaluated the protein expression of AURKA and the potential of AURKA inhibitor, MK-8745, in NHL derived from Mantle cell, Burkitt’s, and natural killer-cell lymphomas. Most of the cell lines analyzed presented enhanced AURKA expression but the results showed that treatment with pan-AKI VE-465 and taxol, when used alone, had higher toxicity than the selective inhibitor. However, the antitumoral effect of AURKA inhibitor was correlated with the expression of AURKA activator TPX2 (70).

Moreover, the pan-AKI AT9283 exhibited highly cytotoxic effects in NHL cells and, when used in combination with docetaxel, enhanced apoptosis, as well as survival rates and reduced tumor volume in the mouse mantle cell lymphoma xenograft model (68). The synergistic effect of AURK inhibition was observed with other antitumoral agents that target the cell cycle and microtubular constituents. The disruption in replication machinery leads to selective cancer cell death, enhancing the antitumoral potential of AKI in oncohematological treatment (108,109).

Kong et al. evaluated the synergism between the inhibitors of AURKA, alisertib, and BCL2 apoptosis regulator (BCL2) ABT-199, in cell lines of the NHL subtype double-hit lymphoma. Interestingly, the BCL2 inhibitor showed better results in vitro than AURKA inhibitor but the ABT-199 plus alisertib treatment synergistically inhibited viability in double-hit lymphoma cells (62). This subtype of NHL is characterized by MYC proto-oncogene (MYC) and BCL2 translocation, MYC and AURKA are regulated by each other in a feedback pathway and exert a strong role in the cell cycle and replicative process (110). BCL2 regulates the apoptotic process and has a function in survival and chemoresistance in hematological malignancies (111,112). Thus, dual inhibitory therapy focusing on AURKA and BCL2 may be an interesting option for treatment in patients with double-hit lymphoma with these specific mutations.

Table II presents clinical trials from the past 10 years utilizing AKIs as a therapeutic option for patients afflicted by leukemia and other hematological disorders. The most common studied disorder was AML, appearing in seven out of 12 studies (113-117,122,124), while the most targeted kinase was AURKA, appearing in 10 of 12 studies (113-118,120-123). AURKC was not a main target in any of the selected studies.

In all clinical trials presented, the most prevalent therapeutic option was alisertib (113,114,118,120,122,123), a selective inhibitor of AURKA that is also under investigation for relevance in the treatment of non-hematological malignancies (125). While it was most effective when treating patients with AML, this finding may also be correlated to its use as a synergistic compound alongside conventional induction chemotherapy (113,114). A study cohort by Goldberg et al. demonstrated alisertib to be potentially clinically effective for patients with AML, while having no efficacy when treating those with myelodysplastic syndrome, hinting towards a better understanding of the drug mechanism of action and biological pathway (122).

While most investigated drugs were AURKA-selective inhibitors or pan-AKIs, AURKB was selectively targeted by the use of barasertib in two studies (119,124). Although its efficacy was demonstrated in the treatment of patients with diffuse large B-cell lymphoma, the low progression-free survival of 60 days attested to by Collins et al. discredits its use as a monotherapy for this disease (119). When treating patients with AML, Kantarjian et al. demonstrated barasertib to achieve better complete response rates as a monotherapy than low-dose cytosine arabinoside, an agent previously demonstrated to be clinically beneficial in clinical trials (124,126).

Other inhibitors that were reported in only one study include AMG 900, AT9283, ENMD-2076 and MK-0457. These drugs had only modest to no efficacy when utilized as single agents and in low, tolerable doses, although ENMD-2076 monotherapy still achieved a 25% overall response rate when treating patients with AML and CML, and should be taken into account when considering possible synergistic interactions (115-117,121).

Overall, the usage of AKIs did not cause unexpected or unmanageable adverse effects in patients and the main ones were related to blood or gastrointestinal disorders (115,117,119-121,123), which is in accordance with the expected effects of aurora inhibition in non-malignant rapidly dividing cells due to mitosis impairment (127,128).

Conclusion

The interest in understanding the involvement of aurora in biological pathways in non-malignant as well as in malignant cells has only grown over the years. While there is still much to be elucidated, it is clear that these kinases and their upstream and downstream regulators play a key role in mitosis and oncogenesis, motivating their investigation as potential targets for oncological treatments. Even though the clinical trials using AKIs as a monotherapy for hematological disorders have not shown great results, the manageable toxicity and potentially synergistic effects still render them a focus of interest for future investigations in combinatorial clinical trials.

Conflicts of Interest

The Authors declare no conflicts of interest regarding this study.

Authors’ Contributions

Machado CB, da Silva EL and Moreira-Nunes CA, performed the study design; Machado CB and Nogueira BMD, prepared the figures; Machado CB, da Silva EL, Nogueira BMD, da Silva JBS, Moraes-Filho MO, Moraes MEA, Montenegro RC and Moreira-Nunes CA wrote the article. All Authors read and approved the final article.

Acknowledgements

This study was supported by Brazilian funding agencies National Counsel of Technological and Scientific Development (CNPq; to ELS, RCM, MEAM, MOMF and CAMN).

References

1 Global Burden of Disease Cancer Collaboration Fitzmaurice C Abate D Abbasi N Abbastabar H Abd-Allah F Abdel-Rahman O Abdelalim A Abdoli A Abdollahpour I Abdulle ASM Abebe ND Abraha HN Abu-Raddad LJ Abualhasan A Adedeji IA Advani SM Afarideh M Afshari M Aghaali M Agius D Agrawal S Ahmadi A Ahmadian E Ahmadpour E Ahmed MB Akbari ME Akinyemiju T Al-Aly Z AlAbdulKader AM Alahdab F Alam T Alamene GM Alemnew BTT Alene KA Alinia C Alipour V Aljunid SM Bakeshei FA Almadi MAH Almasi-Hashiani A Alsharif U Alsowaidi S Alvis-Guzman N Amini E Amini S Amoako YA Anbari Z Anber NH Andrei CL Anjomshoa M Ansari F Ansariadi A Appiah SCY Arab-Zozani M Arabloo J Arefi Z Aremu O Areri HA Artaman A Asayesh H Asfaw ET Ashagre AF Assadi R Ataeinia B Atalay HT Ataro Z Atique S Ausloos M Avila-Burgos L Avokpaho EFGA Awasthi A Awoke N Ayala Quintanilla BP Ayanore MA Ayele HT Babaee E Bacha U Badawi A Bagherzadeh M Bagli E Balakrishnan S Balouchi A Bärnighausen TW Battista RJ Behzadifar M Behzadifar M Bekele BB Belay YB Belayneh YM Berfield KKS Berhane A Bernabe E Beuran M Bhakta N Bhattacharyya K Biadgo B Bijani A Bin Sayeed MS Birungi C Bisignano C Bitew H Bjørge T Bleyer A Bogale KA Bojia HA Borzì AM Bosetti C Bou-Orm IR Brenner H Brewer JD Briko AN Briko NI Bustamante-Teixeira MT Butt ZA Carreras G Carrero JJ Carvalho F Castro C Castro F Catalá-López F Cerin E Chaiah Y Chanie WF Chattu VK Chaturvedi P Chauhan NS Chehrazi M Chiang PP Chichiabellu TY Chido-Amajuoyi OG Chimed-Ochir O Choi JJ Christopher DJ Chu DT Constantin MM Costa VM Crocetti E Crowe CS Curado MP Dahlawi SMA Damiani G Darwish AH Daryani A das Neves J Demeke FM Demis AB Demissie BW Demoz GT Denova-Gutiérrez E Derakhshani A Deribe KS Desai R Desalegn BB Desta M Dey S Dharmaratne SD Dhimal M Diaz D Dinberu MTT Djalalinia S Doku DT Drake TM Dubey M Dubljanin E Duken EE Ebrahimi H Effiong A Eftekhari A El Sayed I Zaki MES El-Jaafary SI El-Khatib Z Elemineh DA Elkout H Ellenbogen RG Elsharkawy A Emamian MH Endalew DA Endries AY Eshrati B Fadhil I Fallah Omrani V Faramarzi M Farhangi MA Farioli A Farzadfar F Fentahun N Fernandes E Feyissa GT Filip I Fischer F Fisher JL Force LM Foroutan M Freitas M Fukumoto T Futran ND Gallus S Gankpe FG Gayesa RT Gebrehiwot TT Gebremeskel GG Gedefaw GA Gelaw BK Geta B Getachew S Gezae KE Ghafourifard M Ghajar A Ghashghaee A Gholamian A Gill PS Ginindza TTG Girmay A Gizaw M Gomez RS Gopalani SV Gorini G Goulart BNG Grada A Ribeiro Guerra M Guimaraes ALS Gupta PC Gupta R Hadkhale K Haj-Mirzaian A Haj-Mirzaian A Hamadeh RR Hamidi S Hanfore LK Haro JM Hasankhani M Hasanzadeh A Hassen HY Hay RJ Hay SI Henok A Henry NJ Herteliu C Hidru HD Hoang CL Hole MK Hoogar P Horita N Hosgood HD Hosseini M Hosseinzadeh M Hostiuc M Hostiuc S Househ M Hussen MM Ileanu B Ilic MD Innos K Irvani SSN Iseh KR Islam SMS Islami F Jafari Balalami N Jafarinia M Jahangiry L Jahani MA Jahanmehr N Jakovljevic M James SL Javanbakht M Jayaraman S Jee SH Jenabi E Jha RP Jonas JB Jonnagaddala J Joo T Jungari SB Jürisson M Kabir A Kamangar F Karch A Karimi N Karimian A Kasaeian A Kasahun GG Kassa B Kassa TD Kassaw MW Kaul A Keiyoro PN Kelbore AG Kerbo AA Khader YS Khalilarjmandi M Khan EA Khan G Khang YH Khatab K Khater A Khayamzadeh M Khazaee-Pool M Khazaei S Khoja AT Khosravi MH Khubchandani J Kianipour N Kim D Kim YJ Kisa A Kisa S Kissimova-Skarbek K Komaki H Koyanagi A Krohn KJ Bicer BK Kugbey N Kumar V Kuupiel D La Vecchia C Lad DP Lake EA Lakew AM Lal DK Lami FH Lan Q Lasrado S Lauriola P Lazarus JV Leigh J Leshargie CT Liao Y Limenih MA Listl S Lopez AD Lopukhov PD Lunevicius R Madadin M Magdeldin S El Razek HMA Majeed A Maleki A Malekzadeh R Manafi A Manafi N Manamo WA Mansourian M Mansournia MA Mantovani LG Maroufizadeh S Martini SMS Mashamba-Thompson TP Massenburg BB Maswabi MT Mathur MR McAlinden C McKee M Meheretu HAA Mehrotra R Mehta V Meier T Melaku YA Meles GG Meles HG Melese A Melku M Memiah PTN Mendoza W Menezes RG Merat S Meretoja TJ Mestrovic T Miazgowski B Miazgowski T Mihretie KMM Miller TR Mills EJ Mir SM Mirzaei H Mirzaei HR Mishra R Moazen B Mohammad DK Mohammad KA Mohammad Y Darwesh AM Mohammadbeigi A Mohammadi H Mohammadi M Mohammadian M Mohammadian-Hafshejani A Mohammadoo-Khorasani M Mohammadpourhodki R Mohammed AS Mohammed JA Mohammed S Mohebi F Mokdad AH Monasta L Moodley Y Moosazadeh M Moossavi M Moradi G Moradi-Joo M Moradi-Lakeh M Moradpour F Morawska L Morgado-da-Costa J Morisaki N Morrison SD Mosapour A Mousavi SM Muche AA Muhammed OSS Musa J Nabhan AF Naderi M Nagarajan AJ Nagel G Nahvijou A Naik G Najafi F Naldi L Nam HS Nasiri N Nazari J Negoi I Neupane S Newcomb PA Nggada HA Ngunjiri JW Nguyen CT Nikniaz L Ningrum DNA Nirayo YL Nixon MR Nnaji CA Nojomi M Nosratnejad S Shiadeh MN Obsa MS Ofori-Asenso R Ogbo FA Oh IH Olagunju AT Olagunju TO Oluwasanu MM Omonisi AE Onwujekwe OE Oommen AM Oren E Ortega-Altamirano DDV Ota E Otstavnov SS Owolabi MO P A M Padubidri JR Pakhale S Pakpour AH Pana A Park EK Parsian H Pashaei T Patel S Patil ST Pennini A Pereira DM Piccinelli C Pillay JD Pirestani M Pishgar F Postma MJ Pourjafar H Pourmalek F Pourshams A Prakash S Prasad N Qorbani M Rabiee M Rabiee N Radfar A Rafiei A Rahim F Rahimi M Rahman MA Rajati F Rana SM Raoofi S Rath GK Rawaf DL Rawaf S Reiner RC Renzaho AMN Rezaei N Rezapour A Ribeiro AI Ribeiro D Ronfani L Roro EM Roshandel G Rostami A Saad RS Sabbagh P Sabour S Saddik B Safiri S Sahebkar A Salahshoor MR Salehi F Salem H Salem MR Salimzadeh H Salomon JA Samy AM Sanabria J Santric Milicevic MM Sartorius B Sarveazad A Sathian B Satpathy M Savic M Sawhney M Sayyah M Schneider IJC Schöttker B Sekerija M Sepanlou SG Sepehrimanesh M Seyedmousavi S Shaahmadi F Shabaninejad H Shahbaz M Shaikh MA Shamshirian A Shamsizadeh M Sharafi H Sharafi Z Sharif M Sharifi A Sharifi H Sharma R Sheikh A Shirkoohi R Shukla SR Si S Siabani S Silva DAS Silveira DGA Singh A Singh JA Sisay S Sitas F Sobngwi E Soofi M Soriano JB Stathopoulou V Sufiyan MB Tabarés-Seisdedos R Tabuchi T Takahashi K Tamtaji OR Tarawneh MR Tassew SG Taymoori P Tehrani-Banihashemi A Temsah MH Temsah O Tesfay BE Tesfay FH Teshale MY Tessema GA Thapa S Tlaye KG Topor-Madry R Tovani-Palone MR Traini E Tran BX Tran KB Tsadik AG Ullah I Uthman OA Vacante M Vaezi M Varona Pérez P Veisani Y Vidale S Violante FS Vlassov V Vollset SE Vos T Vosoughi K Vu GT Vujcic IS Wabinga H Wachamo TM Wagnew FS Waheed Y Weldegebreal F Weldesamuel GT Wijeratne T Wondafrash DZ Wonde TE Wondmieneh AB Workie HM Yadav R Yadegar A Yadollahpour A Yaseri M Yazdi-Feyzabadi V Yeshaneh A Yimam MA Yimer EM Yisma E Yonemoto N Younis MZ Yousefi B Yousefifard M Yu C Zabeh E Zadnik V Moghadam TZ Zaidi Z Zamani M Zandian H Zangeneh A Zaki L Zendehdel K Zenebe ZM Zewale TA Ziapour A Zodpey S & Murray CJL Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2017: a systematic analysis for the global burden of disease study. JAMA Oncol. 5(12) 1749 - 1768 2019. PMID: 31560378. DOI: 10.1001/jamaoncol.2019.2996
2 Hassanpour S & Dehghani M Review of cancer from perspective of molecular. Journal of Cancer Research and Practice. 4(4) 127 - 129 2019. DOI: 10.1016/j.jcrpr.2017.07.001
3 Hanahan D & Weinberg RA The hallmarks of cancer. Cell. 100(1) 57 - 70 2000. PMID: 10647931. DOI: 10.1016/s0092-8674(00)81683-9
4 Foster I Cancer: A cell cycle defect. Radiography. 14(2) 144 - 149 2019. DOI: 10.1016/j.radi.2006.12.001
5 Otto T & Sicinski P Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer. 17(2) 93 - 115 2017. PMID: 28127048. DOI: 10.1038/nrc.2016.138
6 Ma HT & Poon RYC Aurora kinases and DNA damage response. Mutat Res. 821 111716 2020. PMID: 32738522. DOI: 10.1016/j.mrfmmm.2020.111716
7 Vader G & Lens SM The Aurora kinase family in cell division and cancer. Biochim Biophys Acta. 1786(1) 60 - 72 2008. PMID: 18662747. DOI: 10.1016/j.bbcan.2008.07.003
8 Chen HL Tang CJ Chen CY & Tang TK Overexpression of an Aurora-C kinase-deficient mutant disrupts the Aurora-B/INCENP complex and induces polyploidy. J Biomed Sci. 12(2) 297 - 310 2005. PMID: 15917996. DOI: 10.1007/s11373-005-0980-0
9 Carvajal RD Tse A & Schwartz GK Aurora kinases: new targets for cancer therapy. Clin Cancer Res. 12(23) 6869 - 6875 2006. PMID: 17145803. DOI: 10.1158/1078-0432.CCR-06-1405
10 Tang A Gao K Chu L Zhang R Yang J & Zheng J Aurora kinases: novel therapy targets in cancers. Oncotarget. 8(14) 23937 - 23954 2017. PMID: 28147341. DOI: 10.18632/oncotarget.14893
11 Choudary I Barr PM & Friedberg J Recent advances in the development of Aurora kinases inhibitors in hematological malignancies. Ther Adv Hematol. 6(6) 282 - 294 2015. PMID: 26622997. DOI: 10.1177/2040620715607415
12 Glover DM Leibowitz MH McLean DA & Parry H Mutations in aurora prevent centrosome separation leading to the formation of monopolar spindles. Cell. 81(1) 95 - 105 1995. PMID: 7720077. DOI: 10.1016/0092-8674(95)90374-7
13 Stenoien DL Sen S Mancini MA & Brinkley BR Dynamic association of a tumor amplified kinase, Aurora-A, with the centrosome and mitotic spindle. Cell Motil Cytoskeleton. 55(2) 134 - 146 2003. PMID: 12740874. DOI: 10.1002/cm.10120
14 Goldenson B & Crispino JD The aurora kinases in cell cycle and leukemia. Oncogene. 34(5) 537 - 545 2015. PMID: 24632603. DOI: 10.1038/onc.2014.14
15 Petretti C Savoian M Montembault E Glover DM Prigent C & Giet R The PITSLRE/CDK11p58 protein kinase promotes centrosome maturation and bipolar spindle formation. EMBO Rep. 7(4) 418 - 424 2006. PMID: 16462731. DOI: 10.1038/sj.embor.7400639
16 Barr AR & Gergely F Aurora-A: the maker and breaker of spindle poles. J Cell Sci. 120(Pt 17) 2987 - 2996 2007. PMID: 17715155. DOI: 10.1242/jcs.013136
17 Wang G Jiang Q & Zhang C The role of mitotic kinases in coupling the centrosome cycle with the assembly of the mitotic spindle. J Cell Sci. 127(Pt 19) 4111 - 4122 2014. PMID: 25128564. DOI: 10.1242/jcs.151753
18 Abe Y Ohsugi M Haraguchi K Fujimoto J & Yamamoto T LATS2-Ajuba complex regulates gamma-tubulin recruitment to centrosomes and spindle organization during mitosis. FEBS Lett. 580(3) 782 - 788 2006. PMID: 16413547. DOI: 10.1016/j.febslet.2005.12.096
19 Toji S Yabuta N Hosomi T Nishihara S Kobayashi T Suzuki S Tamai K & Nojima H The centrosomal protein Lats2 is a phosphorylation target of Aurora-A kinase. Genes Cells. 9(5) 383 - 397 2004. PMID: 15147269. DOI: 10.1111/j.1356-9597.2004.00732.x
20 Willems E Dedobbeleer M Digregorio M Lombard A Lumapat PN & Rogister B The functional diversity of Aurora kinases: a comprehensive review. Cell Div. 13 7 2018. PMID: 30250494. DOI: 10.1186/s13008-018-0040-6
21 Kimura M Kotani S Hattori T Sumi N Yoshioka T Todokoro K & Okano Y Cell cycle-dependent expression and spindle pole localization of a novel human protein kinase, Aik, related to Aurora of Drosophila and yeast Ipl1. J Biol Chem. 272(21) 13766 - 13771 1997. PMID: 9153231. DOI: 10.1074/jbc.272.21.13766
22 Magnaghi-Jaulin L Eot-Houllier G Gallaud E & Giet R Aurora A protein kinase: to the centrosome and beyond. Biomolecules. 9(1) 28 2019. PMID: 30650622. DOI: 10.3390/biom9010028
23 Kufer TA Silljé HH Körner R Gruss OJ Meraldi P & Nigg EA Human TPX2 is required for targeting Aurora-A kinase to the spindle. J Cell Biol. 158(4) 617 - 623 2002. PMID: 12177045. DOI: 10.1083/jcb.200204155
24 Eyers PA Erikson E Chen LG & Maller JL A novel mechanism for activation of the protein kinase Aurora A. Curr Biol. 13(8) 691 - 697 2003. PMID: 12699628. DOI: 10.1016/s0960-9822(03)00166-0
25 Giet R Uzbekov R Cubizolles F Le Guellec K & Prigent C The Xenopus laevis aurora-related protein kinase pEg2 associates with and phosphorylates the kinesin-related protein XlEg5. J Biol Chem. 274(21) 15005 - 15013 1999. PMID: 10329703. DOI: 10.1074/jbc.274.21.15005
26 Kapitein LC Peterman EJ Kwok BH Kim JH Kapoor TM & Schmidt CF The bipolar mitotic kinesin Eg5 moves on both microtubules that it crosslinks. Nature. 435(7038) 114 - 118 2005. PMID: 15875026. DOI: 10.1038/nature03503
27 Tanenbaum ME Macůrek L Janssen A Geers EF Alvarez-Fernández M & Medema RH Kif15 cooperates with eg5 to promote bipolar spindle assembly. Curr Biol. 19(20) 1703 - 1711 2009. PMID: 19818618. DOI: 10.1016/j.cub.2009.08.027
28 van Heesbeen RGHP Raaijmakers JA Tanenbaum ME Halim VA Lelieveld D Lieftink C Heck AJR Egan DA & Medema RH Aurora A, MCAK, and Kif18b promote Eg5-independent spindle formation. Chromosoma. 126(4) 473 - 486 2017. PMID: 27354041. DOI: 10.1007/s00412-016-0607-4
29 Lens SM Voest EE & Medema RH Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat Rev Cancer. 10(12) 825 - 841 2010. PMID: 21102634. DOI: 10.1038/nrc2964
30 Carmena M Wheelock M Funabiki H & Earnshaw WC The chromosomal passenger complex (CPC): from easy rider to the godfather of mitosis. Nat Rev Mol Cell Biol. 13(12) 789 - 803 2012. PMID: 23175282. DOI: 10.1038/nrm3474
31 Hendzel MJ Wei Y Mancini MA Van Hooser A Ranalli T Brinkley BR Bazett-Jones DP & Allis CD Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma. 106(6) 348 - 360 1997. PMID: 9362543. DOI: 10.1007/s004120050256
32 Hans F & Dimitrov S Histone H3 phosphorylation and cell division. Oncogene. 20(24) 3021 - 3027 2001. PMID: 11420717. DOI: 10.1038/sj.onc.1204326
33 Crosio C Fimia GM Loury R Kimura M Okano Y Zhou H Sen S Allis CD & Sassone-Corsi P Mitotic phosphorylation of histone H3: spatio-temporal regulation by mammalian Aurora kinases. Mol Cell Biol. 22(3) 874 - 885 2002. PMID: 11784863. DOI: 10.1128/mcb.22.3.874-885.2002
34 Giet R & Glover DM Drosophila aurora B kinase is required for histone H3 phosphorylation and condensin recruitment during chromosome condensation and to organize the central spindle during cytokinesis. J Cell Biol. 152(4) 669 - 682 2001. PMID: 11266459. DOI: 10.1083/jcb.152.4.669
35 Wang F Dai J Daum JR Niedzialkowska E Banerjee B Stukenberg PT Gorbsky GJ & Higgins JM Histone H3 Thr-3 phosphorylation by Haspin positions Aurora B at centromeres in mitosis. Science. 330(6001) 231 - 235 2010. PMID: 20705812. DOI: 10.1126/science.1189435
36 Kawashima SA Yamagishi Y Honda T Ishiguro K & Watanabe Y Phosphorylation of H2A by Bub1 prevents chromosomal instability through localizing shugoshin. Science. 327(5962) 172 - 177 2010. PMID: 19965387. DOI: 10.1126/science.1180189
37 Yamagishi Y Honda T Tanno Y & Watanabe Y Two histone marks establish the inner centromere and chromosome bi-orientation. Science. 330(6001) 239 - 243 2010. PMID: 20929775. DOI: 10.1126/science.1194498
38 Broad AJ DeLuca KF & DeLuca JG Aurora B kinase is recruited to multiple discrete kinetochore and centromere regions in human cells. J Cell Biol. 219(3) e201905144 2020. PMID: 32028528. DOI: 10.1083/jcb.201905144
39 Godek KM Kabeche L & Compton DA Regulation of kinetochore-microtubule attachments through homeostatic control during mitosis. Nat Rev Mol Cell Biol. 16(1) 57 - 64 2015. PMID: 25466864. DOI: 10.1038/nrm3916
40 Welburn JP Vleugel M Liu D Yates JR 3rd Lampson MA Fukagawa T & Cheeseman IM Aurora B phosphorylates spatially distinct targets to differentially regulate the kinetochore-microtubule interface. Mol Cell. 38(3) 383 - 392 2010. PMID: 20471944. DOI: 10.1016/j.molcel.2010.02.034
41 London N & Biggins S Signalling dynamics in the spindle checkpoint response. Nat Rev Mol Cell Biol. 15(11) 736 - 747 2014. PMID: 25303117. DOI: 10.1038/nrm3888
42 Trivedi P & Stukenberg PT A centromere-signaling network underlies the coordination among mitotic events. Trends Biochem Sci. 41(2) 160 - 174 2016. PMID: 26705896. DOI: 10.1016/j.tibs.2015.11.002
43 Kitagawa M Fung SY Hameed UF Goto H Inagaki M & Lee SH Cdk1 coordinates timely activation of MKlp2 kinesin with relocation of the chromosome passenger complex for cytokinesis. Cell Rep. 7(1) 166 - 179 2014. PMID: 24656812. DOI: 10.1016/j.celrep.2014.02.034
44 Mierzwa B & Gerlich DW Cytokinetic abscission: molecular mechanisms and temporal control. Dev Cell. 31(5) 525 - 538 2014. PMID: 25490264. DOI: 10.1016/j.devcel.2014.11.006
45 Kitagawa M & Lee SH The chromosomal passenger complex (CPC) as a key orchestrator of orderly mitotic exit and cytokinesis. Front Cell Dev Biol. 3 14 2015. PMID: 25798441. DOI: 10.3389/fcell.2015.00014
46 Steigemann P Wurzenberger C Schmitz MH Held M Guizetti J Maar S & Gerlich DW Aurora B-mediated abscission checkpoint protects against tetraploidization. Cell. 136(3) 473 - 484 2009. PMID: 19203582. DOI: 10.1016/j.cell.2008.12.020
47 Quartuccio SM & Schindler K Functions of Aurora kinase C in meiosis and cancer. Front Cell Dev Biol. 3 50 2015. PMID: 26347867. DOI: 10.3389/fcell.2015.00050
48 Yang KT Li SK Chang CC Tang CJ Lin YN Lee SC & Tang TK Aurora-C kinase deficiency causes cytokinesis failure in meiosis I and production of large polyploid oocytes in mice. Mol Biol Cell. 21(14) 2371 - 2383 2010. PMID: 20484572. DOI: 10.1091/mbc.e10-02-0170
49 Assou S Anahory T Pantesco V Le Carrour T Pellestor F Klein B Reyftmann L Dechaud H De Vos J & Hamamah S The human cumulus – oocyte complex gene-expression profile. Hum Reprod. 21(7) 1705 - 1719 2006. PMID: 16571642. DOI: 10.1093/humrep/del065
50 Sasai K Katayama H Hawke DH & Sen S Aurora-C interactions with survivin and INCENP reveal shared and distinct features compared with Aurora-B chromosome passenger protein complex. PLoS One. 11(6) e0157305 2016. PMID: 27332895. DOI: 10.1371/journal.pone.0157305
51 Fernández-Miranda G Trakala M Martín J Escobar B González A Ghyselinck NB Ortega S Cañamero M Pérez de Castro I & Malumbres M Genetic disruption of aurora B uncovers an essential role for aurora C during early mammalian development. Development. 138(13) 2661 - 2672 2011. PMID: 21613325. DOI: 10.1242/dev.066381
52 Balboula AZ & Schindler K Selective disruption of aurora C kinase reveals distinct functions from aurora B kinase during meiosis in mouse oocytes. PLoS Genet. 10(2) e1004194 2014. PMID: 24586209. DOI: 10.1371/journal.pgen.1004194
53 Cicenas J The Aurora kinase inhibitors in cancer research and therapy. J Cancer Res Clin Oncol. 142(9) 1995 - 2012 2016. PMID: 26932147. DOI: 10.1007/s00432-016-2136-1
54 Yan M Wang C He B Yang M Tong M Long Z Liu B Peng F Xu L Zhang Y Liang D Lei H Subrata S Kelley KW Lam EW Jin B & Liu Q Aurora-A kinase: a potent oncogene and target for cancer therapy. Med Res Rev. 36(6) 1036 - 1079 2016. PMID: 27406026. DOI: 10.1002/med.21399
55 Moreira-Nunes CA Mesquita FP Portilho AJS Mello Júnior FAR Maués JHDS Pantoja LDC Wanderley AV Khayat AS Zuercher WJ Montenegro RC de Moraes-Filho MO & de Moraes MEA Targeting aurora kinases as a potential prognostic and therapeutical biomarkers in pediatric acute lymphoblastic leukaemia. Sci Rep. 10(1) 21272 2020. PMID: 33277547. DOI: 10.1038/s41598-020-78024-8
56 Moore AS Faisal A Mak GWY Miraki-Moud F Bavetsias V Valenti M Box G Hallsworth A de Haven Brandon A Xavier CPR Stronge R Pearson ADJ Blagg J Raynaud FI Chopra R Eccles SA Taussig DC & Linardopoulos S Quizartinib-resistant FLT3-ITD acute myeloid leukemia cells are sensitive to the FLT3-Aurora kinase inhibitor CCT241736. Blood Adv. 4(7) 1478 - 1491 2020. PMID: 32282883. DOI: 10.1182/bloodadvances.2019000986
57 Wang JX Zhang L Huang ZW Zhang XN Jiang YY Liu FJ Long L Xue MJ Lu G Liu Q & Long ZJ Aurora kinase inhibitor restrains STAT5-activated leukemic cell proliferation by inducing mitochondrial impairment. J Cell Physiol. 235(11) 8358 - 8370 2020. PMID: 32239704. DOI: 10.1002/jcp.29680
58 Qi J Gao X Zhong X Zhang N Wang R Zhang H Pan T Liu X Yao Y Wu Q Niu M & Xu K Selective inhibition of Aurora A and B kinases effectively induces cell cycle arrest in t(8;21) acute myeloid leukemia. Biomed Pharmacother. 117 109113 2019. PMID: 31207577. DOI: 10.1016/j.biopha.2019.109113
59 Heo SK Noh EK Jeong YK Ju LJ Sung JY Yu HM Cheon J Koh S Min YJ Choi Y & Jo JC Radotinib inhibits mitosis entry in acute myeloid leukemia cells via suppression of Aurora kinase A expression. Tumour Biol. 41(5) 1010428319848612 2019. PMID: 31074363. DOI: 10.1177/1010428319848612
60 Payton M Cheung HK Ninniri MSS Marinaccio C Wayne WC Hanestad K Crispino JD Juan G & Coxon A Dual Targeting of Aurora Kinases with AMG 900 Exhibits Potent Preclinical Activity Against Acute Myeloid Leukemia with Distinct Post-Mitotic Outcomes. Mol Cancer Ther. 17(12) 2575 - 2585 2018. PMID: 30266802. DOI: 10.1158/1535-7163.MCT-18-0186
61 Okabe S Tauchi T Tanaka Y & Ohyashiki K Therapeutic targeting of Aurora A kinase in Philadelphia chromosome-positive ABL tyrosine kinase inhibitor-resistant cells. Oncotarget. 9(65) 32496 - 32506 2018. PMID: 30197758. DOI: 10.18632/oncotarget.25985
62 Kong L Jia X Song Z Qiu L Li L Qian Z Zhou S Liu X Ren X Meng B Fu K Wang P Xian-huo wang X & Zhang H Co-targeting Aurora kinase A and Bcl-2 synergistically inhibits the viability in double-hit lymphoma cells. Translational Cancer Research. 6(4) 746 - 754 2018. DOI: 10.21037/tcr.2017.06.41
63 Floc’h N Ashton S Taylor P Trueman D Harris E Odedra R Maratea K Derbyshire N Caddy J Jacobs VN Hattersley M Wen S Curtis NJ Pilling JE Pease EJ & Barry ST Optimizing therapeutic effect of aurora B inhibition in acute myeloid leukemia with AZD2811 nanoparticles. Mol Cancer Ther. 16(6) 1031 - 1040 2017. PMID: 28292940. DOI: 10.1158/1535-7163.MCT-16-0580
64 Long ZJ Wang LX Zheng FM Chen JJ Luo Y Tu XX Lin DJ Lu G & Liu Q A novel compound against oncogenic Aurora kinase A overcomes imatinib resistance in chronic myeloid leukemia cells. Int J Oncol. 46(6) 2488 - 2496 2015. PMID: 25872528. DOI: 10.3892/ijo.2015.2960
65 Chen YP Lin HJ Chen JS Tsai MY Hsieh HP Chang JY Chen NF Chang KC Huang WT Su WC Yang ST Chang WC Hung LY & Chen TY CDKN1A-mediated responsiveness of MLL-AF4-positive acute lymphoblastic leukemia to Aurora kinase-A inhibitors. Int J Cancer. 135(3) 751 - 762 2014. PMID: 24382688. DOI: 10.1002/ijc.28708
66 Yamauchi T Uzui K Shigemi H Negoro E Yoshida A & Ueda T Aurora B inhibitor barasertib and cytarabine exert a greater-than-additive cytotoxicity in acute myeloid leukemia cells. Cancer Sci. 104(7) 926 - 933 2013. PMID: 23557198. DOI: 10.1111/cas.12164
67 Fei F Lim M Schmidhuber S Moll J Groffen J & Heisterkamp N Treatment of human pre-B acute lymphoblastic leukemia with the Aurora kinase inhibitor PHA-739358 (Danusertib). Mol Cancer. 11 42 2012. PMID: 22721004. DOI: 10.1186/1476-4598-11-42
68 Qi W Liu X Cooke LS Persky DO Miller TP Squires M & Mahadevan D AT9283, a novel aurora kinase inhibitor, suppresses tumor growth in aggressive B-cell lymphomas. Int J Cancer. 130(12) 2997 - 3005 2012. PMID: 21796626. DOI: 10.1002/ijc.26324
69 Kim SJ Jang JE Cheong JW Eom JI Jeung HK Kim Y Hwang DY & Min YH Aurora A kinase expression is increased in leukemia stem cells, and a selective Aurora A kinase inhibitor enhances Ara-C-induced apoptosis in acute myeloid leukemia stem cells. Korean J Hematol. 47(3) 178 - 185 2012. PMID: 23071472. DOI: 10.5045/kjh.2012.47.3.178
70 Chowdhury A Chowdhury S & Tsai MY A novel Aurora kinase A inhibitor MK-8745 predicts TPX2 as a therapeutic biomarker in non-Hodgkin lymphoma cell lines. Leuk Lymphoma. 53(3) 462 - 471 2012. PMID: 21879811. DOI: 10.3109/10428194.2011.619018
71 Yoon Y Cowley DO Gallant J Jones SN Van Dyke T & Rivera-Pérez JA Conditional Aurora A deficiency differentially affects early mouse embryo patterning. Dev Biol. 371(1) 77 - 85 2012. PMID: 22939930. DOI: 10.1016/j.ydbio.2012.08.010
72 Hégarat N Smith E Nayak G Takeda S Eyers PA & Hochegger H Aurora A and Aurora B jointly coordinate chromosome segregation and anaphase microtubule dynamics. J Cell Biol. 195(7) 1103 - 1113 2011. PMID: 22184196. DOI: 10.1083/jcb.201105058
73 Papaemmanuil E Gerstung M Bullinger L Gaidzik VI Paschka P Roberts ND Potter NE Heuser M Thol F Bolli N Gundem G Van Loo P Martincorena I Ganly P Mudie L McLaren S O’Meara S Raine K Jones DR Teague JW Butler AP Greaves MF Ganser A Döhner K Schlenk RF Döhner H & Campbell PJ Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 374(23) 2209 - 2221 2016. PMID: 27276561. DOI: 10.1056/NEJMoa1516192
74 Sampath D Rao VA & Plunkett W Mechanisms of apoptosis induction by nucleoside analogs. Oncogene. 22(56) 9063 - 9074 2003. PMID: 14663485. DOI: 10.1038/sj.onc.1207229
75 Bavetsias V Crumpler S Sun C Avery S Atrash B Faisal A Moore AS Kosmopoulou M Brown N Sheldrake PW Bush K Henley A Box G Valenti M de Haven Brandon A Raynaud FI Workman P Eccles SA Bayliss R Linardopoulos S & Blagg J Optimization of imidazo[4,5-b]pyridine-based kinase inhibitors: identification of a dual FLT3/Aurora kinase inhibitor as an orally bioavailable preclinical development candidate for the treatment of acute myeloid leukemia. J Med Chem. 55(20) 8721 - 8734 2012. PMID: 23043539. DOI: 10.1021/jm300952s
76 Lin S Ptasinska A Chen X Shrestha M Assi SA Chin PS Imperato MR Aronow BJ Zhang J Weirauch MT Bonifer C & Mulloy JC A FOXO1-induced oncogenic network defines the AML1-ETO preleukemic program. Blood. 130(10) 1213 - 1222 2017. PMID: 28710059. DOI: 10.1182/blood-2016-11-750976
77 Hospital MA Prebet T Bertoli S Thomas X Tavernier E Braun T Pautas C Perrot A Lioure B Rousselot P Tamburini J Cluzeau T Konopacki J Randriamalala E Berthon C Gourin MP Recher C Cahn JY Ifrah N Dombret H & Boissel N Core-binding factor acute myeloid leukemia in first relapse: a retrospective study from the French AML Intergroup. Blood. 124(8) 1312 - 1319 2014. PMID: 25006122. DOI: 10.1182/blood-2014-01-549212
78 Al-Harbi S Aljurf M Mohty M Almohareb F & Ahmed SOA An update on the molecular pathogenesis and potential therapeutic targeting of AML with t(8;21)(q22;q22.1);RUNX1-RUNX1T1. Blood Adv. 4(1) 229 - 238 2020. PMID: 31935293. DOI: 10.1182/bloodadvances.2019000168
79 Terwilliger T & Abdul-Hay M Acute lymphoblastic leukemia: a comprehensive review and 2017 update. Blood Cancer J. 7(6) e577 2017. PMID: 28665419. DOI: 10.1038/bcj.2017.53
80 Scheurer ME Lupo JL & Bondy ML Epidemiology of childhood cancer. In: Principles and Practice of Pediatric Oncology, Seventh Edition. Pizzo PA and Poplack DG. New York, Lippincott Williams & Wilkins. pp. 50 2016.
81 Hartsink-Segers SA Zwaan CM Exalto C Luijendijk MW Calvert VS Petricoin EF Evans WE Reinhardt D de Haas V Hedtjärn M Hansen BR Koch T Caron HN Pieters R & Den Boer ML Aurora kinases in childhood acute leukemia: the promise of aurora B as therapeutic target. Leukemia. 27(3) 560 - 568 2013. PMID: 22940834. DOI: 10.1038/leu.2012.256
82 Hongo T Okada S Inoue N Yamada S Yajima S Watanabe C Fujii Y & Horikoshi Y Two groups of Philadelphia chromosome-positive childhood acute lymphoblastic leukemia classified by pretreatment multidrug sensitivity or resistance in in vitro testing. Int J Hematol. 76(3) 251 - 259 2002. PMID: 12416736. DOI: 10.1007/BF02982795
83 Pui CH Frankel LS Carroll AJ Raimondi SC Shuster JJ Head DR Crist WM Land VJ Pullen DJ & Steuber CP Clinical characteristics and treatment outcome of childhood acute lymphoblastic leukemia with the t(4;11)(q21;q23): a collaborative study of 40 cases. Blood. 77(3) 440 - 447 1991. PMID: 1991161.
Pubmed |
84 Britten O Ragusa D Tosi S & Kamel YM MLL-rearranged acute leukemia with t(4;11)(q21;q23)-current treatment options. Is there a role for CAR-T cell therapy. Cells. 8(11) 1341 2019. PMID: 31671855. DOI: 10.3390/cells8111341
85 Machado C Nogueira B Portilho A Filho M Moraes M & Moreira-nunes C Caracterização do cromossomo Philadephia em tumores não-sólidos: uma abordagem citogenética ao câncer. A Genética e a Construção de Novos Paradigmas nas Ciências da Vida. 14 - 21 2021. DOI: 10.22533/at.ed.1652119032
86 Nowell PC & Hungerford DA Chromosome studies on normal and leukemic human leukocytes. J Natl Cancer Inst. 25 85 - 109 1960. PMID: 14427847.
Pubmed |
87 Aricò M Valsecchi MG Camitta B Schrappe M Chessells J Baruchel A Gaynon P Silverman L Janka-Schaub G Kamps W Pui CH & Masera G Outcome of treatment in children with Philadelphia chromosome-positive acute lymphoblastic leukemia. N Engl J Med. 342(14) 998 - 1006 2000. PMID: 10749961. DOI: 10.1056/NEJM200004063421402
88 Nishiwaki S Kim JH Ito M Maeda M Okuno Y Koyama D Ozawa Y Gunji M Osaki M Kitamura K Ushijima Y Ishikawa Y Miyamura K Sugiura I & Kiyoi H Multi-lineage BCR-ABL expression in Philadelphia chromosome-positive acute lymphoblastic leukemia is associated with improved prognosis but no specific molecular features. Front Oncol. 10 586567 2020. PMID: 33194739. DOI: 10.3389/fonc.2020.586567
89 Schlieben S Borkhardt A Reinisch I Ritterbach J Janssen JW Ratei R Schrappe M Repp R Zimmermann M Kabisch H Janka-Schaub G Bartram CR Ludwig WD Riehm H Lampert F & Harbott J Incidence and clinical outcome of children with BCR/ABL-positive acute lymphoblastic leukemia (ALL). A prospective RT-PCR study based on 673 patients enrolled in the German pediatric multicenter therapy trials ALL-BFM-90 and CoALL-05-92. Leukemia. 10(6) 957 - 963 1996. PMID: 8667652.
Pubmed |
90 Kang ZJ Liu YF Xu LZ Long ZJ Huang D Yang Y Liu B Feng JX Pan YJ Yan JS & Liu Q The Philadelphia chromosome in leukemogenesis. Chin J Cancer. 35 48 2016. PMID: 27233483. DOI: 10.1186/s40880-016-0108-0
91 Gorre ME Mohammed M Ellwood K Hsu N Paquette R Rao PN & Sawyers CL Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 293(5531) 876 - 880 2001. PMID: 11423618. DOI: 10.1126/science.1062538
92 Azam M Latek RR & Daley GQ Mechanisms of autoinhibition and STI-571/imatinib resistance revealed by mutagenesis of BCR-ABL. Cell. 112(6) 831 - 843 2003. PMID: 12654249. DOI: 10.1016/s0092-8674(03)00190-9
93 O’Hare T Corbin AS & Druker BJ Targeted CML therapy: controlling drug resistance, seeking cure. Curr Opin Genet Dev. 16(1) 92 - 99 2006. PMID: 16343892. DOI: 10.1016/j.gde.2005.11.002
94 Haidary AM Ahmed ZA Abdul-Ghafar J Rahmani S Noor S Erfani F Ahmad M Lakanwall N Malakzai HA Ibrahimkhil AS Esmat E Haidari M Yousufzai N Sharif S & Saqib AH Philadelphia chromosome positive chronic myeloid leukemia with 5q deletion at diagnosis. Mol Cytogenet. 14(1) 16 2021. PMID: 33685471. DOI: 10.1186/s13039-021-00539-0
95 Harrison C Philadelphia Chromosome. Encyclopedia of Genetics. 1449 - 1450 2019. DOI: 10.1006/rwgn.2001.0991
96 Van Etten RA Mechanisms of transformation by the BCR-ABL oncogene: new perspectives in the post-imatinib era. Leuk Res. 28(Suppl 1) S21 - S28 2004. PMID: 15036938. DOI: 10.1016/j.leukres.2003.10.005
97 Patel AB O’Hare T & Deininger MW Mechanisms of resistance to ABL kinase inhibition in chronic myeloid leukemia and thedevelopment of next generation ABL kinase inhibitors. Hematol Oncol Clin North Am. 31(4) 589 - 612 2017. PMID: 28673390. DOI: 10.1016/j.hoc.2017.04.007
98 Cammareri P Scopelliti A Todaro M Eterno V Francescangeli F Moyer MP Agrusa A Dieli F Zeuner A & Stassi G Aurora-a is essential for the tumorigenic capacity and chemoresistance of colorectal cancer stem cells. Cancer Res. 70(11) 4655 - 4665 2010. PMID: 20460511. DOI: 10.1158/0008-5472.CAN-09-3953
99 Anand S Penrhyn-Lowe S & Venkitaraman AR AURORA-A amplification overrides the mitotic spindle assembly checkpoint, inducing resistance to Taxol. Cancer Cell. 3(1) 51 - 62 2003. PMID: 12559175. DOI: 10.1016/s1535-6108(02)00235-0
100 He S Feng M Liu M Yang S Yan S Zhang W Wang Z Hu C Xu Q Chen L Zhu H & Xu N P21-activated kinase 7 mediates cisplatin-resistance of esophageal squamous carcinoma cells with Aurora-A overexpression. PLoS One. 9(12) e113989 2014. PMID: 25436453. DOI: 10.1371/journal.pone.0113989
101 Kuang P Chen Z Wang J Liu Z Wang J Gao J & Shen L Characterization of Aurora A and its impact on the effect of cisplatin-based chemotherapy in patients with non-small cell lung cancer. Transl Oncol. 10(3) 367 - 377 2017. PMID: 28431392. DOI: 10.1016/j.tranon.2017.02.010
102 Li Y Tang K Zhang H Zhang Y Zhou W & Chen X Function of Aurora kinase A in Taxol-resistant breast cancer and its correlation with P-gp. Mol Med Rep. 4(4) 739 - 746 2011. PMID: 21584498. DOI: 10.3892/mmr.2011.494
103 Thollet A Vendrell JA Payen L Ghayad SE Ben Larbi S Grisard E Collins C Villedieu M & Cohen PA ZNF217 confers resistance to the pro-apoptotic signals of paclitaxel and aberrant expression of Aurora-A in breast cancer cells. Mol Cancer. 9 291 2010. PMID: 21059223. DOI: 10.1186/1476-4598-9-291
104 Wang L Arras J Katsha A Hamdan S Belkhiri A Ecsedy J & El-Rifai W Cisplatin-resistant cancer cells are sensitive to Aurora kinase A inhibition by alisertib. Mol Oncol. 11(8) 981 - 995 2017. PMID: 28417568. DOI: 10.1002/1878-0261.12066
105 Bowzyk Al-Naeeb A Ajithkumar T Behan S & Hodson DJ Non-Hodgkin lymphoma. BMJ. 362 k3204 2018. PMID: 30135071. DOI: 10.1136/bmj.k3204
106 Singh R Shaik S Negi BS Rajguru JP Patil PB Parihar AS & Sharma U Non-Hodgkin’s lymphoma: A review. J Family Med Prim Care. 9(4) 1834 - 1840 2020. PMID: 32670927. DOI: 10.4103/jfmpc.jfmpc_1037_19
107 Ekström-Smedby K Epidemiology and etiology of non-Hodgkin lymphoma—a review. Acta Oncol. 45(3) 258 - 271 2006. PMID: 16644568. DOI: 10.1080/02841860500531682
108 Ikezoe T Takeuchi T Yang J Adachi Y Nishioka C Furihata M Koeffler HP & Yokoyama A Analysis of Aurora B kinase in non-Hodgkin lymphoma. Lab Invest. 89(12) 1364 - 1373 2009. PMID: 19823168. DOI: 10.1038/labinvest.2009.106
109 Wysong DR Chakravarty A Hoar K & Ecsedy JA The inhibition of Aurora A abrogates the mitotic delay induced by microtubule perturbing agents. Cell Cycle. 8(6) 876 - 888 2009. PMID: 19221504. DOI: 10.4161/cc.8.6.7897
110 Lu L Han H Tian Y Li W Zhang J Feng M & Li Y Aurora kinase A mediates c-Myc’s oncogenic effects in hepatocellular carcinoma. Mol Carcinog. 54(11) 1467 - 1479 2015. PMID: 25284017. DOI: 10.1002/mc.22223
111 Chanan-Khan A Bcl-2 antisense therapy in B-cell malignancies. Blood Rev. 19(4) 213 - 221 2005. PMID: 15784299. DOI: 10.1016/j.blre.2004.11.002
112 Perini GF Ribeiro GN Pinto Neto JV Campos LT & Hamerschlak N BCL-2 as therapeutic target for hematological malignancies. J Hematol Oncol. 11(1) 65 2018. PMID: 29747654. DOI: 10.1186/s13045-018-0608-2
113 Brunner A Blonquist T Deangelo D Mcmasters M Winer E Hobbs G Amrein P Hock H Steensma D Garcia J Luskin M Stone R Ballen K Rosenblatt J Avigan D Mcafee S Moran J Bergeron M Foster J Bertoli C Mcgregor K Fishman K Macrae M Burke M Behnan T Som T Ramos A Vartanian M Nelson N Logan E Lombardi story J Connolly C Neuberg D Chen Y Graubert T & Fathi A Phase II clinical trial of alisertib, an aurora A kinase inhibitor, in combination with induction chemotherapy in high-risk, untreated patients with acute myeloid leukemia. Blood. 132(Suppl 1) 766 - 766 2019. DOI: 10.1182/blood-2018-99-115145
114 Fathi AT Wander SA Blonquist TM Brunner AM Amrein PC Supko J Hermance NM Manning AL Sadrzadeh H Ballen KK Attar EC Graubert TA Hobbs G Joseph C Perry AM Burke M Silver R Foster J Bergeron M Ramos AY Som TT Fishman KM McGregor KL Connolly C Neuberg DS & Chen YB Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia. Haematologica. 102(4) 719 - 727 2017. PMID: 28034990. DOI: 10.3324/haematol.2016.158394
115 Kantarjian HM Schuster MW Jain N Advani A Jabbour E Gamelin E Rasmussen E Juan G Anderson A Chow VF Friberg G Vogl FD & Sekeres MA A phase 1 study of AMG 900, an orally administered pan-aurora kinase inhibitor, in adult patients with acute myeloid leukemia. Am J Hematol. 92(7) 660 - 667 2017. PMID: 28370201. DOI: 10.1002/ajh.24736
116 Vormoor B Veal GJ Griffin MJ Boddy AV Irving J Minto L Case M Banerji U Swales KE Tall JR Moore AS Toguchi M Acton G Dyer K Schwab C Harrison CJ Grainger JD Lancaster D Kearns P Hargrave D & Vormoor J A phase I/II trial of AT9283, a selective inhibitor of aurora kinase in children with relapsed or refractory acute leukemia: challenges to run early phase clinical trials for children with leukemia.. Pediatr Blood Cancer. 64(6) PMID: 27905678. DOI: 10.1002/pbc.26351
117 Yee KW Chen HW Hedley DW Chow S Brandwein J Schuh AC Schimmer AD Gupta V Sanfelice D Johnson T Le LW Arnott J Bray MR Sidor C & Minden MD A phase I trial of the aurora kinase inhibitor, ENMD-2076, in patients with relapsed or refractory acute myeloid leukemia or chronic myelomonocytic leukemia. Invest New Drugs. 34(5) 614 - 624 2016. PMID: 27406088. DOI: 10.1007/s10637-016-0375-2
118 Rosenthal A Kumar S Hofmeister C Laubach J Vij R Dueck A Gano K & Stewart AK A phase Ib study of the combination of the Aurora kinase inhibitor alisertib (MLN8237) and bortezomib in relapsed multiple myeloma. Br J Haematol. 174(2) 323 - 325 2016. PMID: 26403323. DOI: 10.1111/bjh.13765
119 Collins GP Eyre TA Linton KM Radford J Vallance GD Soilleux E & Hatton C A phase II trial of AZD1152 in relapsed/refractory diffuse large B-cell lymphoma. Br J Haematol. 170(6) 886 - 890 2015. PMID: 25721307. DOI: 10.1111/bjh.13333
120 Friedberg JW Mahadevan D Cebula E Persky D Lossos I Agarwal AB Jung J Burack R Zhou X Leonard EJ Fingert H Danaee H & Bernstein SH 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. 32(1) 44 - 50 2014. PMID: 24043741. DOI: 10.1200/JCO.2012.46.8793
121 Seymour JF Kim DW Rubin E Haregewoin A Clark J Watson P Hughes T Dufva I Jimenez JL Mahon FX Rousselot P Cortes J Martinelli G Papayannidis C Nagler A & Giles FJ A phase 2 study of MK-0457 in patients with BCR-ABL T315I mutant chronic myelogenous leukemia and philadelphia chromosome-positive acute lymphoblastic leukemia. Blood Cancer J. 4 e238 2014. PMID: 25127392. DOI: 10.1038/bcj.2014.60
122 Goldberg SL Fenaux P Craig MD Gyan E Lister J Kassis J Pigneux A Schiller GJ Jung J Jane Leonard E Fingert H & Westervelt P An exploratory phase 2 study of investigational Aurora A kinase inhibitor alisertib (MLN8237) in acute myelogenous leukemia and myelodysplastic syndromes. Leuk Res Rep. 3(2) 58 - 61 2014. PMID: 25068104. DOI: 10.1016/j.lrr.2014.06.003
123 Kelly KR Shea TC Goy A Berdeja JG Reeder CB McDonagh KT Zhou X Danaee H Liu H Ecsedy JA Niu H Benaim E & Iyer SP Phase I study of MLN8237—investigational Aurora A kinase inhibitor—in relapsed/refractory multiple myeloma, non-Hodgkin lymphoma and chronic lymphocytic leukemia. Invest New Drugs. 32(3) 489 - 499 2014. PMID: 24352795. DOI: 10.1007/s10637-013-0050-9
124 Kantarjian HM Martinelli G Jabbour EJ Quintás-Cardama A Ando K Bay JO Wei A Gröpper S Papayannidis C Owen K Pike L Schmitt N Stockman PK Giagounidis A & SPARK-AML1 Investigators Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia. Cancer. 119(14) 2611 - 2619 2013. PMID: 23605952. DOI: 10.1002/cncr.28113
125 Liewer S & Huddleston A Alisertib: a review of pharmacokinetics, efficacy and toxicity in patients with hematologic malignancies and solid tumors. Expert Opin Investig Drugs. 27(1) 105 - 112 2018. PMID: 29260599. DOI: 10.1080/13543784.2018.1417382
126 Burnett AK Milligan D Prentice AG Goldstone AH McMullin MF Hills RK & Wheatley K A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 109(6) 1114 - 1124 2007. PMID: 17315155. DOI: 10.1002/cncr.22496
127 Asteriti IA Giubettini M Lavia P & Guarguaglini G Aurora-A inactivation causes mitotic spindle pole fragmentation by unbalancing microtubule-generated forces. Mol Cancer. 10 131 2011. PMID: 22011530. DOI: 10.1186/1476-4598-10-131
128 Li M Dong Q & Zhu B Aurora kinase B phosphorylates histone H3.3 at serine 31 during mitosis in mammalian cells. J Mol Biol. 429(13) 2042 - 2045 2017. PMID: 28137420. DOI: 10.1016/j.jmb.2017.01.016