The Relevance of Aurora Kinase Inhibition in Hematological Malignancies

Page created by Glen Griffin
 
CONTINUE READING
The Relevance of Aurora Kinase Inhibition in Hematological Malignancies
CANCER DIAGNOSIS & PROGNOSIS
1: 111-126 (2021)                                                                                      doi: 10.21873/cdp.10016

Review

The Relevance of Aurora Kinase Inhibition
in Hematological Malignancies
CAIO BEZERRA MACHADO*, EMERSON LUCENA DA SILVA*, BEATRIZ MARIA DIAS NOGUEIRA,
JEAN BRENO SILVEIRA DA SILVA, MANOEL ODORICO DE MORAES FILHO,
RAQUEL CARVALHO MONTENEGRO, MARIA ELISABETE AMARAL DE MORAES
and CAROLINE AQUINO MOREIRA-NUNES

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

Abstract. Aurora kinases are a family of serine/threonine         In 2017 alone, nearly 24.5 million new cases of cancer
protein kinases that play a central role in eukaryotic cell       were diagnosed worldwide, and more than 9.5 million
division. Overexpression of aurora kinases in cancer and          people died from the disease. Cancer can be described as a
their role as major regulators of the cell cycle quickly          heterogeneous set of diseases that share the main clinical
inspired the idea that their inhibition might be a potential      characteristic of uncontrolled cell proliferation in the form
pathway when treating oncologic patients. Over the past           of a solid tumor or as individual cells in the bloodstream
couple of decades, the search for designing and testing of        (1, 2).
molecules capable of inhibiting aurora activities fueled many        Even though cancer is characterized as a multifactorial
pre-clinical and clinical studies. In this study, data from the   disease, the accumulation of genetic mutations, as well as the
past 10 years of in vitro and in vivo investigations, as well     deregulation of the cellular cycle, are both well-established
as clinical trials, utilizing aurora kinase inhibitors as         occurrences in cancerous cells. Failure in the regulation of
therapeutics for hematological malignancies were compiled         the cell cycle is associated with malignant neoplasms since
and discussed, aiming to highlight potential uses of these        it is fundamental for genome maintenance, repairing DNA
inhibitors as a novel monotherapy model or alongside              damage and, consequently, preventing the proliferation of
conventional chemotherapies. While there is still much to be      cells with malignant potential (3, 4).
elucidated, it is clear that these kinases play a key role in        As one of the main factors in cell-cycle control, the
oncogenesis, and their manageable toxicity and potentially        activity of protein kinases is to be highlighted due to their
synergistic effects still render them a focus of interest for     vital roles in several phases of cell division. In recent years,
future investigations in combinatorial clinical trials            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
This article is freely accessible online.
                                                                  of possible clinical relevance are the aurora kinases (5, 6).
*These Authors contributed equally to this work.
                                                                  Role of Aurora Kinases in Cell Division
Correspondence to: Caroline Aquino Moreira-Nunes, Federal
University of Ceará, Coronel Nunes de Melo st, n 1000, Rodolfo    The aurora kinases are a family of serine/threonine protein
Teófilo, CEP: 60416-000 Fortaleza, CE, Brazil. Tel: +55           kinases that play a central role in eukaryotic cell division.
8533668033, e-mail: carolfam@gmail.com
                                                                  Humans express three different types of aurora kinases:
Key Words: Hematological neoplasms, aurora kinases, targeted      aurora kinase A (AURKA), aurora kinase B (AURKB) and
molecular therapy, review.                                        aurora kinase C (AURKC). AURKA and AURKB are the
                                                                  most relevant to mitosis, being expressed in most somatic
©2021 International Institute of Anticancer Resarch               cell types, while AURKC is mostly expressed in germ cells
www.iiar-anticancer.org                                           due to its role in meiosis regulation (7, 8).

                                                                                                                              111
The Relevance of Aurora Kinase Inhibition in Hematological Malignancies
CANCER DIAGNOSIS & PROGNOSIS 1: 111-126 (2021)

   From the first reports of them being essential in cell          AURKB
division to the recent years of thorough molecular studies,
the auroras have been described as oncogenes relevant to           Along with the regulatory proteins inner centromere protein,
many human malignancies (9, 10). Their gene amplification          survivin and borealin, AURKB forms the chromosomal
and overexpression is detected particularly in solid tumors,       passenger complex (CPC) and takes part in regulating crucial
however, their correlation with leukemia development has           pathways for chromosome condensation, sister chromatid
also been reported (10, 11).                                       separation and cytokinesis (30).
   The family name as aurora was conceived when it was                During the G2 phase, at mitosis onset, AURKB is
first discovered that aurora-mutated Drosophila cells failed       responsible for phosphorylation of histone H3 (Figure 2), an
to properly duplicate and separate their centrosomes during        event that coincides with heterochromatin condensation and
mitosis, inducing the formation of monopolar spindles that,        proper chromosome formation (31-33). In vitro experiments
when observed under a microscope, would bear resemblance           utilizing analogous animal models have demonstrated AURKA
to the aurora borealis phenomenon (12). Later studies led to       to be a better kinase for the phosphorylation of H3 than
the determination of the localization of these kinases (Figure     AURKB, however, the intracellular colocalization of AURKB
1) and their actual cellular roles in humans.                      and phosphorylated H3, and the knowledge that a decrease in
                                                                   AURKB level implicates in reduced H3 phosphorylation, leads
AURKA                                                              to the understanding that in vivo models favor AURKB activity
                                                                   for histone phosphorylation (33, 34).
Prior to bipolar spindle formation, from late S phase to pro-         After chromosome condensation, during prophase in early
metaphase, AURKA is mainly concentrated at centrosomes (13,        mitosis, the activation of phosphorylated histones H3 and
14) due to an increase in cyclin-dependent kinase 11 (CDK11)       H2A recruits AURKB to the centromeres (35-37). It has been
activity (15). AURKA is then a part of important processes for     suggested that recruitment of AURKB happens in a two-step
centrosome maturation such as pericentriolar matrix recruitment    process where H2A phosphorylation by BUB1 mitotic
and microtubule nucleation and stabilization (16, 17).             checkpoint serine/threonine protein kinase (BUB1) firstly
   The role of AURKA in maturation occurs through                  creates a pool of kinetochore-bound AURKB and then H3
phosphorylation loops and protein-protein interactions with        phosphorylation by haspin kinase translocates this AURKB
many different kinases. One such pathway is AURKA-                 pool to the inner centromere, where it is mainly located
mediated phosphorylation of a large tumor suppressor 2             during prophase (38).
(LATS2) and ajuba LIM protein (AJUBA) complex which                   Until the onset of anaphase, AURKB phosphorylation of
acts on an auto-phosphorylation loop that directs AURKA to         the KMN network, an association of the conserved
the centrosomes and induces the recruitment of a                   kinetochore proteins kinetochore null protein 1 (KNL1), mis-
centrosomal pool of γ-tubulin, an essential step for               segregation 12 (MIS12) and nuclear division cycle 80
microtubule nucleation and later spindle assembly (17-20).         (NDC80), takes part in regulating kinetochore-microtubule
   During metaphase and throughout mitosis, the bipolar            attachment stabilization (39, 40). While during early mitosis
spindle assembly begins and AURKA also localizes to spindle        the attachment turnover is high, ensuring quickly
microtubules, as well as the centrosomes (21, 22). AURKA           destabilization of incorrect attachments, it tends to slow
localization to spindle microtubules is dependent on activity      down alongside mitosis progression after correct and stable
of TPX2 microtubule nucleation factor (TPX2) which binds           attachments are formed and the spindle assembly checkpoint
to and activates AURKA at the spindles, inhibiting                 is satisfied, allowing for increased activity of anaphase-
dephosphorylation by protein phosphatase 1 (PP1) (22-24).          promoting complex/cyclosome and proper chromosomal
   Aurora-related proteins have been shown to phosphorylate        separation in anaphase (38, 40-42).
kinesin family member 11 (EG5), an important microtubule-             AURKB also exerts major roles in cytokinesis, from initial
motor protein responsible for centrosome separation, in the        anaphase through telophase and the end of mitosis, being
Xenopus laevis model, thus conferring it a major role in           relocated from inner centromeres to the spindle midzone by
bipolar spindle formation (25, 26). However, it has been           the action of mitotic kinesin MKLP2 and then interacting with
demonstrated that AURKA is also involved in EG5-                   kinesins KIF4A and KIF2A to promote microtubule bundling
independent pathways for bipolar spindle assembly through          and consequent central spindle formation, as well as proper
phosphorylation of another motor protein, kinesin-12               cleavage furrow completion (43-45). It has been shown that
(KIF15/HKLP2), in human cell models (27, 28).                      after chromosomaI segregation, the activity of AURKB at the
   Besides its role in cellular spatial organization, AURKA        spindle midzone functions as a checkpoint for the regulation
also exerts a series of nuclear functions related to the control   of incorrectly segregated chromatin, inhibiting the formation
of mitotic checkpoint G2/M (Figure 2), making it a key             of tetraploid daughter cells and protecting lagging
factor in initiation of mitosis (20, 29).                          chromosomes from breakage (45, 46).

112
Machado et al: Aurora Kinases in Hematological Malignancies (Review)

Figure 1. Aurora A (AURKA) and aurora B (AURKB) localization throughout mitosis. Representation of the localization of aurora kinases and their
interactions in cell division. The main interactions of AURKA revolve around centrosomes and microtubule nucleation and stabilization, while
AURKB acts upon proper sister chromatid separation and cytokinesis.

Figure 2. Nuclear roles of aurora kinase A (AURKA) and aurora kinase B (AURKB). AURKA auto-activates and phosphorylates BORA aurora
kinase A activator (BORA), which acts as a cofactor and enhances AURKA activity. Polo-like kinase 1 (PLK1) is also phosphorylated by AURKA
and then acts upon WEE1 G2 checkpoint kinase (WEE1), a kinase responsible for the inhibition of the cyclin-dependent kinase 1 (CDK1)–cyclin B
complex, allowing initiation of mitosis through unlocking of the G2/M checkpoint. After WEE1 activation, PLK1 promotes BORA degradation and
mitosis continuation. AURKB interacts with inner centromere protein (INCEP), survivin and borealin to form mitotic regulator machinery, the
chromosomal passenger complex (CPC). AURKB phosphorylates histone H3 (HisH3) and allows chromatin reorganization in mitosis. Both AURKA
and AURKB undergo ubiquitination and are degraded at the end of mitosis (20, 29, 33).

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

                                                                                                                                           113
CANCER DIAGNOSIS & PROGNOSIS 1: 111-126 (2021)

normal cell division in AURKB-null mice with death                associated with abnormal segregation, resulting in polyploid
occurring only after implantation takes place, making it clear    cells (34). Otherwise, cells lacking both genes are able to
that AURKC may be relevant in the division of somatic cells       finish mitosis without completing anaphase, resulting in
(50, 51). While the non-overlapping functions of AURKB            polyploid cells with just one nucleus (72).
and AURKC are not fully elucidated, both kinases are known           Most studies evaluating the mechanism of AKI action in
to form distinct complexes with chromosomal passenger             hematologic cell lines focus on acute myeloid leukemia
complex proteins and act through distinct mechanisms, while       (AML). This neoplasia is characterized by clonal expansion
still being able to interfere with each other’s activities, as    of undifferentiated myeloid precursors, leading to impaired
shown by experiments of selective inhibition of one or both       hematopoiesis and bone marrow failure (73). In in vitro
kinases (52).                                                     studies, AKIs were evaluated alone or in combination with
                                                                  cytarabine, a nucleoside analog that is incorporated into
Aurora Family as Targets in                                       DNA in cell replication (S-phase) (74). The synergism of
Oncohematological Therapy                                         cytarabine with pan-AKI AMG 900 enhanced the
                                                                  antileukemic effect and attenuated polyploidization (60). In
The finding of overexpression of aurora kinases in cancer and     another study, the AURKB inhibitor barasertib used in
their role as major regulators of the cell cycle quickly led to   combination with cytarabine showed a greater-than-additive
the idea that their inhibition might be a potential pathway       cytotoxic effect with evidence of apoptosis, since barasertib
when treating oncological patients. Over the past couple of       does not prevent DNA synthesis, enabling cytarabine
decades, the quest to design and test molecules capable of        incorporation into DNA (66).
inhibiting aurora activities fueled many pre-clinical and            Internal tandem duplication of FMS-like receptor tyrosine
clinical studies, resulting in the development of a series of     kinase 3 (FLT3) is one of the most common somatic
pan-aurora inhibitors as well as selective inhibitors of one of   mutations characterized in AML. It results in the constant
the three human kinases (53). While some aurora kinase            activation of FLT3 kinase, which is linked to cancer
inhibitors (AKIs) demonstrate promising results as single         resurgence and poor patient survival (75). The dual FLT3
treatments, it is also important to perceive their potential as   inhibitor/AKI CCT241736 showed promising antileukemic
synergistic compounds, being able to restore sensitivity to       effect in vitro and in vivo, as well as in primary samples
chemotherapy agents in tumor cells and interact with other        from patients with AML, including those who were resistant
targeted therapies for increased efficacy (54).                   to the FLT3 inhibitor quizatinib, indicating that dual
   In this scenario, hematological malignant disorders as         inhibition can be a tool to manage therapies in patients with
leukemia presents overexpression of AURK, and the use of          AML presenting internal tandem duplication of FLT3 with
AKIs seems to be an effective pharmacological approach to         resistance to current chemotherapy (56).
treating these diseases (14). Table I presents studies               Translocation t(8;21) is frequent in 4-8% of patients with
published in the past 10 years describing the mechanism of        AML and results in the chimeric protein RUNX family
action of several AKIs in different leukemia cell lines, as       transcription factor 1-RUNX1 partner transcriptional co-
well as in in vivo models, and validation of AURKA and B          repressor 1 (RUNX1-RUNX1T1); 40-60% of patients
overexpression in patient samples. The studies showed that        harboring this translocation are susceptible to relapse after
the antileukemic effect of AKI AURKA and AURKB are                complete remission, this being one of the major causes of
consistently related to G2/M cell-cycle arrest, modulation of     treatment failure (76-78). Qi et al. showed that AURKB
the expression of cell cycle regulators, and polyploidy           inhibitor barasertib was more successful in inhibiting
induction resulting in apoptotic cell death.                      proliferation of AML cell lines with t(8;21) than cytarabine
   AURKA and AURKB have an important role in cell-cycle           and AURKA inhibitor alisertib, and combination of the two
progression, in fact, the transcription of AURK is cell cycle-    AKIs did not show a synergistic effect, indicating that
regulated. AURKA mRNA is found at higher concentrations           AURKB inhibitor may be a more efficient inhibitor in the
in the G2/M phase, its protein level reaching higher a little     treatment of patients with a specific subtype of AML (58).
later; the same happens to AURKB, with enhanced levels of            Acute lymphoblastic leukemia (ALL) is the most common
mRNA and protein expression just after those of AURKA             leukemia diagnosed in adults and the second most frequent
(14). Inhibition of AURKA expression by genetic knockdown         acute subtype in children worldwide, this neoplasia presents
was shown to lead to errors in cell mitotic processes, once       chromosomal and genetic alterations related to the
the spindle checkpoint detects a failure in chromosome            differentiation and proliferation of T and B precursor cells
alignment, it culminates in mitotic arrest and cellular death     (79, 80). AURKA and AURKB are overexpressed in samples
by apoptosis (71). The knockdown of AURKB leads to                from patients with ALL (55, 65, 81) and some research
reduction in histone H3 phosphorylation required in               described the mechanism of action of AKI in ALL cell lines
chromosome condensation and cytokinesis, these errors are         alone or in combination with other antineoplastics agents.

114
Machado et al: Aurora Kinases in Hematological Malignancies (Review)

   Genetic alterations as t(4;11) with mixed lineage leukemia      without BCR–ABL1T315I mutation, and also in ALL Ph− cell
(MLL) fusion–associated gene AF4 (MLL-AF4) and t(9;22)             lines. Danusertib induced cytotoxicity in all cells
with breakpoint cluster region–Abelson murine leukemia 1           independently of BCR–ABL1T315I mutation and Ph presence,
(BCR–ABL1) are frequent in ALL and are related to treatment        as well as reducing the percentage of leukemia cells
resistance, poor outcome, and disease relapse (65, 82).            (CD10+/CD19+) and enhancing the survival rates of mice
Rearrangements of MLL gene represents 10% of ALL cases             transplanted with cells with BCR–ABL1T315I mutation (67).
and MLL–AF4 fusion is present in the majority of ALL cases         The study also attested to synergism in the cytotoxic effect
(57%), is highly associated with rapid cancer development,         of the pan-AKI danusertib in combination with
fast progression, and poor prognosis in comparison with            farnesyltransferase inhibitor, vincristine, and with the TKI
patients without this rearrangement (83, 84).                      dasatinib independently of BCR–ABL1T315I mutation,
   In in vitro studies evaluating the cytotoxicity of the pan-     suggesting danusertib as a new option for chemotherapy in
AKIs VX-680 and VE-465 against a panel of ALL cell lines,          patients with ALL, since it is effective independently of Ph
cell lines with MLL–AF4 fusion showed higher sensitivity to        presence or BCR–ABL1T315I mutation, and, especially for
AKI treatment, and VE-465 gave better results when                 patients with BCR–ABL1T315I mutation who are resistant to
compared to VX-68. Curiously, the same study reported that         actual treatment with TKIs.
these cell lines presented AURKB protein expression higher            CML is basically, but not only, characterized by the
than AURKA (65). A recent study conducted by Moreira-              presence of Ph+ cells, since around 95% of cases present this
Nunes et al. observed no difference between AURKA and              translocation (94, 95). Thus, the malignant potential of CML
AURKB gene expression in samples with MLL–AF4 fusion               cells is related to the BCR–ABL1 chimeric gene, which is
from patients with ALL when compared to other frequent             also its pharmacologic target (85, 96). Resistance to TKIs in
fusions in leukemia [BCR–ABL1, transcription factor 3 (E2A         CML is widely documented and occurs in 20-40% of cases
immunoglobulin enhancer-binding factors E12/E47) and pre-          (97), furthermore, the overexpression of AURKA in several
B-cell leukemia transcription factor 1 (E2A–PBX1)                  cancer types is described as one mechanism of
translocation, stem leukemia cell interrupting locus and T-        chemoresistance (98-104).
cell acute lymphocytic leukemia protein 1 (SIL–TAL1)                  In vitro studies monitored the antileukemia effect of pan-
fusion, translocation–Ets–leukemia and acute myeloid               AKIs GW809897X and GW806742X in a non-resistant
leukemia 1 protein (TEL–AML1) fusion]. In contrast, when           CML cell line, with promising results (55). Long et al.
the expression of AUKA and B were analyzed in all patients         showed that AURKA inhibitor AKI603 suppressed cell
in the study, AURKB expression was higher than that of             growth and induced cell differentiation of both imatinib-
AURKA and overexpression was correlated with lower                 resistant (BCR–ABL1T315I) and non-resistant cell lines (64).
survival rates (55). Thus, due to the role of AURKB in ALL,        A similar study showed that AURKA inhibitor alisertib,
and even the lack of evidence that correlates the expression       when used alone, reduced viability of CML cell lines, both
of this kinase with outcome in those with MLL–AF4, more            non-resistant and resistant to imatinib and nilotinib, as well
studies evaluating AURKB inhibitors in therapy of MLL–             as cells harboring BCR–ABL1T315I mutations. Moreover,
AF4 ALL are still necessary.                                       when administered in combination with the TKIs imatinib,
   The Philadelphia chromosome (Ph+) is characterized by           ponatinib or nilotinib, there was a disruption in the resistance
the translocation between chromosomes 9 and 22, generating         mechanism and reduced rates of cell growth were observed
the chimeric gene BCR–ABL1 (85, 86). Although this                 in the respective resistant cell lines. Moreover, alisertib plus
translocation is more described in chronic myeloid leukemia        ponatinib showed an additive effect in reducing tumor
(CML), the presence of Ph affects around 3-5% of children          volume and enhanced the rate of survival in mice
and 25% of adults with ALL, and is related to a poor               transplanted with CML BCR–ABL1T315I+ cells (61).
prognosis (87-89). The chimeric BCR–ABL1 gene is                   Inhibition of AURKA seems to reduce the profile of
constitutively active and overexpresses a tyrosine-kinase          resistance in CML cell lines in vitro and in vivo; the
receptor that is involved in signaling pathways linked to          utilization of AKIs in combination with conventional
cellular proliferation, de-differentiation, and apoptosis escape   chemotherapy may improve the outcome of patients that are
(90). Although tyrosine-kinase inhibitors (TKIs) such as           unresponsive to chemotherapy for CML.
imatinib, bosutinib, dasatinib, and nilotinib were developed,         Lymphomas are a group of malignant diseases that can be
mutations in catalytic enzymatic domains as ABL1T315I result       derived from constituent cells of the lymphoid tissue,
in TKI resistance, leading to risk of treatment failure in         lymphocytes and histiocytes. Malignant lymphomas are
patients with CML or ALL Ph+ (67, 91-93).                          divided into Hodgkin’s and non-Hodgkin’s lymphoma
   Due to the efficacy of AKIs in leukemia cell lines in vitro     (NHL) and are more common in the head and neck region,
and in in vivo studies, Fei et al. evaluated the pan-AKI           but NHL can also be found in extranodal regions (25% of
danusertib (PHA-739358) in human ALL cell lines with and           the cases), with or without lymph node involvement. NHL is

                                                                                                                               115
CANCER DIAGNOSIS & PROGNOSIS 1: 111-126 (2021)

Table I. In vitro studies utilizing Aurora-kinase inhibitors as a monotherapy or in combination with synergetic treatments in the past 10 years.

Kinase              Study         Leukemia         Cell    Genetic alterations      Aurora      Synergistic            Mechanism            Reference
assessed            phase          subtype         line        assessed            inhibitor     treatment

AURKA/B/C       In vitro with       ALL           K-562        BRC–ABL1          GW809897X          NR                Cell death               55
                validation in       CML                         E2A–PBX          GW806742X                     Caspase 3 and 7 activation
               patient samples                                 MLL–AF4                                           G2/M cell-cycle arrest
                                                                SIL–TAL
                                                               EL–AML1

AURKA/B/C In vitro, in vivo         AML        MOLM-13          FLT3–ITD         CCT241736          NR             Reduced viability           56
          with validation                       MV4-11                                                                 Apoptosis
             in patient                                                                                          G2/M cell-cycle arrest
              samples                                                                                           Reduces phosphorylation
                                                                                                                   of FLT3, STAT5,
                                                                                                                 MAPK and AURKA/B
                                                                                                                 Reduced tumor growth
                                                                                                                and STAT5 and AURKA
                                                                                                                    phosphorylation

AURKA            In vitro and       AML          HL-60             NR              AKI604           NR          Inhibition of proliferation    57
                    in vivo                      U937                                                             and colony formation
                                                                                                                Mitochondrial impairment
                                                                                                               Elevation of ROS as well as
                                                                                                              cellular calcium (Ca2+) levels
                                                                                                                Increase in polyploid cells
                                                                                                               Inhibition of tumor growth

AURKA/B/C          In vitro         AML         Kasumi-1        RUNX1–            Alisertib         NR         Reduction in proliferation      58
                                                 Skno-1         RUNX1T1          (MLN8237)                      G2/M cell-cycle arrest
                                                                                  Barasertib                  Modulation in the expression
                                                                              (AZD1152-HQPA)                    of cell cycle regulators

AURKA              In vitro         AML         Kasumi-1           NR              Radotinib        NR                Cytotoxicity             59
                                                                                                                 Reduced expression of
                                                                                                                    cyclin-dependent
                                                                                                                 kinase 1 and cyclin B1
                                                                                                                Reduced phosphorylation
                                                                                                                       of AURKA
                                                                                                                 G2/M cell cycle arrest

AURKA/B/C          In vitro         AML        MOLM-13          FLT3–ITD          AMG 900       Cytarabine     Reduction in proliferation      60
                                                MV-411            TP53                                         Induces polyploidization
                                                 KG-1             P–gp                                              and apoptosis
                                                MEGAL
                                                 U937
                                                 HEL
                                              CHRF-288-11

AURKA            In vitro and       CML          K-562         BRC–ABL1           Alisertib Imatinib and Blocked cell proliferation            61
                    in vivo                      Ba/F3           BRC–            (MLN8237) Ponatinib            Apoptosis
                                               K-562/IR        ABL1T315I                                 AURKA phosphorylation
                                              K-562/NILR                                                  G2/M cell cycle arrest
                                                                                                          Enhanced ROS levels
                                                                                                          Reduced tumor growth

AURKA              In vitro         NHL          DOHH2            MYC             Alisertib      ABT-199        Suppressed cell growth         62
                                   (DHL)          VAL             BCL2           (MLN8237)                       G2/M cell cycle arrest
                                                                                                                       Apoptosis
                                                                                                              Enhanced expression of cell
                                                                                                              intrinsic apoptotic proteins
                                                                                                                Reduction in AURKA
                                                                                                                  phosphorylation and
                                                                                                                    MYC expression

                                                                                                                                  Table I. Continued

116
Machado et al: Aurora Kinases in Hematological Malignancies (Review)

Table I. Continued

Kinase               Study        Leukemia       Cell      Genetic alterations     Aurora      Synergistic          Mechanism               Reference
assessed             phase         subtype       line          assessed           inhibitor     treatment

AURKB                In vivo        AML        HL-60           FLT3–ITD           Barasertib   Cytarabine      Inhibited tumor growth          63
                                              OCI-AML3                           (AZD1152-                   Increased phospho-histone
                                               MV4-11                              HQPA)                     H3 inhibition, polyploidy,
                                              MOLM-13                                                         and tumor cell apoptosis

AURKA                In vitro       CML        KBM-5          BRC–ABL1            AKI603          NR           Reduced cell viability          64
                                              KBM-5T315I        BRC–                                         Inhibited phosphorylation
                                                              ABL1T315I                                        of AURKA at Thr288
                                                                                                             G2/M cell-cycle arrest with
                                                                                                              polyploidy accumulation
                                                                                                                     Apoptosis

AURKA/B/C       In vitro with       ALL         RS4;11         MLL–AF4            VE-465          NR                Cytotoxicity               65
                validation in                   MV4;11          TP53              VX-680                       Inhibition of AURKA
               patient samples                 CCRF-SB                                                            phosphorylation
                                               MOLT-3                                                          G2/M cell-cycle arrest
                                               MOLT-4                                                              Apoptosis and
                                              RPMI-8402                                                            accumulation
                                               SUP-B15                                                             of polyploidy
                                              CCRF-CEM

AURKB                In vitro       AML         HL-60             NR              Barasertib   Cytarabine     Increase in cytotoxicity         66
                                             HL-60/ara-C20                       (AZD1152-                      Induced polyploidy
                                                 U937                              HQPA)                             Apoptosis
                                                                                                                 Inhibited AURKB
                                                                                                               auto-phosphorylation

AURKA/B/C       In vitro and        ALL        UCSF02         BRC–ABL1           Danusertib Lonafarnib,          Reduced viability             67
                   in vivo                      TXL2            BRC–               (PHA-    Vincristine          Induced apoptosis
                                                BLQ1          ABL1T315I           739358) and Dasatinib            and polyploidy
                                                 Pt2            TP53                                                  G2/M cell
                                                 US6                                                                 cycle arrest
                                                 US7                                                            Reduced BCR–ABL
                                                US7R                                                                and AURKB
                                                                                                               phosphorylation in vivo

AURKB           In vitro and        NHL     TOLEDO               TP53             AT9283       Docetaxel     Inhibited cell proliferation      68
                   in vivo        (DLBCL,      RL                 p16                                               and AURKB
                                 MCL, T-FL,   MCL                                                                 phosphorylation
                                  Burkitt’s Granta-519                                                        Increased polyploid cells
                                 lymphoma)   Granta-4                                                                 Apoptosis
                                            SUDHL-4                                                            Inhibited tumor growth
                                            SUDHL-6
                                               Raji

AURKA           In vitro with  AML stem          NB4              NR               C1368       Cytarabine        Inhibition of cell            69
                validation in    cells           KG1                                                               proliferation
               patient samples                                                                                      Apoptosis
                                                                                                               Reduction of AURKA
                                                                                                                 phosphorylation
                                                                                                                  and expression

AURKA                In vitro       NHL     Granta-519           TP53            MK-8745          NR          G2/M cell-cycle arrest           70
                                   (MCL,      Jeko1                                                          Inhibited phosphorylation
                                  Burkitt’s   JVM2                                                                  of AURKA
                                 Lymphoma,    Z138C                                                                  Apoptosis
                                    and       Akata
                                  NK-cells   DHL16
                                 lymphoma) SUDHL-16
                                              KAI3
                                              SNK6

ALL: Acute lymphoblastic leukemia; AML: acute myeloid leukemia; ARA-C: cytarabine; AURKA: aurora kinase a; AURKB: aurora kinase b; CLL:
chronic lymphoblastic leukemia; CML: chronic myeloid leukemia; DHL: double-hit lymphoma; DLBCL: diffuse large B-cell lymphoma; IR: imatinib
resistant; MCL: mantle-cell lymphoma; NHL: non-Hodgkin lymphoma; NR: not reported; NK: natural killer; NILR: nilotinib resistant; T-FL:
transformed follicular lymphoma.

                                                                                                                                                    117
CANCER DIAGNOSIS & PROGNOSIS 1: 111-126 (2021)

more frequent than Hodgkin’s disease and corresponds to            123), a selective inhibitor of AURKA that is also under
around 90% of head and neck malignancies (105, 106). Of            investigation for relevance in the treatment of non-
NHL subtypes, follicular and diffuse large B-cell lymphomas        hematological malignancies (125). While it was most
are the most frequently diagnosed, corresponding to about          effective when treating patients with AML, this finding may
20% and 30%, respectively. Other subtypes account for              also be correlated to its use as a synergistic compound
fewer than 10% of the cases (107).                                 alongside conventional induction chemotherapy (113, 114).
   Studies have shown that AURKA is overexpressed in               A study cohort by Goldberg et al. demonstrated alisertib to
several NHL subtypes when compared to normal tissues and           be potentially clinically effective for patients with AML,
B-cells, Chowdhury et al. evaluated the protein expression         while having no efficacy when treating those with
of AURKA and the potential of AURKA inhibitor, MK-8745,            myelodysplastic syndrome, hinting towards a better
in NHL derived from Mantle cell, Burkitt’s, and natural            understanding of the drug mechanism of action and
killer-cell lymphomas. Most of the cell lines analyzed             biological pathway (122).
presented enhanced AURKA expression but the results                   While most investigated drugs were AURKA-selective
showed that treatment with pan-AKI VE-465 and taxol,               inhibitors or pan-AKIs, AURKB was selectively targeted by
when used alone, had higher toxicity than the selective            the use of barasertib in two studies (119, 124). Although its
inhibitor. However, the antitumoral effect of AURKA                efficacy was demonstrated in the treatment of patients with
inhibitor was correlated with the expression of AURKA              diffuse large B-cell lymphoma, the low progression-free
activator TPX2 (70).                                               survival of 60 days attested to by Collins et al. discredits its
   Moreover, the pan-AKI AT9283 exhibited highly cytotoxic         use as a monotherapy for this disease (119). When treating
effects in NHL cells and, when used in combination with            patients with AML, Kantarjian et al. demonstrated
docetaxel, enhanced apoptosis, as well as survival rates and       barasertib to achieve better complete response rates as a
reduced tumor volume in the mouse mantle cell lymphoma             monotherapy than low-dose cytosine arabinoside, an agent
xenograft model (68). The synergistic effect of AURK               previously demonstrated to be clinically beneficial in
inhibition was observed with other antitumoral agents that         clinical trials (124, 126).
target the cell cycle and microtubular constituents. The              Other inhibitors that were reported in only one study
disruption in replication machinery leads to selective cancer      include AMG 900, AT9283, ENMD-2076 and MK-0457.
cell death, enhancing the antitumoral potential of AKI in          These drugs had only modest to no efficacy when utilized as
oncohematological treatment (108, 109).                            single agents and in low, tolerable doses, although ENMD-
   Kong et al. evaluated the synergism between the inhibitors      2076 monotherapy still achieved a 25% overall response rate
of AURKA, alisertib, and BCL2 apoptosis regulator (BCL2)           when treating patients with AML and CML, and should be
ABT-199, in cell lines of the NHL subtype double-hit               taken into account when considering possible synergistic
lymphoma. Interestingly, the BCL2 inhibitor showed better          interactions (115-117, 121).
results in vitro than AURKA inhibitor but the ABT-199 plus            Overall, the usage of AKIs did not cause unexpected or
alisertib treatment synergistically inhibited viability in         unmanageable adverse effects in patients and the main ones
double-hit lymphoma cells (62). This subtype of NHL is             were related to blood or gastrointestinal disorders (115, 117,
characterized by MYC proto-oncogene (MYC) and BCL2                 119-121, 123), which is in accordance with the expected
translocation, MYC and AURKA are regulated by each other           effects of aurora inhibition in non-malignant rapidly dividing
in a feedback pathway and exert a strong role in the cell          cells due to mitosis impairment (127, 128).
cycle and replicative process (110). BCL2 regulates the
apoptotic process and has a function in survival and               Conclusion
chemoresistance in hematological malignancies (111, 112).
Thus, dual inhibitory therapy focusing on AURKA and                The interest in understanding the involvement of aurora in
BCL2 may be an interesting option for treatment in patients        biological pathways in non-malignant as well as in malignant
with double-hit lymphoma with these specific mutations.            cells has only grown over the years. While there is still much
   Table II presents clinical trials from the past 10 years        to be elucidated, it is clear that these kinases and their
utilizing AKIs as a therapeutic option for patients afflicted by   upstream and downstream regulators play a key role in
leukemia and other hematological disorders. The most common        mitosis and oncogenesis, motivating their investigation as
studied disorder was AML, appearing in seven out of 12             potential targets for oncological treatments. Even though the
studies (113-117, 122, 124), while the most targeted kinase was    clinical trials using AKIs as a monotherapy for hematological
AURKA, appearing in 10 of 12 studies (113-118, 120-123).           disorders have not shown great results, the manageable
AURKC was not a main target in any of the selected studies.        toxicity and potentially synergistic effects still render them
   In all clinical trials presented, the most prevalent            a focus of interest for future investigations in combinatorial
therapeutic option was alisertib (113, 114, 118, 120, 122,         clinical trials.

118
Machado et al: Aurora Kinases in Hematological Malignancies (Review)

Table II. Clinical trials utilizing aurora-kinase inhibitors as a monotherapy or in combination with synergetic treatments in the past 10 years.

Kinase assessed     Study              Patients             Leukemia          Aurora           Synergistic            Prognostic         Reference
                    phase              enrolled              subtype         inhibitor          treatment            significance

AURKA                 II          39 (64% Male)                AML          Alisertib        Cytarabine and         Remission was           113
                                Median age=67 years                        (MLN8237)           idarubicin          achieved in 64%

AURKA                 I           22 (68% Male)                AML          Alisertib        Cytarabine and         Remission was           114
                               Median age=62.7 years                       (MLN8237)           idarubicin          achieved in 86%

AURKA/B               I           35 (66% Male)                AML          AMG 900                NR            Complete response          115
                                Median age=69 years                                                             with incomplete count
                                                                                                                    recovery was
                                                                                                                   achieved in 9%

AURKA/B              I/II          7 (71% Male)                AML           AT9283                NR            None of the patients       116
                                 Median age=3 years            ALL                                               achieved a partial or
                                                                                                                  complete remission

AURKA                 I           27 (74% Male)                AML         ENMD-2076               NR              Overall response         117
                                Median age=69 years            CML                                                  rate was 25%

AURKA                 I          26 (61.5% Male)               MM           Alisertib          Bortezomib          Overall response         118
                               Median age=64.5 years                       (MLN8237)                               rate was 26.9%

AURKB                 II          15 (53% Male)              DLBCL         Barasertib              NR              Overall response         119
                                Median age=65 years                     (AZD1152-HQPA)                              rate was 20%

AURKA                 II          48 (73% Male)           Non-Hodgkin       Alisertib              NR              Overall response         120
                               Median age=67.5 years       lymphoma        (MLN8237)                                rate was 27%

AURKA/B               II          52 (65% Male)                CML          MK-0457                NR          Only 3.8% had complete       121
                                Median age=52 years            ALL                                                 cytogenetic and
                                                                                                               hematological response

AURKA                 II          57 (56% Male)                AML          Alisertib              NR              Overall response         122
                                Median age=72 years            MDS         (MLN8237)                                rate was 13%

AURKA                 I           58 (47% Male)                MM           Alisertib              NR              Overall response         123
                                Median age=61 years            NHL         (MLN8237)                                rate was 27%
                                                               CLL

AURKB                 II         77 (58.4% Male)               AML         Barasertib              NR             Complete response         124
                                Median age=76 years                     (AZD1152-HQPA)                              rate of 35.4%

ALL: Acute lymphoid leukemia; AML: acute myeloid leukemia; AURKA: aurora kinase A; AURKB: aurora kinase B; CML: chronic myeloid
leukemia; DLBCL: diffuse large B-cell lymphoma; MDS: myelodysplastic syndrome; MM: multiple myeloma; NHL: non-Hodgkin’s lymphoma;
NR: not reported.

Conflicts of Interest                                                       Moraes-Filho MO, Moraes MEA, Montenegro RC and Moreira-
                                                                            Nunes CA wrote the article. All Authors read and approved the final
The Authors declare no conflicts of interest regarding this study.          article.

Authors’ Contributions                                                      Acknowledgements

Machado CB, da Silva EL and Moreira-Nunes CA, performed the                 This study was supported by Brazilian funding agencies National
study design; Machado CB and Nogueira BMD, prepared the                     Counsel of Technological and Scientific Development (CNPq; to
figures; Machado CB, da Silva EL, Nogueira BMD, da Silva JBS,               ELS, RCM, MEAM, MOMF and CAMN).

                                                                                                                                                   119
CANCER DIAGNOSIS & PROGNOSIS 1: 111-126 (2021)

References                                                                Kaul A, Keiyoro PN, Kelbore AG, Kerbo AA, Khader YS,
                                                                          Khalilarjmandi M, Khan EA, Khan G, Khang YH, Khatab K,
1     Global Burden of Disease Cancer Collaboration, Fitzmaurice C,       Khater A, Khayamzadeh M, Khazaee-Pool M, Khazaei S, Khoja
      Abate D, Abbasi N, Abbastabar H, Abd-Allah F, Abdel-Rahman          AT, Khosravi MH, Khubchandani J, Kianipour N, Kim D, Kim
      O, Abdelalim A, Abdoli A, Abdollahpour I, Abdulle ASM, Abebe        YJ, Kisa A, Kisa S, Kissimova-Skarbek K, Komaki H, Koyanagi
      ND, Abraha HN, Abu-Raddad LJ, Abualhasan A, Adedeji IA,             A, Krohn KJ, Bicer BK, Kugbey N, Kumar V, Kuupiel D, La
      Advani SM, Afarideh M, Afshari M, Aghaali M, Agius D,               Vecchia C, Lad DP, Lake EA, Lakew AM, Lal DK, Lami FH, Lan
      Agrawal S, Ahmadi A, Ahmadian E, Ahmadpour E, Ahmed MB,             Q, Lasrado S, Lauriola P, Lazarus JV, Leigh J, Leshargie CT, Liao
      Akbari ME, Akinyemiju T, Al-Aly Z, AlAbdulKader AM,                 Y, Limenih MA, Listl S, Lopez AD, Lopukhov PD, Lunevicius
      Alahdab F, Alam T, Alamene GM, Alemnew BTT, Alene KA,               R, Madadin M, Magdeldin S, El Razek HMA, Majeed A, Maleki
      Alinia C, Alipour V, Aljunid SM, Bakeshei FA, Almadi MAH,           A, Malekzadeh R, Manafi A, Manafi N, Manamo WA,
      Almasi-Hashiani A, Alsharif U, Alsowaidi S, Alvis-Guzman N,         Mansourian M, Mansournia MA, Mantovani LG, Maroufizadeh
      Amini E, Amini S, Amoako YA, Anbari Z, Anber NH, Andrei CL,         S, Martini SMS, Mashamba-Thompson TP, Massenburg BB,
      Anjomshoa M, Ansari F, Ansariadi A, Appiah SCY, Arab-Zozani         Maswabi MT, Mathur MR, McAlinden C, McKee M, Meheretu
      M, Arabloo J, Arefi Z, Aremu O, Areri HA, Artaman A, Asayesh        HAA, Mehrotra R, Mehta V, Meier T, Melaku YA, Meles GG,
      H, Asfaw ET, Ashagre AF, Assadi R, Ataeinia B, Atalay HT, Ataro     Meles HG, Melese A, Melku M, Memiah PTN, Mendoza W,
      Z, Atique S, Ausloos M, Avila-Burgos L, Avokpaho EFGA,              Menezes RG, Merat S, Meretoja TJ, Mestrovic T, Miazgowski B,
      Awasthi A, Awoke N, Ayala Quintanilla BP, Ayanore MA, Ayele         Miazgowski T, Mihretie KMM, Miller TR, Mills EJ, Mir SM,
      HT, Babaee E, Bacha U, Badawi A, Bagherzadeh M, Bagli E,            Mirzaei H, Mirzaei HR, Mishra R, Moazen B, Mohammad DK,
      Balakrishnan S, Balouchi A, Bärnighausen TW, Battista RJ,           Mohammad KA, Mohammad Y, Darwesh AM, Mohammadbeigi
      Behzadifar M, Behzadifar M, Bekele BB, Belay YB, Belayneh           A, Mohammadi H, Mohammadi M, Mohammadian M,
      YM, Berfield KKS, Berhane A, Bernabe E, Beuran M, Bhakta N,         Mohammadian-Hafshejani A, Mohammadoo-Khorasani M,
      Bhattacharyya K, Biadgo B, Bijani A, Bin Sayeed MS, Birungi         Mohammadpourhodki R, Mohammed AS, Mohammed JA,
      C, Bisignano C, Bitew H, Bjørge T, Bleyer A, Bogale KA, Bojia       Mohammed S, Mohebi F, Mokdad AH, Monasta L, Moodley Y,
      HA, Borzì AM, Bosetti C, Bou-Orm IR, Brenner H, Brewer JD,          Moosazadeh M, Moossavi M, Moradi G, Moradi-Joo M, Moradi-
      Briko AN, Briko NI, Bustamante-Teixeira MT, Butt ZA, Carreras       Lakeh M, Moradpour F, Morawska L, Morgado-da-Costa J,
      G, Carrero JJ, Carvalho F, Castro C, Castro F, Catalá-López F,      Morisaki N, Morrison SD, Mosapour A, Mousavi SM, Muche
      Cerin E, Chaiah Y, Chanie WF, Chattu VK, Chaturvedi P,              AA, Muhammed OSS, Musa J, Nabhan AF, Naderi M, Nagarajan
      Chauhan NS, Chehrazi M, Chiang PP, Chichiabellu TY, Chido-          AJ, Nagel G, Nahvijou A, Naik G, Najafi F, Naldi L, Nam HS,
      Amajuoyi OG, Chimed-Ochir O, Choi JJ, Christopher DJ, Chu           Nasiri N, Nazari J, Negoi I, Neupane S, Newcomb PA, Nggada
      DT, Constantin MM, Costa VM, Crocetti E, Crowe CS, Curado           HA, Ngunjiri JW, Nguyen CT, Nikniaz L, Ningrum DNA, Nirayo
      MP, Dahlawi SMA, Damiani G, Darwish AH, Daryani A, das              YL, Nixon MR, Nnaji CA, Nojomi M, Nosratnejad S, Shiadeh
      Neves J, Demeke FM, Demis AB, Demissie BW, Demoz GT,                MN, Obsa MS, Ofori-Asenso R, Ogbo FA, Oh IH, Olagunju AT,
      Denova-Gutiérrez E, Derakhshani A, Deribe KS, Desai R,              Olagunju TO, Oluwasanu MM, Omonisi AE, Onwujekwe OE,
      Desalegn BB, Desta M, Dey S, Dharmaratne SD, Dhimal M, Diaz         Oommen AM, Oren E, Ortega-Altamirano DDV, Ota E,
      D, Dinberu MTT, Djalalinia S, Doku DT, Drake TM, Dubey M,           Otstavnov SS, Owolabi MO, P A M, Padubidri JR, Pakhale S,
      Dubljanin E, Duken EE, Ebrahimi H, Effiong A, Eftekhari A, El       Pakpour AH, Pana A, Park EK, Parsian H, Pashaei T, Patel S,
      Sayed I, Zaki MES, El-Jaafary SI, El-Khatib Z, Elemineh DA,         Patil ST, Pennini A, Pereira DM, Piccinelli C, Pillay JD, Pirestani
      Elkout H, Ellenbogen RG, Elsharkawy A, Emamian MH,                  M, Pishgar F, Postma MJ, Pourjafar H, Pourmalek F, Pourshams
      Endalew DA, Endries AY, Eshrati B, Fadhil I, Fallah Omrani V,       A, Prakash S, Prasad N, Qorbani M, Rabiee M, Rabiee N, Radfar
      Faramarzi M, Farhangi MA, Farioli A, Farzadfar F, Fentahun N,       A, Rafiei A, Rahim F, Rahimi M, Rahman MA, Rajati F, Rana
      Fernandes E, Feyissa GT, Filip I, Fischer F, Fisher JL, Force LM,   SM, Raoofi S, Rath GK, Rawaf DL, Rawaf S, Reiner RC,
      Foroutan M, Freitas M, Fukumoto T, Futran ND, Gallus S,             Renzaho AMN, Rezaei N, Rezapour A, Ribeiro AI, Ribeiro D,
      Gankpe FG, Gayesa RT, Gebrehiwot TT, Gebremeskel GG,                Ronfani L, Roro EM, Roshandel G, Rostami A, Saad RS,
      Gedefaw GA, Gelaw BK, Geta B, Getachew S, Gezae KE,                 Sabbagh P, Sabour S, Saddik B, Safiri S, Sahebkar A, Salahshoor
      Ghafourifard M, Ghajar A, Ghashghaee A, Gholamian A, Gill PS,       MR, Salehi F, Salem H, Salem MR, Salimzadeh H, Salomon JA,
      Ginindza TTG, Girmay A, Gizaw M, Gomez RS, Gopalani SV,             Samy AM, Sanabria J, Santric Milicevic MM, Sartorius B,
      Gorini G, Goulart BNG, Grada A, Ribeiro Guerra M, Guimaraes         Sarveazad A, Sathian B, Satpathy M, Savic M, Sawhney M,
      ALS, Gupta PC, Gupta R, Hadkhale K, Haj-Mirzaian A, Haj-            Sayyah M, Schneider IJC, Schöttker B, Sekerija M, Sepanlou SG,
      Mirzaian A, Hamadeh RR, Hamidi S, Hanfore LK, Haro JM,              Sepehrimanesh M, Seyedmousavi S, Shaahmadi F, Shabaninejad
      Hasankhani M, Hasanzadeh A, Hassen HY, Hay RJ, Hay SI,              H, Shahbaz M, Shaikh MA, Shamshirian A, Shamsizadeh M,
      Henok A, Henry NJ, Herteliu C, Hidru HD, Hoang CL, Hole MK,         Sharafi H, Sharafi Z, Sharif M, Sharifi A, Sharifi H, Sharma R,
      Hoogar P, Horita N, Hosgood HD, Hosseini M, Hosseinzadeh M,         Sheikh A, Shirkoohi R, Shukla SR, Si S, Siabani S, Silva DAS,
      Hostiuc M, Hostiuc S, Househ M, Hussen MM, Ileanu B, Ilic           Silveira DGA, Singh A, Singh JA, Sisay S, Sitas F, Sobngwi E,
      MD, Innos K, Irvani SSN, Iseh KR, Islam SMS, Islami F, Jafari       Soofi M, Soriano JB, Stathopoulou V, Sufiyan MB, Tabarés-
      Balalami N, Jafarinia M, Jahangiry L, Jahani MA, Jahanmehr N,       Seisdedos R, Tabuchi T, Takahashi K, Tamtaji OR, Tarawneh MR,
      Jakovljevic M, James SL, Javanbakht M, Jayaraman S, Jee SH,         Tassew SG, Taymoori P, Tehrani-Banihashemi A, Temsah MH,
      Jenabi E, Jha RP, Jonas JB, Jonnagaddala J, Joo T, Jungari SB,      Temsah O, Tesfay BE, Tesfay FH, Teshale MY, Tessema GA,
      Jürisson M, Kabir A, Kamangar F, Karch A, Karimi N, Karimian        Thapa S, Tlaye KG, Topor-Madry R, Tovani-Palone MR, Traini
      A, Kasaeian A, Kasahun GG, Kassa B, Kassa TD, Kassaw MW,            E, Tran BX, Tran KB, Tsadik AG, Ullah I, Uthman OA, Vacante

120
Machado et al: Aurora Kinases in Hematological Malignancies (Review)

     M, Vaezi M, Varona Pérez P, Veisani Y, Vidale S, Violante FS,                centrosome maturation and bipolar spindle formation. EMBO Rep
     Vlassov V, Vollset SE, Vos T, Vosoughi K, Vu GT, Vujcic IS,                  7(4): 418-424, 2006. PMID: 16462731. DOI: 10.1038/
     Wabinga H, Wachamo TM, Wagnew FS, Waheed Y, Weldegebreal                     sj.embor.7400639
     F, Weldesamuel GT, Wijeratne T, Wondafrash DZ, Wonde TE,                16   Barr AR and Gergely F: Aurora-A: the maker and breaker of
     Wondmieneh AB, Workie HM, Yadav R, Yadegar A, Yadollahpour                   spindle poles. J Cell Sci 120(Pt 17): 2987-2996, 2007. PMID:
     A, Yaseri M, Yazdi-Feyzabadi V, Yeshaneh A, Yimam MA, Yimer                  17715155. DOI: 10.1242/jcs.013136
     EM, Yisma E, Yonemoto N, Younis MZ, Yousefi B, Yousefifard              17   Wang G, Jiang Q and Zhang C: The role of mitotic kinases in
     M, Yu C, Zabeh E, Zadnik V, Moghadam TZ, Zaidi Z, Zamani M,                  coupling the centrosome cycle with the assembly of the mitotic
     Zandian H, Zangeneh A, Zaki L, Zendehdel K, Zenebe ZM,                       spindle. J Cell Sci 127(Pt 19): 4111-4122, 2014. PMID:
     Zewale TA, Ziapour A, Zodpey S and Murray CJL: Global,                       25128564. DOI: 10.1242/jcs.151753
     regional, and national cancer incidence, mortality, years of life       18   Abe Y, Ohsugi M, Haraguchi K, Fujimoto J and Yamamoto T:
     lost, years lived with disability, and disability-adjusted life-years        LATS2-Ajuba complex regulates gamma-tubulin recruitment to
     for 29 cancer groups, 1990 to 2017: a systematic analysis for the            centrosomes and spindle organization during mitosis. FEBS Lett
     global burden of disease study. JAMA Oncol 5(12): 1749-1768,                 580(3): 782-788, 2006. PMID: 16413547. DOI: 10.1016/
     2019. PMID: 31560378. DOI: 10.1001/jamaoncol.2019.2996                       j.febslet.2005.12.096
2    Hassanpour S and Dehghani M: Review of cancer from                      19   Toji S, Yabuta N, Hosomi T, Nishihara S, Kobayashi T, Suzuki S,
     perspective of molecular. Journal of Cancer Research and Practice            Tamai K and Nojima H: The centrosomal protein Lats2 is a
     4(4): 127-129, 2019. DOI: 10.1016/j.jcrpr.2017.07.001                        phosphorylation target of Aurora-A kinase. Genes Cells 9(5): 383-
3    Hanahan D and Weinberg RA: The hallmarks of cancer. Cell                     397, 2004. PMID: 15147269. DOI: 10.1111/j.1356-9597.2004.
     100(1): 57-70, 2000. PMID: 10647931. DOI: 10.1016/s0092-                     00732.x
     8674(00)81683-9                                                         20   Willems E, Dedobbeleer M, Digregorio M, Lombard A, Lumapat
4    Foster I: Cancer: A cell cycle defect. Radiography 14(2): 144-149,           PN and Rogister B: The functional diversity of Aurora kinases: a
     2019. DOI: 10.1016/j.radi.2006.12.001                                        comprehensive review. Cell Div 13: 7, 2018. PMID: 30250494.
5    Otto T and Sicinski P: Cell cycle proteins as promising targets in           DOI: 10.1186/s13008-018-0040-6
     cancer therapy. Nat Rev Cancer 17(2): 93-115, 2017. PMID:               21   Kimura M, Kotani S, Hattori T, Sumi N, Yoshioka T, Todokoro
     28127048. DOI: 10.1038/nrc.2016.138                                          K and Okano Y: Cell cycle-dependent expression and spindle pole
6    Ma HT and Poon RYC: Aurora kinases and DNA damage                            localization of a novel human protein kinase, Aik, related to
     response. Mutat Res 821: 111716, 2020. PMID: 32738522. DOI:                  Aurora of Drosophila and yeast Ipl1. J Biol Chem 272(21):
     10.1016/j.mrfmmm.2020.111716                                                 13766-13771, 1997. PMID: 9153231. DOI: 10.1074/jbc.272.
7    Vader G and Lens SM: The Aurora kinase family in cell division               21.13766
     and cancer. Biochim Biophys Acta 1786(1): 60-72, 2008. PMID:            22   Magnaghi-Jaulin L, Eot-Houllier G, Gallaud E and Giet R: Aurora
     18662747. DOI: 10.1016/j.bbcan.2008.07.003                                   A protein kinase: to the centrosome and beyond. Biomolecules
8    Chen HL, Tang CJ, Chen CY and Tang TK: Overexpression of an                  9(1): 28, 2019. PMID: 30650622. DOI: 10.3390/biom9010028
     Aurora-C kinase-deficient mutant disrupts the Aurora-B/INCENP           23   Kufer TA, Silljé HH, Körner R, Gruss OJ, Meraldi P and Nigg
     complex and induces polyploidy. J Biomed Sci 12(2): 297-310,                 EA: Human TPX2 is required for targeting Aurora-A kinase to the
     2005. PMID: 15917996. DOI: 10.1007/s11373-005-0980-0                         spindle. J Cell Biol 158(4): 617-623, 2002. PMID: 12177045.
9    Carvajal RD, Tse A and Schwartz GK: Aurora kinases: new                      DOI: 10.1083/jcb.200204155
     targets for cancer therapy. Clin Cancer Res 12(23): 6869-6875,          24   Eyers PA, Erikson E, Chen LG and Maller JL: A novel
     2006. PMID: 17145803. DOI: 10.1158/1078-0432.CCR-06-1405                     mechanism for activation of the protein kinase Aurora A. Curr
10   Tang A, Gao K, Chu L, Zhang R, Yang J and Zheng J: Aurora                    Biol 13(8): 691-697, 2003. PMID: 12699628. DOI:
     kinases: novel therapy targets in cancers. Oncotarget 8(14):                 10.1016/s0960-9822(03)00166-0
     23937-23954, 2017. PMID: 28147341. DOI: 10.18632/                       25   Giet R, Uzbekov R, Cubizolles F, Le Guellec K and Prigent C:
     oncotarget.14893                                                             The Xenopus laevis aurora-related protein kinase pEg2 associates
11   Choudary I, Barr PM and Friedberg J: Recent advances in the                  with and phosphorylates the kinesin-related protein XlEg5. J Biol
     development of Aurora kinases inhibitors in hematological                    Chem 274(21): 15005-15013, 1999. PMID: 10329703. DOI:
     malignancies. Ther Adv Hematol 6(6): 282-294, 2015. PMID:                    10.1074/jbc.274.21.15005
     26622997. DOI: 10.1177/2040620715607415                                 26   Kapitein LC, Peterman EJ, Kwok BH, Kim JH, Kapoor TM and
12   Glover DM, Leibowitz MH, McLean DA and Parry H: Mutations                    Schmidt CF: The bipolar mitotic kinesin Eg5 moves on both
     in aurora prevent centrosome separation leading to the formation             microtubules that it crosslinks. Nature 435(7038): 114-118, 2005.
     of monopolar spindles. Cell 81(1): 95-105, 1995. PMID:                       PMID: 15875026. DOI: 10.1038/nature03503
     7720077. DOI: 10.1016/0092-8674(95)90374-7                              27   Tanenbaum ME, Macůrek L, Janssen A, Geers EF, Alvarez-
13   Stenoien DL, Sen S, Mancini MA and Brinkley BR: Dynamic                      Fernández M and Medema RH: Kif15 cooperates with eg5 to
     association of a tumor amplified kinase, Aurora-A, with the                  promote bipolar spindle assembly. Curr Biol 19(20): 1703-
     centrosome and mitotic spindle. Cell Motil Cytoskeleton 55(2):               1711, 2009. PMID: 19818618. DOI: 10.1016/j.cub.
     134-146, 2003. PMID: 12740874. DOI: 10.1002/cm.10120                         2009.08.027
14   Goldenson B and Crispino JD: The aurora kinases in cell cycle           28   van Heesbeen RGHP, Raaijmakers JA, Tanenbaum ME, Halim
     and leukemia. Oncogene 34(5): 537-545, 2015. PMID: 24632603.                 VA, Lelieveld D, Lieftink C, Heck AJR, Egan DA and Medema
     DOI: 10.1038/onc.2014.14                                                     RH: Aurora A, MCAK, and Kif18b promote Eg5-independent
15   Petretti C, Savoian M, Montembault E, Glover DM, Prigent C and               spindle formation. Chromosoma 126(4): 473-486, 2017. PMID:
     Giet R: The PITSLRE/CDK11p58 protein kinase promotes                         27354041. DOI: 10.1007/s00412-016-0607-4

                                                                                                                                              121
CANCER DIAGNOSIS & PROGNOSIS 1: 111-126 (2021)

29 Lens SM, Voest EE and Medema RH: Shared and separate                          Sci 41(2): 160-174, 2016. PMID: 26705896. DOI: 10.1016/
   functions of polo-like kinases and aurora kinases in cancer. Nat              j.tibs.2015.11.002
   Rev Cancer 10(12): 825-841, 2010. PMID: 21102634. DOI:                   43   Kitagawa M, Fung SY, Hameed UF, Goto H, Inagaki M and Lee
   10.1038/nrc2964                                                               SH: Cdk1 coordinates timely activation of MKlp2 kinesin with
30 Carmena M, Wheelock M, Funabiki H and Earnshaw WC: The                        relocation of the chromosome passenger complex for cytokinesis.
   chromosomal passenger complex (CPC): from easy rider to the                   Cell Rep 7(1): 166-179, 2014. PMID: 24656812. DOI:
   godfather of mitosis. Nat Rev Mol Cell Biol 13(12): 789-803,                  10.1016/j.celrep.2014.02.034
   2012. PMID: 23175282. DOI: 10.1038/nrm3474                               44   Mierzwa B and Gerlich DW: Cytokinetic abscission: molecular
31 Hendzel MJ, Wei Y, Mancini MA, Van Hooser A, Ranalli T,                       mechanisms and temporal control. Dev Cell 31(5): 525-538, 2014.
   Brinkley BR, Bazett-Jones DP and Allis CD: Mitosis-specific                   PMID: 25490264. DOI: 10.1016/j.devcel.2014.11.006
   phosphorylation of histone H3 initiates primarily within                 45   Kitagawa M and Lee SH: The chromosomal passenger complex
   pericentromeric heterochromatin during G2 and spreads in an                   (CPC) as a key orchestrator of orderly mitotic exit and
   ordered fashion coincident with mitotic chromosome                            cytokinesis. Front Cell Dev Biol 3: 14, 2015. PMID: 25798441.
   condensation. Chromosoma 106(6): 348-360, 1997. PMID:                         DOI: 10.3389/fcell.2015.00014
   9362543. DOI: 10.1007/s004120050256                                      46   Steigemann P, Wurzenberger C, Schmitz MH, Held M, Guizetti
32 Hans F and Dimitrov S: Histone H3 phosphorylation and cell                    J, Maar S and Gerlich DW: Aurora B-mediated abscission
   division. Oncogene 20(24): 3021-3027, 2001. PMID: 11420717.                   checkpoint protects against tetraploidization. Cell 136(3): 473-
   DOI: 10.1038/sj.onc.1204326                                                   484, 2009. PMID: 19203582. DOI: 10.1016/j.cell.2008.12.020
33 Crosio C, Fimia GM, Loury R, Kimura M, Okano Y, Zhou H, Sen              47   Quartuccio SM and Schindler K: Functions of Aurora kinase C in
   S, Allis CD and Sassone-Corsi P: Mitotic phosphorylation of                   meiosis and cancer. Front Cell Dev Biol 3: 50, 2015. PMID:
   histone H3: spatio-temporal regulation by mammalian Aurora                    26347867. DOI: 10.3389/fcell.2015.00050
   kinases. Mol Cell Biol 22(3): 874-885, 2002. PMID: 11784863.             48   Yang KT, Li SK, Chang CC, Tang CJ, Lin YN, Lee SC and Tang
   DOI: 10.1128/mcb.22.3.874-885.2002                                            TK: Aurora-C kinase deficiency causes cytokinesis failure in
34 Giet R and Glover DM: Drosophila aurora B kinase is required                  meiosis I and production of large polyploid oocytes in mice. Mol
   for histone H3 phosphorylation and condensin recruitment during               Biol Cell 21(14): 2371-2383, 2010. PMID: 20484572. DOI:
   chromosome condensation and to organize the central spindle                   10.1091/mbc.e10-02-0170
   during cytokinesis. J Cell Biol 152(4): 669-682, 2001. PMID:             49   Assou S, Anahory T, Pantesco V, Le Carrour T, Pellestor F, Klein
   11266459. DOI: 10.1083/jcb.152.4.669                                          B, Reyftmann L, Dechaud H, De Vos J and Hamamah S: The
35 Wang F, Dai J, Daum JR, Niedzialkowska E, Banerjee B,                         human cumulus – oocyte complex gene-expression profile. Hum
   Stukenberg PT, Gorbsky GJ and Higgins JM: Histone H3 Thr-3                    Reprod 21(7): 1705-1719, 2006. PMID: 16571642. DOI:
   phosphorylation by Haspin positions Aurora B at centromeres in                10.1093/humrep/del065
   mitosis. Science 330(6001): 231-235, 2010. PMID: 20705812.               50   Sasai K, Katayama H, Hawke DH and Sen S: Aurora-C
   DOI: 10.1126/science.1189435                                                  interactions with survivin and INCENP reveal shared and distinct
36 Kawashima SA, Yamagishi Y, Honda T, Ishiguro K and                            features compared with Aurora-B chromosome passenger protein
   Watanabe Y: Phosphorylation of H2A by Bub1 prevents                           complex. PLoS One 11(6): e0157305, 2016. PMID: 27332895.
   chromosomal instability through localizing shugoshin. Science                 DOI: 10.1371/journal.pone.0157305
   327(5962): 172-177, 2010. PMID: 19965387. DOI: 10.1126/                  51   Fernández-Miranda G, Trakala M, Martín J, Escobar B, González
   science.1180189                                                               A, Ghyselinck NB, Ortega S, Cañamero M, Pérez de Castro I and
37 Yamagishi Y, Honda T, Tanno Y and Watanabe Y: Two histone                     Malumbres M: Genetic disruption of aurora B uncovers an
   marks establish the inner centromere and chromosome bi-                       essential role for aurora C during early mammalian development.
   orientation. Science 330(6001): 239-243, 2010. PMID: 20929775.                Development 138(13): 2661-2672, 2011. PMID: 21613325. DOI:
   DOI: 10.1126/science.1194498                                                  10.1242/dev.066381
38 Broad AJ, DeLuca KF and DeLuca JG: Aurora B kinase is                    52   Balboula AZ and Schindler K: Selective disruption of aurora
   recruited to multiple discrete kinetochore and centromere regions             C kinase reveals distinct functions from aurora B kinase
   in human cells. J Cell Biol 219(3): e201905144, 2020. PMID:                   during meiosis in mouse oocytes. PLoS Genet 10(2):
   32028528. DOI: 10.1083/jcb.201905144                                          e1004194, 2014. PMID: 24586209. DOI: 10.1371/journal.
39 Godek KM, Kabeche L and Compton DA: Regulation of                             pgen.1004194
   kinetochore-microtubule attachments through homeostatic control          53   Cicenas J: The Aurora kinase inhibitors in cancer research and
   during mitosis. Nat Rev Mol Cell Biol 16(1): 57-64, 2015. PMID:               therapy. J Cancer Res Clin Oncol 142(9): 1995-2012, 2016.
   25466864. DOI: 10.1038/nrm3916                                                PMID: 26932147. DOI: 10.1007/s00432-016-2136-1
40 Welburn JP, Vleugel M, Liu D, Yates JR 3rd, Lampson MA,                  54   Yan M, Wang C, He B, Yang M, Tong M, Long Z, Liu B, Peng
   Fukagawa T and Cheeseman IM: Aurora B phosphorylates                          F, Xu L, Zhang Y, Liang D, Lei H, Subrata S, Kelley KW, Lam
   spatially distinct targets to differentially regulate the kinetochore-        EW, Jin B and Liu Q: Aurora-A kinase: a potent oncogene and
   microtubule interface. Mol Cell 38(3): 383-392, 2010. PMID:                   target for cancer therapy. Med Res Rev 36(6): 1036-1079, 2016.
   20471944. DOI: 10.1016/j.molcel.2010.02.034                                   PMID: 27406026. DOI: 10.1002/med.21399
41 London N and Biggins S: Signalling dynamics in the spindle               55   Moreira-Nunes CA, Mesquita FP, Portilho AJS, Mello Júnior
   checkpoint response. Nat Rev Mol Cell Biol 15(11): 736-747,                   FAR, Maués JHDS, Pantoja LDC, Wanderley AV, Khayat AS,
   2014. PMID: 25303117. DOI: 10.1038/nrm3888                                    Zuercher WJ, Montenegro RC, de Moraes-Filho MO and de
42 Trivedi P and Stukenberg PT: A centromere-signaling network                   Moraes MEA: Targeting aurora kinases as a potential prognostic
   underlies the coordination among mitotic events. Trends Biochem               and therapeutical biomarkers in pediatric acute lymphoblastic

122
Machado et al: Aurora Kinases in Hematological Malignancies (Review)

     leukaemia. Sci Rep 10(1): 21272, 2020. PMID: 33277547. DOI:          66 Yamauchi T, Uzui K, Shigemi H, Negoro E, Yoshida A and Ueda
     10.1038/s41598-020-78024-8                                               T: Aurora B inhibitor barasertib and cytarabine exert a greater-
56   Moore AS, Faisal A, Mak GWY, Miraki-Moud F, Bavetsias V,                 than-additive cytotoxicity in acute myeloid leukemia cells. Cancer
     Valenti M, Box G, Hallsworth A, de Haven Brandon A, Xavier               Sci 104(7): 926-933, 2013. PMID: 23557198. DOI: 10.1111/
     CPR, Stronge R, Pearson ADJ, Blagg J, Raynaud FI, Chopra R,              cas.12164
     Eccles SA, Taussig DC and Linardopoulos S: Quizartinib-resistant     67 Fei F, Lim M, Schmidhuber S, Moll J, Groffen J and Heisterkamp
     FLT3-ITD acute myeloid leukemia cells are sensitive to the FLT3-         N: Treatment of human pre-B acute lymphoblastic leukemia with
     Aurora kinase inhibitor CCT241736. Blood Adv 4(7): 1478-1491,            the Aurora kinase inhibitor PHA-739358 (Danusertib). Mol
     2020. PMID: 32282883. DOI: 10.1182/bloodadvances.                        Cancer 11: 42, 2012. PMID: 22721004. DOI: 10.1186/1476-4598-
     2019000986                                                               11-42
57   Wang JX, Zhang L, Huang ZW, Zhang XN, Jiang YY, Liu FJ,              68 Qi W, Liu X, Cooke LS, Persky DO, Miller TP, Squires M and
     Long L, Xue MJ, Lu G, Liu Q and Long ZJ: Aurora kinase                   Mahadevan D: AT9283, a novel aurora kinase inhibitor,
     inhibitor restrains STAT5-activated leukemic cell proliferation by       suppresses tumor growth in aggressive B-cell lymphomas. Int J
     inducing mitochondrial impairment. J Cell Physiol 235(11): 8358-         Cancer 130(12): 2997-3005, 2012. PMID: 21796626. DOI:
     8370, 2020. PMID: 32239704. DOI: 10.1002/jcp.29680                       10.1002/ijc.26324
58   Qi J, Gao X, Zhong X, Zhang N, Wang R, Zhang H, Pan T, Liu           69 Kim SJ, Jang JE, Cheong JW, Eom JI, Jeung HK, Kim Y, Hwang
     X, Yao Y, Wu Q, Niu M and Xu K: Selective inhibition of Aurora           DY and Min YH: Aurora A kinase expression is increased in
     A and B kinases effectively induces cell cycle arrest in t(8;21)         leukemia stem cells, and a selective Aurora A kinase inhibitor
     acute myeloid leukemia. Biomed Pharmacother 117: 109113,                 enhances Ara-C-induced apoptosis in acute myeloid leukemia
     2019. PMID: 31207577. DOI: 10.1016/j.biopha.2019.109113                  stem cells. Korean J Hematol 47(3): 178-185, 2012. PMID:
59   Heo SK, Noh EK, Jeong YK, Ju LJ, Sung JY, Yu HM, Cheon J,                23071472. DOI: 10.5045/kjh.2012.47.3.178
     Koh S, Min YJ, Choi Y and Jo JC: Radotinib inhibits mitosis          70 Chowdhury A, Chowdhury S and Tsai MY: A novel Aurora kinase
     entry in acute myeloid leukemia cells via suppression of Aurora          A inhibitor MK-8745 predicts TPX2 as a therapeutic biomarker
     kinase A expression. Tumour Biol 41(5): 1010428319848612,                in non-Hodgkin lymphoma cell lines. Leuk Lymphoma 53(3):
     2019. PMID: 31074363. DOI: 10.1177/1010428319848612                      462-471, 2012. PMID: 21879811. DOI: 10.3109/104281
60   Payton M, Cheung HK, Ninniri MSS, Marinaccio C, Wayne                    94.2011.619018
     WC, Hanestad K, Crispino JD, Juan G and Coxon A: Dual                71 Yoon Y, Cowley DO, Gallant J, Jones SN, Van Dyke T and
     Targeting of Aurora Kinases with AMG 900 Exhibits Potent                 Rivera-Pérez JA: Conditional Aurora A deficiency differentially
     Preclinical Activity Against Acute Myeloid Leukemia with                 affects early mouse embryo patterning. Dev Biol 371(1): 77-85,
     Distinct Post-Mitotic Outcomes. Mol Cancer Ther 17(12): 2575-            2012. PMID: 22939930. DOI: 10.1016/j.ydbio.2012.08.010
     2585, 2018. PMID: 30266802. DOI: 10.1158/1535-7163.MCT-              72 Hégarat N, Smith E, Nayak G, Takeda S, Eyers PA and
     18-0186                                                                  Hochegger H: Aurora A and Aurora B jointly coordinate
61   Okabe S, Tauchi T, Tanaka Y and Ohyashiki K: Therapeutic                 chromosome segregation and anaphase microtubule dynamics. J
     targeting of Aurora A kinase in Philadelphia chromosome-positive         Cell Biol 195(7): 1103-1113, 2011. PMID: 22184196. DOI:
     ABL tyrosine kinase inhibitor-resistant cells. Oncotarget 9(65):         10.1083/jcb.201105058
     32496-32506, 2018. PMID: 30197758. DOI: 10.18632/                    73 Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka
     oncotarget.25985                                                         P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem
62   Kong L, Jia X, Song Z, Qiu L, Li L, Qian Z, Zhou S, Liu X, Ren           G, Van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S,
     X, Meng B, Fu K, Wang P, Xian-huo wang X and Zhang H: Co-                O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves
     targeting Aurora kinase A and Bcl-2 synergistically inhibits the         MF, Ganser A, Döhner K, Schlenk RF, Döhner H and Campbell
     viability in double-hit lymphoma cells. Translational Cancer             PJ: Genomic classification and prognosis in acute myeloid
     Research 6(4): 746-754, 2018. DOI: 10.21037/tcr.2017.06.41               leukemia. N Engl J Med 374(23): 2209-2221, 2016. PMID:
63   Floc’h N, Ashton S, Taylor P, Trueman D, Harris E, Odedra R,             27276561. DOI: 10.1056/NEJMoa1516192
     Maratea K, Derbyshire N, Caddy J, Jacobs VN, Hattersley M,           74 Sampath D, Rao VA and Plunkett W: Mechanisms of apoptosis
     Wen S, Curtis NJ, Pilling JE, Pease EJ and Barry ST: Optimizing          induction by nucleoside analogs. Oncogene 22(56): 9063-9074,
     therapeutic effect of aurora B inhibition in acute myeloid               2003. PMID: 14663485. DOI: 10.1038/sj.onc.1207229
     leukemia with AZD2811 nanoparticles. Mol Cancer Ther 16(6):          75 Bavetsias V, Crumpler S, Sun C, Avery S, Atrash B, Faisal A,
     1031-1040, 2017. PMID: 28292940. DOI: 10.1158/1535-                      Moore AS, Kosmopoulou M, Brown N, Sheldrake PW, Bush K,
     7163.MCT-16-0580                                                         Henley A, Box G, Valenti M, de Haven Brandon A, Raynaud FI,
64   Long ZJ, Wang LX, Zheng FM, Chen JJ, Luo Y, Tu XX, Lin DJ,               Workman P, Eccles SA, Bayliss R, Linardopoulos S and Blagg J:
     Lu G and Liu Q: A novel compound against oncogenic Aurora                Optimization of imidazo[4,5-b]pyridine-based kinase inhibitors:
     kinase A overcomes imatinib resistance in chronic myeloid                identification of a dual FLT3/Aurora kinase inhibitor as an orally
     leukemia cells. Int J Oncol 46(6): 2488-2496, 2015. PMID:                bioavailable preclinical development candidate for the treatment
     25872528. DOI: 10.3892/ijo.2015.2960                                     of acute myeloid leukemia. J Med Chem 55(20): 8721-8734,
65   Chen YP, Lin HJ, Chen JS, Tsai MY, Hsieh HP, Chang JY, Chen              2012. PMID: 23043539. DOI: 10.1021/jm300952s
     NF, Chang KC, Huang WT, Su WC, Yang ST, Chang WC, Hung               76 Lin S, Ptasinska A, Chen X, Shrestha M, Assi SA, Chin PS,
     LY and Chen TY: CDKN1A-mediated responsiveness of MLL-                   Imperato MR, Aronow BJ, Zhang J, Weirauch MT, Bonifer C and
     AF4-positive acute lymphoblastic leukemia to Aurora kinase-A             Mulloy JC: A FOXO1-induced oncogenic network defines the
     inhibitors. Int J Cancer 135(3): 751-762, 2014. PMID: 24382688.          AML1-ETO preleukemic program. Blood 130(10): 1213-1222,
     DOI: 10.1002/ijc.28708                                                   2017. PMID: 28710059. DOI: 10.1182/blood-2016-11-750976

                                                                                                                                           123
You can also read