Next Article in Journal
An Insight into Biomolecules for the Treatment of Skin Infectious Diseases
Next Article in Special Issue
Behind the Adaptive and Resistance Mechanisms of Cancer Stem Cells to TRAIL
Previous Article in Journal
Clinical Advances of siRNA-Based Nanotherapeutics for Cancer Treatment
Previous Article in Special Issue
Light- and Melanin Nanoparticle-Induced Cytotoxicity in Metastatic Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Second-Generation Antimitotics in Cancer Clinical Trials

1
CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
2
Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
3
ICBAS, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
4
GreenUPorto (Sustainable Agrifood Production) Research Center, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
*
Author to whom correspondence should be addressed.
Pharmaceutics 2021, 13(7), 1011; https://doi.org/10.3390/pharmaceutics13071011
Submission received: 24 May 2021 / Revised: 24 June 2021 / Accepted: 29 June 2021 / Published: 2 July 2021
(This article belongs to the Special Issue Novel Anticancer Strategies (Volume II))

Abstract

:
Mitosis represents a promising target to block cancer cell proliferation. Classical antimitotics, mainly microtubule-targeting agents (MTAs), such as taxanes and vinca alkaloids, are amongst the most successful anticancer drugs. By disrupting microtubules, they activate the spindle assembly checkpoint (SAC), which induces a prolonged delay in mitosis, expected to induce cell death. However, resistance, toxicity, and slippage limit the MTA’s effectiveness. With the desire to overcome some of the MTA’s limitations, mitotic and SAC components have attracted great interest as promising microtubule-independent targets, leading to the so-called second-generation antimitotics (SGAs). The identification of inhibitors against most of these targets, and the promising outcomes achieved in preclinical assays, has sparked the interest of academia and industry. Many of these inhibitors have entered clinical trials; however, they exhibited limited efficacy as monotherapy, and failed to go beyond phase II trials. Combination therapies are emerging as promising strategies to give a second chance to these SGAs. Here, an updated view of the SGAs that reached clinical trials is here provided, together with future research directions, focusing on inhibitors that target the SAC components.

1. Introduction

The cell cycle is a tightly regulated process in which a parental cell gives rise to two genetically identical daughter cells. Cell cycle progression is under the control of the family of serine/threonine kinases cyclin-dependent kinases (Cdk 1, 2, 4, and 6) and their regulatory subunits cyclins (A, B, D, and E). While Cdks’ concentration is constant throughout the cell cycle, their activation depends on the oscillation of cyclin levels at different phases of the cell cycle [1]. The cell cycle is divided into two phases, interphase and mitosis. Interphase is a time of synthesis and growth, occurring according to the consecutive phases G1, S, and G2, during which the DNA is replicated. Mitosis consists of five active phases: prophase, prometaphase, metaphase, anaphase, and telophase, followed by cytokinesis. During prophase, chromosomes start to condense and the centrosomes start to migrate to the opposite sides of the mitotic cell. After nuclear envelope breakdown (NEBD), at the onset of prometaphase, microtubules emanating from centrosomes grow to assemble the mitotic spindle and capture the chromosomes by attaching to their kinetochores. Chromosomes then align at the spindle equator, forming the metaphase plate. When all chromosomes are bipolarly attached to spindle microtubules, sister chromatids are separated and segregate at the anaphase. The nuclear envelope reassembles at the telophase and the cytoplasm divides (cytokinesis), giving rise to two genetically equal daughter cells (Figure 1a,b).
A successful mitosis relies on equal chromosome segregation at the transition from metaphase to anaphase. Kinetochore–microtubule attachment defects lead to the missegregation of chromosomes resulting in genome instability, a hallmark of cancer [2,3]. Fortunately, the fidelity of chromosome segregation is ensured by the spindle assembly checkpoint (SAC) (Figure 2). The SAC consists of a protein network that delays the anaphase in the presence of erroneous kinetochore–microtubule attachments or the absence of attachments [4]. SAC activation is mediated by the orderly orchestrated recruitment to the unattached kinetochores of the SAC proteins monopolar spindle 1 (Mps1), Aurora kinase B, budding uninhibited by benomyl 1 (Bub1), and mitotic arrest deficiency 1 (Mad1). Consequently, the mitotic checkpoint complex (MCC) is assembled, which is formed by Mad2, Bub1-related 1 (BubR1), Bub3 and Cdc20. By sequestering Cdc20, the MCC inhibits the ultimate target of SAC, the anaphase promoting complex/cyclosome (APC/C), an E3 ubiquitin ligase. When all kinetochores are correctly attached to microtubules, the SAC is turned off and APC/C becomes active, targeting securin and cyclin B for proteolysis. The degradation of securin releases the protease separase, which thus cleaves the cohesin rings, allowing sister chromatids to separate, whereas the proteolysis of cyclin B triggers mitotic exit [4].
Based on the uncontrolled proliferation of many cancers, anticancer drugs have been developed to block the cell cycle, particularly mitosis. In this review, we will briefly discuss the current antimitotic approaches, and focus on the new generation of promising antimitotics that have reached clinical trials, with particular emphasis on their clinical efficacy. These so-called second-generation antimitotics (SGAs) target the mitotic kinases and spindle motor proteins. Possible research directions will be discussed.

2. Limitations of Current Microtubule-Targeting Agents

Microtubule-targeting agents (MTAs) are the main current antimitotic drugs in the clinic, and are widely used for the treatment of several cancers [5]. MTAs are divided into two groups, based on their action mechanism: microtubule destabilizers, such as the vinca alkaloids, that inhibit microtubule polymerization; and microtubule stabilizers, such as taxanes, that enhance microtubule polymerization [6]. Both classes impair a functional mitotic spindle, leading to SAC activation and subsequent mitotic arrest, which usually results in cell death by apoptosis [7]. However, other outcomes are possible after MTA treatment. After a prolonged mitotic arrest, cells may exit mitosis without undergoing cytokinesis, originating tetraploid cells, a process known as mitotic slippage, which results from a constant and slow degradation of cyclin B even when SAC is on [8]. The slipped cells can follow three possible fates: become senescent, undergo post-mitotic death, or continue dividing [9]. Therefore, mitotic slippage, together with efflux pumps, mutations in tubulin genes, and deficient apoptotic signaling, represent the main reasons for the therapeutic failure of MTAs [10]. Additionally, MTAs treatment is also frequently associated with neurological and myeloid toxicity [10].

3. Second-Generation Antimitotics in Clinical Trials

Due to the aforementioned limitation of MTAs, alternative approaches to directly targeting microtubules were developed to block cells in mitosis. These new strategies consist of inhibiting the activity of mitotic proteins, especially kinases and motor proteins, that play crucial roles in different processes during mitosis, such as mitotic entry, spindle assembly, chromosome congression, or SAC regulation. The inhibition of these proteins is made possible through small molecules or small interfering RNAs (siRNAs), known as the second-generation antimitotics (SGAs), with promising outcomes in preclinical assays [11]. Here, we will focus on those SGAs that reached clinical trials, namely, inhibitors of Mps1, polo-like kinase 1 (Plk1), Aurora kinases, Eg-5, and centromeric protein E (CENP-E), review their clinical outcomes, and provide future research directions.

3.1. Mps1

Mps1, also known as TKK, is a dual-specificity protein kinase phosphorylating serine/threonine and tyrosine residues [12]. Mps1 is recruited early in mitosis to unattached kinetochores, where it is responsible for SAC activation through the recruitment of several SAC components to kinetochores and subsequent MCC formation [13]. It has also been involved in DNA damage checkpoint response, chromosome alignment, meiosis, cytokinesis, centrosome duplication, and error-correction of kinetochore–microtubule attachment [14,15,16]. Mps1 is overexpressed in various tumors, correlating with poor prognosis [17]. The inhibition of Mps1 activity compromises the SAC, which leads to premature mitotic exit, resulting in massive aberrant chromosome segregation and subsequent cell death [18,19]. Furthermore, inhibition of Mps1 has been shown to sensitize cancer cells to Paclitaxel [18,20]. Hence, Mps1 became an attractive target for cancer therapy, and a variety of small molecules that inhibit Mps1 have been developed. As with most kinases, Mps1 inhibitors are ATP competitive molecules. So far, five Mps1 inhibitors have been approved to begin clinical trials, namely, BOS 172722, BAY 1217389, BAY1161909, CFI-402257 and S 81694. Six phase I/II studies have been undertaken: two have been completed, one has been terminated, two are recruiting participants and another one is ongoing (Figure 1 and Table 1).

3.1.1. BAY 1217389 and BAY1161909

BAY 1217389, an Imidazopyridazine, and BAY1161909, a Triazolopyridine, inhibit Mps1 with an IC50 of 0.34 nM and 0.63 nM, respectively, in vitro [21]. Both compounds were developed by Bayer and showed similar behavior in vivo, with modest efficacy as single agents. However, both compounds demonstrate synergistic effects when combined with Paclitaxel on the growth inhibition of human tumor xenografts in immunocompromised mice [21]. In the clinical trial involving BAY1161909 (NCT02138812) for the treatment of advanced solid malignancies, in a combinational treatment with 75 mg/m2 and 90 mg/m2 of Paclitaxel, five (14%) and four (14%) partial responses (PRs) were reported, respectively [22]. However, Bayer decided to interrupt clinical trials with BAY1161909 in favor of BAY 1217389, which was being developed in parallel. The BAY 1217389 phase I study (NCT02366949) in combination with Paclitaxel was completed in March 2018, but the outcomes are not available yet.

3.1.2. S 81694

S 81694 inhibits Mps1 with an IC50 of 3 nM in vitro, and was first discovered by Nerviano Medical Sciences (NMS-P153), and thereafter acquired and further developed by Servier [23]. S 81694 pre-clinical studies demonstrated that triple-negative breast cancer cell lines were particularly sensitive to S 81694 and, in an MDA-MB-231 xenograft and orthotopic model of tumor regression, the curing of animals and metastasis reduction were observed [24]. A phase I/II trial with S 81694 (NCT03411161) for the treatment of breast cancer was completed in June of 2020, the outcomes of which are not published yet.

3.1.3. BOS 172722

BOS 172722 is a pyrido[3,4-d]pyrimidine, developed by Boston Pharmaceuticals, and was shown to inhibit Mps1 with an IC50 of 11 nM, in vitro [25]. It exhibited modest efficacy as a monotherapy in xenograft studies, but exerted strong synergistic effects in combination with Paclitaxel in triple-negative breast cancer (TNBC) cell lines [26]. A phase I trial (NCT03328494) is currently ongoing with patients with advanced nonhematologic malignancies.

3.1.4. CFI-402257

CFI-402257, a pyrazolo[1,5-a]pyrimidine, is a potent Mps1 inhibitor with an IC50 of 1.2 nM in vitro, and it induced tumor regression in murine colon and lung cancer models [27,28]. Currently, two phase I/II trials are recruiting participants: one for the treatment of advanced solid tumors, including breast cancer, as a monotherapy and in combination with Fulvestrant (NCT02792465), and one in combination with Paclitaxel for breast cancer (NCT03568422).
Based on the promising results of in vivo preclinical studies that have demonstrated a synergistic effect between Mps1 inhibitors and taxanes, all small molecules that entered clinical trials were combined with Paclitaxel. However, it is too early to draw a conclusion on the clinical efficacy and safety of these combinations, as only the outcomes of BAY 1161909 have been published to date. Nevertheless, in this trial, the combinational treatment was generally well tolerated, with the most common AEs being gastrointestinal and hematological. Objective responses were observed with BAY 1161909. The rationale behind these combinations is interesting: silencing the SAC by Mps1 inhibition leads to premature mitotic exit with chromosome missegregation, while the affecting of microtubule dynamics by the taxanes further enhances chromosome misalignment and chromosome missegregation, culminating in massive cell death [20]. Inhibition of Mps1 also demonstrated a synergic effect with cisplatin in malignant mesothelioma cisplatin-resistant cell lines in vitro. Therefore, it will be interesting to evaluate the clinical efficacy of Mps1 inhibitors combined with anti-cancer drugs other than taxanes, in particular with platinum-based agents [29]. It should be noted that all clinical trials enrolled patients with solid tumors. Recently, the inhibition of Mps1 was shown to induce apoptosis in multiple myeloma cell lines, but further research is required to evaluate the potential effectiveness of Mps1 inhibitors in hematological tumors [30].
Table 1. Mps1 inhibitors in clinical trials 1.
Table 1. Mps1 inhibitors in clinical trials 1.
CompoundClinical
Trials
Current
Status
ConditionsInterventionsOutcomes 2Ref.
BAY 1217389
Pharmaceutics 13 01011 i001
NCT02366949Phase I
Completed
Advanced Solid MalignanciesCombination with PaclitaxelNot published-
BAY 1161909 (Empesertib)
Pharmaceutics 13 01011 i002
NCT02138812Phase I
Terminated
Advanced Solid MalignanciesCombination with PaclitaxelBest response was PR[22]
S 81694
Structure Undisclosed
NCT03411161Phase I
Phase II
Completed
Metastatic Breast Cancer,
Metastatic Triple-Negative Breast Cancer
Combination with PaclitaxelNot published-
BOS 172722
Pharmaceutics 13 01011 i003
NCT03328494Phase I
Ongoing
Advanced Nonhematologic MalignanciesCombination with Paclitaxel--
CFI-402257
Pharmaceutics 13 01011 i004
NCT03568422Phase I
Phase II
Recruiting
Breast CancerCombination with Paclitaxel--
NCT02792465Phase I,
Recruiting
Breast Cancer,
Advanced Solid Malignancies
Monotherapy
Combination with Fulvestrant
--
1 Data collected from clinicaltrials.gov. 2 PR, partial response.

3.2. Plk1

Plk1 is a cell cycle-regulating serine/threonine kinase implicated in centrosome maturation and separation, mitotic entry, spindle assembly, kinetochore–microtubule attachment, the SAC, DNA damage checkpoint activation, and cytokinesis [31,32,33]. Knockdown or inhibition through small molecules of Plk1 leads to monopolar spindle formation, G2/M arrest, and polyploidy, which ultimately leads to cell death [34,35,36]. Plk1 is overexpressed in several tumors, associated with poor prognosis, and has been associated with resistance to chemotherapeutics such as Doxorubicin, Gemcitabine, and Paclitaxel [37,38]. Taken together, these facts led to the development of several small molecules that target Plk1 for cancer therapy. Nine Plk1 inhibitors have already entered clinical trials: CYC 140, GSK461364, TAK-960, NMS-1286937 (Onvansertib), BI 6727 (Volasertib), BI 2536, Rigosertib, MK-1496, and lipid nanoparticles carrying the siRNA TKM-080301 (Figure 1 and Table 2). Seventy-nine phase I/II clinical trials with solid and hematological tumor patients have been initiated: fifty-two completed, five ongoing, fifteen terminated/withdrawn, and seven are recruiting participants. Additionally, three phase III studies with metastatic pancreatic adenocarcinoma and myelodysplastic syndrome (MDS) patients have been completed, and two studies with acute myeloid leukemia (AML) and MDS patients are ongoing.

3.2.1. BI 2536 and BI6727 (Volasertib)

BI 2536 and Volasertib are two similar molecules developed by Boehringer Ingelheim. BI2536 is a potent ATP-competitive Plk1 inhibitor with an IC50 of 0.83 nM in vitro [39]. Several studies have been undertaken with this compound. Overall, the most common AEs were neutropenia and leukopenia. PR were reported in non-small-cell lung cancer patients as monotherapy (2.1%, NCT00376623) and in combination with pemetrexed (5.2%, NCT02211833), and in pancreatic cancer patients (2.3%, NCT00710710) as a monotherapy [40,41,42]. Nevertheless, the greatest efficacy was observed in AML patients (NCT00701766), in which two complete remissions (CRs, 3.7%) and three PRs (5.5%) were reported, and in non-Hodgkin’s lymphoma patients (NCT00243087), also as monotherapy, in which three CR (17.7%) and one PR (5.9%) were achieved [43,44]. However, clinical trials with BI 2536 have been terminated as single agent, and the second-generation inhibitor Volasertib has been chosen for further clinical development.
Volasertib is a dihydropteridinone derivative that inhibits the Plk1 with an IC50 of 0.84 nM in vitro [45]. The first-in-human trial of Volasertib was initiated in 2005 against solid tumors as monotherapy (NCT02273388) [46]. The most common AEs were predominately hematological, and the main dose-limiting toxicities (DLT) were thrombocytopenia, neutropenia, and febrile neutropenia. The MTD was established as 400 mg. However, due to overall tolerability, 300 mg was set as the recommended dose for further development [46]. Three PRs (4.6%) were reported in patients with melanoma, ovarian and urothelial cancer, and 40% of patients had the best response of SD [46]. Since then, at least twenty-four more phase I/II trials (twelve completed, six withdrawn, four terminated and two ongoing) have begun against solid and hematological tumors. Several PRs have been reported with Volasertib as a single agent or in combination with other drugs against many solid tumors. In two phase I trials with Volasertib as the single agent against solid tumors, three PRs (3.3%) were reported in patients with melanoma, ureteral cancer (NCT00969553), and gastric cancer (6.7%, NCT01348347) [47,48]. Based on this phase I result of Volasertib as a monotherapy, two phase II trials were conducted in patients with urothelial cancer (NCT01023958) and platinum-resistant/refractory ovarian cancer (NCT01121406). In the study with urothelial cancer patients, seven PRs (14%) were reported and 26% of patients had SD. The safety profile was considered acceptable, but Volasertib demonstrated insufficient antitumor activity for further evaluation as a monotherapy in these patients [49]. Additionally, in the trial with platinum-resistant/refractory ovarian cancer patients, seven PRs (13%) were reported and 44.4% of patients had SD. The expression of Plk1 was evaluated in 47.3% of patients, but the results demonstrated no relationship between Plk1 levels and Volasertib response. In this regard, it was suggested that further clinical development of Volasertib as a single agent in these patients should only be performed after biomarker analysis, in order to select patients with higher chances of response [50]. Another phase II trial was conducted in patients with non-small-cell lung cancer treated with Volasertib as a single agent, in combination with pemetrexed, or pemetrexed alone (NCT00824408). The combinational regimen did not increase toxicity compared to pemetrexed as a single agent. Three PRs (8.1%) were reported with Volasertib as a single agent, and ten (21.3%) in the combinational arm; however, the combination treatment did not demonstrate an efficacy improvement [51]. More objective responses were reported in other studies with solid tumor patients treated with Volasertib in combination with several drugs. In a phase I trial in combination with Nintedanib (NCT01022853), one CR (3.3%) in a breast cancer patient and one PR (3.3%) in a patient with non-small-cell lung cancer were reported [52]. In another phase I study in combination with Afatinib (NCT01206816), two PRs (3.3%) were observed in patients with non-small-cell lung cancer and head and neck cancer of the tongue [53]. Additionally, in a phase I trial, Volasertib was combined with platinum agents (Cisplatin or Carboplatin, NCT00969761); two PRs (6.7%) were reported with Volasertib plus cisplatin in patients with follicular dendritic reticulum cell carcinoma of the palatine tonsil and follicular dendritic reticulum cell retroperitoneal sarcoma, and two additional PRs (6.5%) were also reported with Volasertib plus Carboplatin in patients with non-small-cell lung cancer and hypopharynx carcinoma [54]. Still, the best response was achieved in patients with AML, in which three CRs (15.8%) and another three CRs with incomplete blood count remission (15.8%, CRi) were reported as monotherapy (NCT01662505) [55]. In this trial, the MTD was established as 450 mg, administered intravenously on days 1 and 15 in a 28-day cycle, and grade 3 or higher AEs were mainly hematological (neutropenia and thrombocytopenia). However, the safety profile was considered acceptable and could be managed through the supportive care of granulocyte colony-stimulating factor (G-CSF) [55]. These results prompted the use of Volasertib in a phase II (NCT00804856) and a phase III (NCT01721876) trial against AML, both in combination with cytarabine, which are currently ongoing.

3.2.2. ON 01910.Na (Rigosertib)

Rigosertib inhibits Plk1 with an IC50 of 9 nM in vitro [56]. At least thirty-two phase I/II trials have begun (twenty-four completed, one terminated, two withdrawn, three planned/recruiting participants and two ongoing) as monotherapy and in combinational treatments against solid and hematological tumors. Three PRs (9.3%) were reported in Hodgkin lymphoma, thymic cancer and pancreatic ductal adenocarcinoma patients in combination with Gemcitabine (NCT01125891) [57]. These led Rigosertib to a phase III trial against pancreatic adenocarcinoma in combination with Gemcitabine (NCT01360853), which was completed in 2015. In this study the best response was PR in 19% of patients, and SD in 50% of patients; however, the combinational treatment did not demonstrate an improvement in survival compared to Gemcitabine as a single agent [58]. Nevertheless, the phase I/II outcomes in patients with myelodysplastic syndrome (MDS) were the most promising, and Rigosertib was approved for three phase III trials as monotherapy. One is ongoing (NCT02562443), and two have been completed (NCT01241500 and NCT01928537), in which the best overall response was marrow complete remission (mCR) in 20% and 22% of patients, respectively [59,60]. Additionally, in a phase I/II trial in combination with Azacitidine in patients with MDS (NCT01926587), two CRs (12.5%) were reported and the phase II trial is ongoing [61]. Currently, two phase I/II trials with Rigosertib as monotherapy are planned, recruiting participants against recessive dystrophic epidermolysis bullosa (NCT03786237 and NCT04177498), and also a phase I study in patients with non-small-cell lung cancer with Rigosertib in combination with nivolumab is recruiting participants (NCT04263090).

3.2.3. GSK461364

GSK461364 is a thiophene amide that inhibits Plk1 with an IC50 of 7 nM in vitro, and it was developed by GlaxoSmithKline [62]. GSK461364 entered a phase I trial for the treatment of advanced solid malignancies as monotherapy (NCT00536835). No objective responses were reported, and only six stable diseases (SD, 15%) were observed in patients with esophageal, endometrial carcinoma, and ovarian cancers, all treated with doses at or above the MTD [63]. The most common AEs were infusion site reactions and phlebitis, and due to the high incidence of venous thromboembolism (20%) no further development is planned.

3.2.4. MK-1496

MK-1496, developed by Merck Sharp & Dohme Corp., has completed a phase I trial as monotherapy for the treatment of advanced solid tumors in 2009 (NCT00880568). The MTD was determined as 80 mg/m2, and reversible hematotoxicity was the main side effect [64]. Two PRs (11.8%) were reported in patients with parotid gland carcinoma and small-cell lung cancer, but no further development is planned for MK-1496.

3.2.5. TAK-960

TAK-960, developed by Millennium Pharmaceuticals, inhibits Plk1 with an IC50 of 2 nM in vitro [65]. TAK-960 has demonstrated substantial antitumor activity in various human cancer models [66]. A phase I trial was started in 2010 with TAK-960 as monotherapy for the treatment of advanced nonhematologic malignancies (NCT01179399), but has been terminated early due to lack of efficacy, and further development has been halted.

3.2.6. NMS-1286937 (Onvansertib)

Onvansertib, from Nerviano Medical Sciences, is another potent Plk1 inhibitor with an IC50 of 2 nM in vitro [67]. In a phase I trial as monotherapy, the best response was SD (26%) in patients with colorectal cancer, pancreatic carcinoma with a K-RAS mutation, head and neck squamous cell carcinoma, and basal cell carcinoma (NCT01014429) [68]. Despite the lack of efficacy as a monotherapy, three phase I/II studies are currently recruiting participants: one for AML in combination with decitabine or cytarabine (NCT03303339), one for metastatic prostate cancer in combination with FOLFIRI (folinic acid, fluorouracil, and irinotecan) and bevacizumab (NCT03829410), and one for metastatic colorectal cancer with a KRAS mutation in combination with abiraterone and prednisone (NCT03414034).

3.2.7. TKM-080301

TKM-080301 is distinctive among the anti-Plk1 since it is a lipid nanoparticle formulation of an siRNA targeting the Plk1 gene transcript. Phase I/II studies on TKM-080301 as monotherapy in patients with advanced hepatocellular carcinomas (HCC, NCT02191878), adrenocortical cancer (ACC, NCT01262235) and other solid tumors (NCT01437007) were completed. TKM-080301 was generally well tolerated, but did not demonstrate clinical antitumor activity against HCC. However, it showed a better response against ACC with a PR (12.5%) reported, and further clinical evaluation is warranted [69,70].

3.2.8. CYC 140

CYC 140, from Cyclacel Pharmaceuticals, inhibits Plk1 with an IC50 of 3 nM in vitro, and has demonstrated antitumor activity in human tumor xenografts at non-toxic doses [71]. CYC 140 is the most recent Plk1 inhibitor in clinical trials, and a phase I study using it against hematological malignancies as monotherapy is currently recruiting participants (NCT03884829).
The AEs associated with Plk1 inhibitors were mainly hematological, namely, neutropenia and thrombocytopenia, but the safety profile of most molecules was considered acceptable and manageable. The efficacy outcomes in patients with solid tumors were modest, either as monotherapy or in combinational treatments. Inhibition of Plk1 has been suggested to be more effective in tumors with high levels of Plk1 and mutated p53, thus selection of patients with these tumor characteristics may improve the clinical outcomes of Plk1 inhibitors against solid tumors [74,75]. Interestingly, better responses were achieved in patients with hematological tumors, especially against AML and MDS, with Volasertib and Rigosertib, respectively, representing the most promising molecules among Plk1 inhibitors. Both inhibitors have demonstrated antitumor activity as a monotherapy, which was improved when combined with other chemotherapeutic drugs. Therefore, future efforts should focus on combinational treatments in order to increase efficacy while reducing drug dosage, DLTs, and development of resistance. Additionally, further investigation of biomarkers that predict small molecule efficiency towards inhibiting Plk1 is needed.
Table 2. Plk1 inhibitors in clinical trials 1.
Table 2. Plk1 inhibitors in clinical trials 1.
CompoundClinical
Trials
Current
Status
ConditionsInterventionsOutcomes 2Refs.
BI 2536
Pharmaceutics 13 01011 i005
11 Clinical trialsPhase I/II
10 Completed
1 Terminated
Advanced Solid Tumors, Acute Myeloid Leukemia, Non-Hodgkin’s LymphomaMonotherapyBest responses were CR for acute myeloid leukemia and non-Hodgkin’s lymphoma; PR for non-small-cell lung cancer, pancreatic cancer, acute myeloid leukemia, and non-Hodgkin’s lymphoma[40,41,42,43,44]
Combination with Pemetrexed and GemcitabineBest response was PR for adenocarcinoma and squamous cell carcinoma
BI 6727 (Volasertib)
Pharmaceutics 13 01011 i006
26 Clinical trials25 Phase I/II
12 Completed
4 Terminated
6 Withdrawn
3 Ongoing
Acute Myeloid Leukemia Pediatric Patients with Advanced Cancers, Myelodysplastic Syndromes, Non-Hodgkin’s Lymphoma, Urothelial, Ovarian, Lung Pancreatic, Colorectal and Prostate CancerMonotherapyBest responses were CR for acute myeloid leukemia; PR for non-small cell lung cancer, melanoma, ovarian cancer, acute myeloid leukemia, gastric cancer, and urothelial cancer[46,47,48,49,50,51,52,53,54,55]
Combination with Cytarabine, Pemetrexed, Azacitidine, Afatinib, Decitabine, Daunorubicin, Nintedanib, Mitoxantrone and ItraconazoleBest responses were CR for breast cancer in combination with Nintedanib, PR for non-small-lung cancer in combination with Pemetrexed, Nintedanib, Afatinib, and Carboplatin, PR for head and neck carcinoma in combination with Afatinib, PR for undifferentiated follicular dendritic reticulum cell sarcoma and differentiated follicular dendritic reticulum cell retroperitoneal sarcoma in combination with Cisplatin, PR for differentiated hypopharynx carcinoma in combination with Carboplatin
1 Phase III OngoingAcute Myeloid LeukemiaCombination with Cytarabine-
ON 01910.Na (Rigosertib)
Pharmaceutics 13 01011 i007
36 Clinical trials32 Phase I/II
24 Completed
1 Terminated
2 Withdrawn
3 Recruiting/ Planned
2 Ongoing
Refractory Leukemia, Myelodysplastic Syndrome, Squamous Cell Carcinoma Acute/Chronic Myeloid Leukemia, Ovarian and Lung Cancer, Advanced Solid TumorsMonotherapyBest response was PR for non-Hodgkin’s lymphoma, thymic cancer, and pancreatic ductal adenocarcinoma[57,58,59,60,61,72,73]
Combination with Nivolumab, Cisplatin, Azacitidine, Oxaplatin, Gemcitabine and IrinotecanBest responses were CR and mCR for myelodysplastic syndrome in combination with Azacitidine
4 Phase III
3 Completed
1 Ongoing
Metastatic Pancreatic Adenocarcinoma, Myelodysplastic SyndromesMonotherapyBest responses were SD and mCR for myelodysplastic syndromes
Combination with GemcitabineBest response was PR for pancreatic adenocarcinoma
GSK461364
Pharmaceutics 13 01011 i008
NCT00536835Phase I
Completed
Non-Hodgkin Lymphoma,
Advanced Solid Malignancies
MonotherapyBest response was SD for esophageal and ovarian cancers and endometrial carcinoma[63]
MK-1496
Pharmaceutics 13 01011 i009
NCT00880568Phase I
Completed
Advanced Solid TumorsMonotherapyBest response was PR for parotid gland carcinoma and small cell lung cancer[64]
TAK-960
Pharmaceutics 13 01011 i010
NCT01179399Phase I
Terminated
Advanced Nonhematologic MalignanciesMonotherapyDiscontinued strategically by sponsor due to lack of efficacy-
NMS-1286937/Onvansertib
Pharmaceutics 13 01011 i011
4 Clinical trialsPhase I/II
1 Completed
3 Recruiting
Advanced or Metastatic Solid TumorsMonotherapyBest response was SD for colorectal cancer, pancreatic carcinoma with a K-RAS mutation, head and neck squamous cell carcinoma, and basal cell carcinoma[68]
Acute Myeloid Leukemia, Metastatic Prostate Cancer, Metastatic Colorectal Cancer with a KRAS mutationCombination with Decitabine, Cytarabine, Abiraterone, Prednisone, FOLFIRI and Bevacizumab-
TKM-080301
(siRNA)
3 Clinical trialsPhase I/II
Completed
Hepatocellular Carcinoma, Colorectal, Pancreas, Breast and Ovarian
Cancer with Hepatic Metastases, Adrenocortical Carcinoma, Neuroendocrine Tumors
MonotherapyBest response was PR for adrenocortical carcinoma[69,70]
CYC 140
Pharmaceutics 13 01011 i012
NCT03884829Phase I
Recruiting
Acute Myeloid Leukemia,
Myelodysplastic Syndromes,
Acute Lymphoblastic Leukemia
Monotherapy--
1 Data collected from clinicaltrials.gov. 2 CR, complete remission; mCR, marrow complete remission; PR, partial response; SD, stable disease.

4. Aurora Kinases

Aurora kinases A, B, and C (AurA, AurB, and AurC) are a family of serine/threonine kinases that play critical functions during mitosis. AurA is implicated in centrosome maturation and separation, cytokinesis, and bipolar spindle assembly, whereas AurB is involved in chromosome condensation and alignment, kinetochore–microtubule attachments, SAC activation, and cytokinesis [76,77]. All Aurora family members are overexpressed in several tumors, and have been related to improving the survival and proliferation of tumor cells [77]. The inhibition of AurA and AurB results in cell death through different mechanisms. AurB inhibition leads to defects in kinetochore–microtubule attachments that override the SAC, resulting in extensive aneuploidy and cell death [78,79]. In contrast, inhibition of AurA provokes a shortened and disorganized microtubule spindle, inducing a transient SAC activation and subsequent mitotic arrest, followed by mitotic exit and apoptosis [80,81]. For these reasons, Aurora kinases were seen as attractive targets for cancer therapy. Therefore, several small molecules with broad-spectrum inhibition activity against Aurora kinases have been developed in pre-clinical studies, known as pan-Aurora inhibitors. Twelve compounds have reached clinical trials: VX-680 (Tozasertib), CYC 116, PHA739358 (Danusertib), SNS-314 Mesylate, PF-03814735, AS703569 (Cenisertib), TAK-901, ABT-348 (Ilorasertib), AMG-900, GSK1070916 (NIM-900), AT9283, and BI-847325 (Figure 1 and Table 3).

4.1. Pan-Aurora Inhibitors

4.1.1. ABT-348 (Ilorasertib)

Ilorasertib was reported to inhibit AurA with an IC50 of 120 nM, AurB with an IC50 of 7 nM, and AurC with an IC50 of 1nM, in vitro, and is also able to inhibit the VEGF [82]. At least four phase I/II trials have been initiated with Ilorasertib against solid and hematological tumors. A phase I trial was conducted with patients with solid tumors in which Ilorasertib was administered as single agent, or in combination with Docetaxel or Carboplatin (NCT01110486). However, only the outcomes as monotherapy were published. In total, 10% of patients experienced a total of ten DLTs, and the study was terminated by strategic decision of the sponsor without establishment of MTD. Nevertheless, two PRs (2.4%) were reported in patients with basal cell carcinoma and adenocarcinoma [83]. In a phase I trial in patients with hematological tumors, Ilorasertib was administered in combination with Azacitidine (NCT01110473). The DLTs observed were pancreatitis, acute kidney injury, and hypertension. Among the fifty-two patients, twelve had SD as the best response, and three objective responses (CR, CRi, and PR) were reported in patients with AML [84]. Currently, a phase II trial is ongoing with Ilorasertib as a single agent (NCT02478320).

4.1.2. AS703569/MSC1992371A (Cenisertib)

Cenisertib displays a broad inhibitory activity against a number of kinases, including AurA and B [85]. A phase I trial using it as monotherapy was conducted in patients with solid tumors (NCT00391521). Neutropenia, febrile neutropenia, and thrombocytopenia were the most common DLTs observed, and no objective responses were observed [86]. In another phase I trial with patients with solid tumors, Cenisertib was administered in combination with Gemcitabine (NCT01097512). The MTD was 37 mg/m2 of Cenisertib with the standard 1000 mg/m2 of Gemcitabine, and neutropenia was the main DLT reported. The best response observed was two PRs (3%) in patients with non-small-cell lung cancer and hepatocellular carcinoma [87]. A phase I trial was also conducted with patients with hematological malignancies with Cenisertib as single agent in two schedules (NCT01080664). In the first schedule, Cenisertib was given on days 1–3 and 8–10, and in the second schedule on days 1–6, both in a 21-day cycle. The MTDs were established as 37 mg/m2 and 28 mg/m2 for the first and second schedules, respectively. The DLTs reported were severe neutropenia with infection and sepsis, mucositis, and diarrhea. Overall, two CRs (2.7%) were observed in patients with AML, and a CRi (1.3%) was reported in a patient with acute lymphoid leukemia with Philadelphia chromosome [88].

4.1.3. VX-680 (Tozasertib)

Tozasertib, also known as VX-680 or MK-0457, was developed by Merck Sharp & Dohme Corp., and was shown to inhibit AurA with an IC50 of 0.6 nM, AurB with an IC50 of 18 nM, and AurC with an IC50 of 4.6 nM, in vitro [89]. Tozasertib has entered five trials as monotherapy against solid and hematological tumors, and in combination with Dasatinib in patients with chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia (NCT00500006). In the first-in-human study in patients with solid tumors (NCT02532868), the MTD was identified as 64 mg/m2, and Tozasertib was generally well tolerated, with neutropenia and a herpes zoster reactivation being the DLTs observed. No objective responses were reported, and twelve patients (44.4%) had SD as best response [90]. The best response to Tozasertib treatment was observed in a trial with patients with leukemia (NCT00111683), in which a CR (1.3%) was reported in a patient with chronic myeloid leukemia [91].

4.1.4. BI-847325

BI-847325, from Boehringer Ingelheim, is a potent inhibitor of all Aurora family members as well as MEK1/2 kinases [92]. A phase I study with BI-847325 as the single agent in patients with solid tumors was initiated in 2011 (NCT01324830). The DLTs observed were primarily hematological (neutropenia, febrile neutropenia, and thrombocytopenia) and gastrointestinal (vomiting and diarrhea). In total, 45% of patients had SD as the best response, and one PR (1.4%) was reported in a patient with squamous cell carcinoma of the esophagus [93].

4.1.5. AT9283

AT9283, from Astex Pharmaceuticals, inhibits AurA and AurB with an IC50 of 3 nM in vitro. Additionally, AT9283 was also found to inhibit other kinases including JAK2, Flt3, and Abl (T315I) [94]. At least five trials have been completed with AT9283 as monotherapy. A phase I trial in patients with solid tumors or non-Hodgkin’s lymphoma was started in 2007 (NCT00443976). The MTD was determined to be 47 mg/m2/day, and febrile neutropenia and wound infection were the DLTs observed. Four patients (12.5%) achieved SD as best response and one PR (3.1%) was reported in a patient with squamous cell carcinoma of the anal canal [95]. In a phase II trial with patients with leukemia or myelofibrosis (NCT00522990), the MTD was established as 324 mg/m2/72h, and tolerability was strongly dose-dependent. No CR or PR were observed [96]. A phase I trial was conducted in children and adolescents with solid tumors (NCT00985868). The MTD was 18.5 mg/m2/day, and the most common DLTs observed were neutropenia and febrile neutropenia. The best response reported was a PR (4.3%) in a patient with central nervous system-primitive neuroectodermal tumor [97].

4.1.6. AMG-900

AMG-900, developed by Amgen, inhibits Aurora A, B, and C in vitro, with IC50 values of 5 nM, 4 nM, and 1 nM, respectively [98]. The first-in-human trial of AMG-900 was conducted in patients with solid tumors (NCT00858377). The MTD was 25 mg/day, and neutropenia was the most common DLT observed. Consequently, G-CSF support was included in treatment regimen, and the MTD was established as a higher dose of 40 mg/day. In total, 56% of the patients had SD and one PR (2.4%) was reported in a patient with clear-cell endometrial cancer [99]. In a phase I trial against AML (NCT01380756) with AMG-900 as monotherapy, the most common AEs were nausea, diarrhea, and febrile neutropenia. Three patients had a best response of CRi (9%) and no other responses were observed [100].

4.1.7. PHA739358 (Danusertib)

Danusertib is a pyrrolo-pyrazole developed by Nerviano Medical Sciences, and inhibits AurA with an IC50 of 13 nM, AurB with an IC50 of 79 nM, and AurC with an IC50 of 61 nM, in vitro [101]. A phase II trial in patients with multiple myeloma was terminated earlier due to low recruitment rate (NCT00872300). Another phase II trial with Danusertib as single agent against metastatic hormone-refractory prostate cancer was completed (NCT00766324). Danusertib was generally well tolerated with neutropenia and fatigue being the most common AEs observed. No objective responses were observed, and 21 (25.9%) patients achieved SD as the best response [102].

4.1.8. SNS-314 Mesylate

SNS-314 Mesylate, from Sunesis Pharmaceuticals, inhibits AurA with an IC50 of 9 nM, AurB with an IC50 of 31 nM, and AurC with an IC50 of 3 nM, in vitro [103]. SNS-314 Mesylate entered a phase I trial (NCT00519662) in patients with solid tumors as a single agent. It was generally well tolerated, with nausea, fatigue, vomiting, constipation, and pain being the most AEs commonly observed. In total, 18.8% of patients had SD as the best response, and no objective responses were reported [104].

4.1.9. TAK-901

TAK-901 was developed by Millennium Pharmaceuticals (Takeda) and inhibits AurA and B in vitro, with IC50 values of 21 nM and 15 nM, respectively [105]. TAK-901 entered two phase I trials for solid and hematological malignancies (NCT00935844 and NCT00807677). Both trials were completed but no data are available yet.

4.1.10. CYC116

CYC116, from Cyclacel Pharmaceuticals, inhibits AurA with a Ki of 8 nM and AurB with a Ki of 9.2 nM, and showed antitumor activity in vivo [106]. CYC116 entered a phase I trial in patients with solid tumors as monotherapy (NCT00560716); however, the study was terminated early by sponsor decision.

4.1.11. GSK1070916 (NIM-900)

GSK1070916, also known as NIM-900, is a potent Aurora B and C inhibitor, with IC50 values of 5 nM and 6.5 nM, respectively, in vitro [107]. A phase I trial in patients with advanced solid tumors was completed in 2013 with GSK1070916 as monotherapy (NCT01118611), but the outcomes are not published yet.

4.1.12. PF-03814735

PF-03814735, developed by Pfizer, is a potent Aurora A and B inhibitor with IC50 values of 0.8 nM and 5 nM, respectively, in vitro [108]. PF-03814735 entered a phase I trial against solid tumors as a single agent (NCT00424632). The DLTs observed were neutropenia, febrile neutropenia, increase in aspartate amino transferase, and left ventricular dysfunction. No objective responses were reported, with 35.5% of patients achieving SD as the best response [109].
Table 3. Pan-Aurora inhibitors in clinical trials 1.
Table 3. Pan-Aurora inhibitors in clinical trials 1.
CompoundClinical
Trials
Current
Status
ConditionsInterventionsOutcomes 2Refs.
ABT-348 (Ilorasertib)
Pharmaceutics 13 01011 i013
4 Clinical trialsPhase I/II
3 Completed
1 Ongoing
Solid Tumors, Advanced Hematological MalignanciesMonotherapyBest response was PR for basal cell carcinoma, and adenocarcinoma[83,84]
Combination with Carboplatin, Docetaxel and AzacitidineBest responses were CR, PR, and CRi for acute myeloid leukemia in combination with Azacitidine
AS703569/MSC1992371A (Cenisertib)
Pharmaceutics 13 01011 i014
3 Clinical trialsPhase I/II
2 Completed
1 Terminated
Solid Tumors, Advanced MalignanciesMonotherapyBest response was CR for acute myeloid leukemia and CRi for acute lymphoid leukemia with Philadelphia chromosome[86,87,88]
Combination with GemcitabineBest response was PR for non-small-cell lung cancer and hepatocellular carcinoma
VX-680/MK-0457 (Tozasertib)
Pharmaceutics 13 01011 i015
6 Clinical trialsPhase I/II
1 Completed
5 Terminated
Chronic Myeloid Leukemia, Philadelphia Chromosome-Positive, Acute Lymphoblastic Leukemia, Non-Small-Cell Lung Carcinoma, Advanced Solid TumorsMonotherapy
Combination with Dasatinib
Best response was one CR for chronic myeloid leukemia with BCR–ABL T315I mutation phenotype as monotherapy[90,91,110]
BI-847325
Pharmaceutics 13 01011 i016
NCT01324830Phase I
Completed
Solid TumorsMonotherapyBest response was PR for esophageal cancer[93]
AT9283
Pharmaceutics 13 01011 i017
5 Clinical trialsPhase I/II
4 Completed
1 Terminated
Multiple Myeloma, Acute Leukemia, Myelofibrosis, Advanced or metastatic solid tumors, Non-Hodgkin’s lymphoma, Solid TumorsMonotherapyBest response was PR for nervous system primitive neuroectodermal tumor and squamous cell carcinoma[95,96,97]
AMG-900
Pharmaceutics 13 01011 i018
NCT00858377Phase I
Completed
Advanced Solid TumorsMonotherapyBest response was PR for clear-cell endometrial cancer[99]
NCT01380756Phase I
Completed
Acute Myeloid LeukemiaMonotherapyBest response was CRi[100]
PHA739358, (Danusertib)
Pharmaceutics 13 01011 i019
NCT00766324Phase II
Completed
Metastatic Hormone Refractory Prostate CancerMonotherapyThe best response was SD[102]
NCT00872300Phase II
Terminated
Multiple Myeloma--
SNS-314 Mesylate
Pharmaceutics 13 01011 i020
NCT00519662Phase I
Completed
Advanced Solid TumorsMonotherapyBest response was SD[104]
TAK-901
Pharmaceutics 13 01011 i021
NCT00935844Phase I
Completed
Advanced Solid Tumors
Lymphoma
MonotherapyNot published-
NCT00807677Phase I
Completed
Advanced Hematologic Malignancies
CYC116
Pharmaceutics 13 01011 i022
NCT00560716Phase I
Terminated
Advanced Solid TumorsMonotherapy--
GSK1070916/NMI-900
Pharmaceutics 13 01011 i023
NCT01118611Phase I
Completed
Advanced Solid TumorsMonotherapyNot published-
PF-03814735
Pharmaceutics 13 01011 i024
NCT00424632Phase I
Completed
Advanced Solid TumorsMonotherapyBest response was SD for non-small-cell lung cancer, melanoma, renal cell carcinoma, and neuroendocrine tumor[109]
1 Data collected from clinicaltrials.gov. 2 CR, complete remission; Cri, complete remission with incomplete blood count remission; PR, partial response; SD, stable disease.

4.2. Aurora B inhibitors

In addition to inhibitors with a broad-spectrum for Aurora kinases, several small molecules have been developed with selectivity to each Aurora family member. Four AurB-specific inhibitors (Chiauranib, AZD1152 or Barasertib, BI-811283, and BI-831266) have entered clinical trials for the treatment of solid and hematological tumors (Figure 1 and Table 4).

4.2.1. AZD1152 (Barasertib)

Barasertib, from AstraZeneca, is a potent AurB inhibitor with an IC50 of 0.37 nM in vitro [111]. Barasertib entered at least ten clinical trials for the treatment of solid and hematological tumors. A phase I trial as single agent was conducted in patients with solid tumors (NCT00338182). Fatigue, neutropenia, and nausea were the most common AEs, and neutropenia was the main DLT observed. No objective responses were reported, and 26.5% of patients had SD as the best response [112]. In another phase I trial with patients with solid tumors (NCT00497731), the outcomes were similar to the previous study, in which neutropenia was the main DLT observed and the best response reported was SD in 25.5% of patients [113]. Despite the lack of efficacy in solid tumors, Barasertib demonstrated better antitumor activity in patients with AML. In a phase I/II trial (NCT00497991) using it as a single agent, the MTD was established as 1200 mg, with febrile neutropenia and stomatitis/mucosal inflammation being the most common AEs observed. Sixteen (25%) objective responses were observed, including three CRs, six CRi, and seven PRs [114]. Furthermore, additional responses (CR, CRi, or PR) were reported in other trials (NCT00530699, NCT00952588, and NCT01019161) with Barasertib as a single agent in patients with AML [115,116,117]. Another phase I trial was conducted in AML patients in combination with low doses of Cytarabine (NCT00926731). The MTD was established as 1000 mg of Barasertib and 400 mg of Cytarabine, with the most common AEs being febrile neutropenia, nausea, diarrhea, peripheral edema, and stomatitis. Ten patients (45%) had a response to treatment, including six CR, two CRi, and two PRs [118]. A phase I/II trial with AML patients is currently recruiting participants with Barasertib as a single agent, or in combination with Venetoclax and Azacitidine (NCT03217838). A phase II trial was also conducted with patients with diffuse large B-cell lymphoma (NCT01354392) with Barasertib as a single agent. Patients received up to six cycles of 800 mg of Barasertib starting on day 1 of the 21-day cycle, and G-CSF support was added to the regimen if grade 3 or higher neutropenia occurred. The most common AEs were neutropenia, nausea, diarrhea, anemia, fatigue, and mucositis. In total, 33% of patients had SD and three PRs (20%) were reported [119].

4.2.2. BI-831266

BI-831266 inhibits AurB with an IC50 of 42 nM in vitro and has demonstrated antitumor activity in murine xenograft tumor models [120]. A phase I trial as monotherapy was conducted with patients with solid tumors (NCT00756223). The most common AEs were fatigue, neutropenia, and alopecia, with febrile neutropenia being the only DLT observed. In total, 16% of patients had SD, and one PR (4%) was reported in a patient with cervical cancer [121].

4.2.3. BI-811283

BI-811283 is another small molecule developed by Boehringer Ingelheim that inhibits AurB with an IC50 of 9 nM in vitro [122]. A phase I study with patients with solid tumors was completed with BI-811283 as a single agent (NCT00701324). The DLTs observed were mainly hematological, including neutropenia and thrombocytopenia. No objective responses were reported, and 37% of patients had SD as the best response [123]. Better results were observed in a phase II trial with BI-811283 in combination with a low dose of cytarabine in AML patients (NCT00632749). MTD was established as 100 mg of BI-811283, with anemia, nausea, pyrexia, and febrile neutropenia being the most common AEs observed. In total, 14% of patients showed treatment responses, including seven CR, one CRi, and one PR [124].

4.2.4. Chiauranib

Chiauranib, developed by Chipscreen Biosciences, is a potent AurB inhibitor with an IC50 of 9 nM in vitro. Chiauranib also exerts an inhibitory activity against VEGF receptors and the colony-stimulating receptor 1 (CSF-1R), and is the most recent AurB inhibitor tested in trials [125]. A phase I trial monotherapy was conducted with patients with solid tumors (NCT02122809). Chiauranib was generally well tolerated, with fatigue, proteinuria, hematuria, hypothyroidism, hypertriglyceridemia, and hypertension being the most common AEs observed. No objective responses were observed, with 66.7% of patients achieving SD as best response [126]. Currently, a phase II trial is ongoing with ovarian cancer patients (NCT03901118), and another three phase I/II studies are planned, recruiting participants with hepatocellular carcinoma (NCT03245190), non-Hodgkin’s lymphoma (NCT03974243), and small-cell lung cancer (NCT03216343).
Table 4. Aurora B inhibitors in clinical trials 1.
Table 4. Aurora B inhibitors in clinical trials 1.
CompoundClinical
Trials
Current
Status
ConditionsInterventionsOutcomes 2Refs.
AZD1152 (Barasertib)
Pharmaceutics 13 01011 i025
10 Clinical trialsPhase I/II
8 Completed
2 Terminated
Lymphoma, Advanced Solid Tumors, Acute Myeloid LeukemiaMonotherapyBest response was CR for acute myeloid leukemia, and PR for lymphoma and acute myeloid leukemia[112,113,114,115,116,117,118,119]
Combination with low dose of Cytarabine, Venetoclax, AzacitidineBest response was CR and PR for acute myeloid leukemia in combination with low dose of Cytarabine
BI-831266
Pharmaceutics 13 01011 i026
NCT00756223Phase I
Completed
Advanced Solid TumorsMonotherapyBest response was PR for cervical cancer[121]
BI-811283
(Structure Undisclosed)
NCT00701324Phase I
Completed
Solid TumorsMonotherapyBest response was SD[123]
NCT00632749Phase II
Completed
Acute Myeloid LeukemiaCombination with CytarabineBest responses were CR, PR, and CRi[124]
Chiauranib
Pharmaceutics 13 01011 i027
7 Clinical trialsPhase I/II
2 Completed
1 Terminated
3 Recruiting/ Planned
1 Ongoing
Ovarian Cancer, Non-Hodgkin’s Lymphoma, Hepatocellular Carcinoma, Small Cell Lung Cancer, Other Advanced Solid TumorsMonotherapyBest response was SD[126]
1 Data collected from clinicaltrials.gov. 2 CR, complete remission; Cri, complete remission with incomplete blood count remission; PR, partial response; SD, stable disease.

4.3. Aurora A inhibitors

Inhibitors with selectivity to AurA were also developed. In fact, more AurA inhibitors have reached clinical trials than AurB inhibitors, including MLN8237 (Alisertib), LY3295668 (Erbumine), TAS-119, ENMD-2076, MK-5108/VX-689, KW-2449, and MLN8054. At least seventy-three trials involving these compounds have been initiated for the treatment of solid and hematological tumors (Figure 1 and Table 5).

4.3.1. MLN8237 (Alisertib)

Alisertib is a potent oral AurA inhibitor with an IC50 of 1.2 nM in vitro [127]. Alisertib has been extensively tested in clinical trials, with at least fifty-four trials initiated as monotherapy, and in several combinational treatments against solid and hematological tumors.
The first-in-human trial was conducted in patients with solid tumors with Alisertib as a single agent (NCT00500903). Patients were administered orally with Alisertib for 7, 14, or 21 consecutive days, followed by a 14-day recovery period. MTD was established as 50 mg twice a day for 7 consecutive days, and the main DLTs observed were neutropenia and thrombocytopenia. Alisertib was generally well tolerated, with 38% of patients achieving SD as the best response, and one PR (1.1%) was reported in a patient with platinum- and radiation-refractory ovarian cancer lasting for more than one year [128]. These results prompted the undertaking of more trials with Alisertib as a single agent in patients with solid tumors. Objective responses were reported. In a phase II trial in patients with ovarian, fallopian tube, or peritoneal carcinoma (NCT00853307), two (6%) PRs were reported in patients with ovarian carcinoma [129]. In two phase II trials in patients with metastatic sarcoma (NCT01653028) and metastatic castrate-resistant and neuroendocrine prostate cancer (NCT01799278), two PRs in each study (2.7% and 3.3%, respectively) were reported [130,131]. Additionally, several studies tested the combination of Alisertib with taxane drugs (Paclitaxel and Docetaxel) in patients with solid tumors. A phase I/II trial was conducted in patients with breast or ovarian carcinoma, who received Alisertib in combination with Paclitaxel or Paclitaxel alone (NCT01091428). The MTD was established as 10 mg of Alisertib twice a day plus 80 mg/m2 of Paclitaxel. Febrile neutropenia, neutropenia, stomatitis, and diarrhea were the DLTs observed, although this combination has demonstrated a manageable safety profile. Seven CRs (11%) and 23 PRs (37%) were reported in the combinational treatment schedule, representing a 10% improvement in the ORR, in comparison to Paclitaxel as single agent (30% vs. 20%) [132]. More objective responses with Alisertib in combination with Paclitaxel were also reported in other solid tumors. For instance, in a phase II trial for the treatment of small-cell lung cancer (NCT02038647), nineteen PRs (21%) and one CR were reported (1.1%), and in a phase II with urothelial cancer patients (NCT02109328), two PRs (9.1%) were observed [133,134]. The efficacy outcomes of a phase I trial which combined Alisertib with Docetaxel in solid tumor patients (NCT01094288) were similar to those for the combination with Paclitaxel, in which seven PRs (25%) were reported in patients with angiosarcoma and castration-resistant prostate cancer, and a CR (3,6%) was observed in a patient with bladder cancer [135]. Furthermore, additional PRs in patients with solid tumors were reported with Alisertib in combination with other drugs, such as Pazopanib against breast cancer and mesothelioma (7%, NCT01639911), Irinotecan and Temozolomide against neuroblastoma (12.5%, NCT01601535), Oxaliplatin, Leucovorin, and Fluorouracil against colorectal cancer (8.3%, NCT02319018), and Fulvestrant against lobular ER+/PR+/HER2-breast cancer (22.2%, NCT02219789) [136,137,138,139]. Currently, several phase I/II trials are ongoing, namely, with lung cancer and mesothelioma patients treated with Alisertib as single agent (NCT02293005), with solid tumor patients in combination with Gemcitabine (NCT01924260), with breast cancer patients in combination with Paclitaxel (NCT02187991), and with solid tumor and breast cancer patients in combination with MLN0128 (NCT02719691). Additionally, other trials are recruiting participants for the treatment of head and neck squamous cell carcinoma and malignant solid neoplasm with Alisertib in combination with Pembrolizumab (NCT04555837), and non-small-cell lung cancer (NCT04479306) and EGFR-mutant lung cancer (NCT04085315) with Alisertib in combination with Pembrolizumab.
The clinical trial results of Alisertib against hematological tumors were also very promising. A phase I monotherapy study was conducted (NCT00697346). The recommended phase II dose was 50 mg twice a day for 7 days, followed by a recovery period of 14 days, in a 21-day cycle. The DLTs observed were mainly hematological, including neutropenia, febrile neutropenia, and thrombocytopenia, with 9% of patients who had their treatment discontinued due to AEs. However, Alisertib was generally well tolerated at the recommended phase II dose. In total, 27.6% of patients had SD as the best response, and six PRs (12.8%) were reported in patients with follicular lymphoma, multiple myeloma, peripheral T-cell lymphoma, and diffuse large B-cell lymphoma [140]. These results encouraged the use of Alisertib in a phase II study in patients with T-cell lymphoma as monotherapy (NCT01466881). Patients received the recommended phase II dose established in the previous study. The most common grade 3 or higher AEs were neutropenia, anemia, and thrombocytopenia. In total, 18.9% of patients had SD, and the best responses observed were two CRs (5.4%) and seven PRs (18.9%) [141]. Based on the efficacy results observed in this study, a phase III trial (NCT01482962) was conducted in patients with peripheral T-cell lymphoma, in which Alisertib efficacy as a single agent was evaluated and compared to a comparator (Pralatrexate, Gemcitabine, or Romidepsin). Despite the ORR with Alisertib being 33% (18 CRs and 16 PRs), it was not statistically significantly superior to the comparator arm [142]. However, objective responses were reported in other hematological tumors too. In a phase II trial with non-Hodgkin lymphoma patients treated with Alisertib as a single agent (NCT00807495), 10% of patients had CR and 17% had PR [143]. In a phase I/II trial conducted in patients with non-Hodgkin lymphomas (NCT01397825), treated with Alisertib in combination with Rituximab or with Rituximab plus Vincristine, 38% ORR was observed, including seven CRs and seven PRs [144]. Additionally, six PRs (23%) were observed in a phase I/II trial with multiple myeloma patients (NCT01034553) treated with Alisertib in combination with Bortezomib [145]. In a phase II trial in patients with AML (NCT02560025), treated with Alisertib combined with induction chemotherapy (Daunorubicin or Idarubicin plus Cytarabine), 51% of patients had CR and five CRi (13%) were also reported [146]. Currently, a phase I trial is ongoing in patients with non-Hodgkin lymphomas, with Alisertib in combination with Bortezomib and Rituximab (NCT01695941).
Alisertib was also evaluated in children with solid and hematological tumors. A phase I trial was conducted in pediatric patients with solid tumors, with Alisertib as single agent (NCT02444884). Neutropenia was the most common DLT observed, and the MTD was established as 80 mg/m2 once daily for 7 days followed by 2 weeks rest in a 21-day cycle. In total, 18% of patients had SD and one PR (3%) was reported in a patient with hepatoblastoma [147]. A phase II trial combined Alisertib with Paclitaxel in patients with solid tumors or leukemia (NCT01154816). The most common AEs were neutropenia, anemia, leukopenia, and thrombocytopenia. One CR (0.7%) and two PRs (1.5%) were observed in patients with neuroblastoma and another patient with Wilms tumor also had a CR [148]. Currently, a phase II trial using Alisertib as a single agent against rhabdoid tumor is recruiting participants (NCT02114229).

4.3.2. ENMD-2076

ENMD-2076 is an orally administered AurA inhibitor with an IC50 of 14 nM, and is also capable of inhibiting multiple tyrosine kinases in vitro [149]. ENMD-2076 entered eight phase I/II trials for the treatment of solid and hematological tumors, all as monotherapy. The first-in-human trial was conducted in patients with solid tumors (NCT00658671). MTD was established as 160 mg/m2, and the DLTs observed were neutropenia and hypertension. In total, 85% of patients achieved SD as the best response, and two PRs (3%) were reported in patients with platinum-refractory/resistant ovarian cancer [150]. These results prompted the conduction of two phase II studies with ENMD-2076 in patients with ovarian cancer. Overall, ENMD-2076 was well tolerated, with hypertension and diarrhea being the most common AEs in both studies. In the first trial (NCT01104675), with patients with platinum-refractory/resistant ovarian cancer, the majority of patients (52%) had progressive diseases, although five PRs (8%) were reported [151], whereas in the other study (NCT01914510), with patients with clear-cell ovarian cancer, 55% of patients achieved SD and two PRs (7.9%) were observed [152]. Additional objective responses were also observed in other tumors. A phase II trial was conducted in patients with soft tissue sarcoma (NCT01719744), in which 35% of patients had SD and two PRs (9%) were reported in patients with angiosarcoma and undifferentiated pleomorphic sarcoma [153]. In a phase II trial with triple-negative breast cancer patients (NCT01639248), 38.9% of patients had SD and two PRs (5.2%) were reported [154]. Furthermore, in a phase II trial with fibrolamellar carcinoma patients (NCT02234986), one PR (3%) was reported and 57% of patients had SD [155]. Additionally, ENMD-2076 entered two trials against hematological tumors. A phase I trial with patients with AML or chronic myelomonocytic leukemia was completed in 2011 (NCT00904787). MTD was established as 225 mg once a day, with typhilis, fatigue, syncope, and QTc prolongation being the DLTs observed. The best response was observed in three patients who had CRi (19%) [156].

4.3.3. LY3295668 (Erbumine)

LY3295668 is a potent AurA inhibitor with an IC50 of 1.12 nM in vitro, and it has demonstrated antitumor activity in xenograft cancer models [80]. LY3295668 is the most recently tested inhibitor in clinical trials, with two trials completed. The first-in-human trial with LY3295668 as monotherapy was conducted in patients with solid tumors (NCT03092934). MTD was 25 mg twice a day, with diarrhea, corneal deposits, and mucositis being the DLTs observed. The best response was SD achieved in 69% of patients [157]. The other phase I/II study, with metastatic breast cancer patients, was completed in 2020; the results are yet to be announced (NCT03955939). Currently, a phase I trial is ongoing for the treatment of small-cell lung cancer (NCT03898791) with LY3295668 as monotherapy, and another phase I study with LY3295668 in combination with Topotecan and Cyclophosphamide in patients with neuroblastoma is recruiting participants (NCT04106219).

4.3.4. MLN8054

MLN8054, from Millennium Pharmaceuticals, inhibits AurA with an IC50 of 4 nM in vitro [158]. The first-in-human trial was conducted with patients with solid tumors (NCT00249301), in which MLN8054 was given orally for 7, 14, or 21 days, followed by a 14-day recovery period. The MTD was 60 mg, with the most common DLT observed being somnolence. No objective responses were observed, and 15% of patients had SD as the best response [159]. In another phase I trial with patients with solid tumors (NCT00652158), the efficacy outcomes were similar, in which the best response observed was SD [160].

4.3.5. MK-5108 (VX-689)

MK-5108, from Merck Sharp & Dohme, is a highly potent AurA inhibitor with an IC50 of 0.064 nM in vitro [161]. A phase I trial was conducted in patients with solid tumors, in which MK-5108 was given as single agent and in combination with Docetaxel (NCT00543387). Its MTD as a single agent was not established because no patients experienced a DLT, whereas the MTD of the combinational treatment was 300 mg/day. Overall, the drug-related AEs were mainly blood and lymphatic system disorders. No objective responses were reported with MK-5108 as a single agent. However, a PR (5.9%) was observed in a patient who received 450mg/day and a standard dose of Docetaxel [162].

4.3.6. TAS-119

TAS-119, developed by Taiho Oncology, is a potent AurA inhibitor with an IC50 of 1 nM in vitro [163]. The first-in-human phase I study with TAS-119 as monotherapy was conducted in patients with solid tumors (NCT02448589). The MTD was established as 250 mg twice a day, with fatigue, pain, and diarrhea being the most common AEs observed. No objective responses were observed, and 35% of patients had SD as the best response [164]. In a phase I trial for the treatment of solid tumors, TAS-119 was given in combination with Paclitaxel (NCT02134067). The MTD was established as 80 mg/m2 of Paclitaxel with 75 mg of TAS-119 twice a day. Neutropenia and elevated AST were the DLTs observed. In total, 45% of patients had SD, and four PRs (15.4%) were reported in patients with ovarian/fallopian tube cancers [165].

4.3.7. KW-2449

KW-2449, developed by Kyowa Kirin Pharmaceutical Development, is a multikinase inhibitor, active against AurA with an IC50 of 48 nM in vitro [166]. KW-2449 entered two phase I trials with patients with hematological tumors. However, both were terminated due to a suboptimal dosing schedule and failure to identify a tolerable dose that had potential efficacy (NCT00346632 and NCT00779480).
Table 5. Aurora A kinase-specific inhibitors in clinical trials 1.
Table 5. Aurora A kinase-specific inhibitors in clinical trials 1.
CompoundClinical
Trials
Current
Status
ConditionsInterventionsOutcomes 2Refs.
MLN8237 (Alisertib)
Pharmaceutics 13 01011 i028
53 Clinical trialsPhase I/II
38 completed
7 terminated
4 ongoing
4 recruiting
Advanced Hematological Malignancies, Advanced Solid Tumors, Lymphoma, Prostate Cancer, Ovarian, Fallopian Tube and Peritoneal Carcinoma, Leiomyosarcoma, Neuroblastoma, Acute Myeloid Leukemia, Mantle Cell Lymphoma, Burkitt’s Lymphoma, Breast Carcinoma, Glioma, Non-Hodgkin’s Lymphoma, Bladder Cancer, Non-Small-Cell Lung Cancer,
Mesothelioma,
Small-Cell Lung Cancer
Adenocarcinoma,
Melanoma,
Head and Neck Squamous Cell Carcinoma,
EGFR-mutant Lung Cancer,
Rhabdoid Tumor
MonotherapyBest responses were CR for neuroblastoma, Wilms tumor, non-Hodgkin’s lymphoma, and acute myeloid leukemia;
PR for lymphoma, multiple myeloma, angiosarcoma, ovarian cancer, prostate cancer, hepatoblastoma, neuroblastoma, non-Hodgkin’s lymphoma, and acute myeloid leukemia
[128,129,130,131,132,133,134,135,136,137,138, 139,140,141,143,144,144,145,146,147,148]
Combination with Pazopanib, Bortezomib, Docetaxel, Irinotecan, Temozolomide, Daunorubicin, Idarubicin, Cytarabine, Rituximab, Vincristine, Paclitaxel, Oxaliplatin, Leucovorin, Fluorouracil, Esomeprazole, Rifampin, Fulvestrant, Itraconazole, Idarubicin, Cytarabine, Vorinostat, MLN0128, Abiraterone Acetate, Prednisone, Erlotinib, Gemcitabine, Pembrolizumab, Osimertinib, Romidepsin, BortezomibBest responses were CR for multiple myeloma in combination with Bortezomib, ovarian cancer in combination with Paclitaxel acute myeloid leukemia in combination with Daunorubicin, Idarubicin, and Cytarabine, refractory aggressive B-cell lymphoma in combination with Rituximab and Vincristine, breast cancer in combination with Paclitaxel, small-cell lung cancer in combination with Paclitaxel;
PR for urothelial cancer in combination with Paclitaxel, breast cancer in combination with Pazopanib, Fulvestrant and Paclitaxel, mesothelioma in combination with Pazopanib, multiple myeloma in combination with Bortezomib, angiosarcoma in combination with Docetaxel, castration-resistant prostate cancer in combination with Docetaxel, neuroblastoma in combination with Temozolomide, acute myeloid leukemia in combination with Daunorubicin, Idarubicin, and Cytarabine, refractory aggressive B-cell lymphoma in combination with Rituximab and Vincristine, ovarian cancer in combination with Paclitaxel, colon cancer in combination with Oxaliplatin, Leucovorin, and Fluorouracil, small-cell lung cancer in combination with Paclitaxel
MLN8237 (Alisertib)
Pharmaceutics 13 01011 i029
NCT01482962Phase III
Completed
Relapsed/Refractory
Peripheral T-Cell Lymphoma
MonotherapyBest responses were CR and PR[142]
ENMD-2076
Pharmaceutics 13 01011 i030
8 Clinical trialsPhase I/II
Completed
Soft Tissue Sarcoma, Ovarian Cancer,
Triple-Negative Breast Cancer,
Relapsed or Refractory Hematological Malignancies,
Advanced Fibrolamellar Carcinoma,
Multiple Myeloma,
Advanced Malignancies
MonotherapyBest response was PR for ovarian cancer, triple-negative breast cancer, advanced fibrolamellar carcinoma, undifferentiated pleomorphic sarcoma, and angiosarcoma; and CRi for relapsed or refractory hematological malignancies[150,151,152,153,154,155,156,167]
LY3295668 (Erbumine)
Pharmaceutics 13 01011 i031
4 Clinical trialsPhase I/II
2 Completed
1 Recruiting
1 Planned
Small-Cell Lung Cancer, Metastatic Breast Cancer, Neuroblastoma, Solid TumorsMonotherapy
Combination with Topotecan and Cyclophosphamide
Best response was SD[157]
MLN8054
Pharmaceutics 13 01011 i032
NCT00249301Phase I
Terminated
Solid TumorsMonotherapyBest response was SD[159]
NCT00652158Phase I
Terminated
Advanced MalignanciesMonotherapy[160]
MK-5108/VX-689
Pharmaceutics 13 01011 i033
NCT00543387Phase I
Completed
Solid TumorsMonotherapyBest response was SD[162]
Combination with DocetaxelBest response was PR
TAS-119
(Structure Undisclosed)
NCT02448589Phase I
Terminated
Advanced Solid TumorsMonotherapy Best response was SD [164]
NCT02134067Phase I
Terminated
Combination with PaclitaxelBest response was PR for ovarian/fallopian tube cancers[165]
KW-2449
Pharmaceutics 13 01011 i034
NCT00346632Phase I
Terminated
Acute Leukemias
Myelodysplastic Syndromes—Chronic Myelogenous Leukemia
MonotherapyTerminated due to suboptimal dosing schedule-
NCT00779480Phase I
Terminated
Acute Myelogenous LeukemiaMonotherapyFailure to demonstrate a tolerable dose that had potential for efficacy
1 Data collected from clinicaltrials.gov. 2 CR, complete remission; Cri, complete remission with incomplete blood count remission; PR, partial response; SD, stable disease.
In summary, considering all Aurora inhibitors, either those with broad-spectrum or with selective activity, the AEs observed were mainly hematological, primarily neutropenia, which was manageable through G-CSF support. As to pan-Aurora inhibitors, the majority of small molecules were used as monotherapy and demonstrated a poor or modest efficacy in solid and hematological tumors, except for the clinical trials with Cenisertib and Tozasertib, in which CRs were reported in patients with leukemia. CRs in leukemia patients were also observed with Ilorasertib in combination with Azacitidine. Similar efficacy results were observed with the AurB-selective inhibitors. The best responses were achieved in patients with AML with Barasertib as monotherapy and in combination with Cytarabine, while in patients with solid tumors, all inhibitors showed modest efficacy. AurA inhibitors have demonstrated better responses in patients with solid tumors comparatively to AurB and pan-Aurora inhibitors, especially with Alisertib. Even so, the efficacy outcomes of AurA inhibitors were better in hematological tumors than in solid tumors. There is a substantial debate as to whether it is more efficient to inhibit AurA, AurB, or both simultaneously. A pre-clinical study in pancreatic cancer cells has pointed to AurA as a better target than AurB [168]. However, another pre-clinical study has demonstrated that colon cancer cells were more sensitive to AurB inhibition compared to AurA [78]. In fact, the molecule with better efficacy outcomes in trials was the AurA inhibitor Alisertib, but both strategies have demonstrated antitumor activity, especially against hematological tumors. Perhaps, some tumors may be more sensitive to inhibition of one of the two kinases, but further studies are required to address this question. An important aspect of Aurora kinase inhibition is the existence of biomarkers that permit access to its cellular activity, such as histone H3 phosphorylation or autophosphorylation on T288 of AurA, enabling one to verify whether the inhibitors are efficiently targeting the kinases [169]. In sum, either AurA or AurB inhibition seem to be sustainable approaches for cancer therapy that could be improved in combination with other drugs.

5. CENP-E Kinesin

Centromere-associated protein E (CENP-E) is a plus end-directed motor protein that plays a crucial role in cytokinesis, chromosome congression and alignment, and in SAC signaling through modulation of BubR1 function [170,171,172]. Inhibition of CENP-E results in mitotic arrest due to unaligned chromosomes, which activates the SAC and has demonstrated antitumor activity in human cancer models [173,174,175]. Some CENP-E inhibitors have been tested in pre-clinical studies, but only one small molecule, GSK923295, has reached clinical trials (Figure 1 and Table 6). GSK923295 is an allosteric inhibitor of CENP-E with an IC50 of 1.6 nM in vitro [176]. In the phase I trial (NCT00504790), the MTD was established as 190 mg/m2 [177]. GSK923295 was generally well tolerated; the most common AEs were fatigue, diarrhea, and decreased appetite. Five patients (12.8%) experienced DLTs such as increase in aspartate aminotransferase (AST), fatigue, hypoxia, and hypokalemia [177]. Antitumor activity was modest, and the best response was one PR (3%) in a patient with urothelial carcinoma treated with a dose above the MTD (250 mg/m2), and 33% had SD [177]. More studies are needed to overcome the challenges of using the CENP-E inhibition approach, in order to develop novel CENP-E inhibitors and to test combinational treatments with other drugs for possible synergistic effects.
Table 6. CENP-E inhibitors in clinical trials 1.
Table 6. CENP-E inhibitors in clinical trials 1.
CompoundClinical TrialsCurrent StatusConditionsInterventionsOutcomes 2Ref.
GSK923295
Pharmaceutics 13 01011 i035
NCT00504790Phase I
Completed
Refractory CancerMonotherapyBest response was PR for urothelial carcinoma[177]
1 Data collected from clinicaltrials.gov. 2 PR, partial response.

6. Eg-5 Kinesin

Eg-5 kinesin is a plus end-directed motor protein that plays a critical role in bipolar spindle assembly [178,179]. The inhibition of Eg-5 results in monopolar spindles, which leads to SAC activation and mitotic arrest, and has demonstrated antitumor activity in human xenografts models [180,181,182]. Together with its overexpression in several tumors, this makes Eg-5 an attractive target for cancer therapy [183,184,185,186]. Conversely to the kinesin CENP-E, ten Eg-5 inhibitors have reached clinical trials: 4SC-205, ARRY-520 (Filanesib), AZD4877, MK-0731, SB-715992 (Ispinesib), LY2523355 (Litronesib), SB-743921, EMD 534085, ARQ 621, and ALN-VSP02 (Figure 1 and Table 7). Forty-five phase I/II trials against solid and hematological tumors, in monotherapy or in combinational treatments, have been completed or terminated.

6.1. SB-715992 (Ispinesib) and SB-743921

Ispinesib, developed by Cytokinetics and GlaxoSmithKline, was the first Eg-5 inhibitor to enter clinical trials. Thirteen studies have been completed or terminated as monotherapy against solid and hematological tumors, and three phase I/II trials in combination with Docetaxel (NCT00169520), Capecitabine (NCT00119171), and Carboplatin (NCT00136578) against solid tumors have also been completed [187,188,189]. In the phase I study in combination with Docetaxel, the MTD was established as 10 mg/m2 of Ispinesib and 60 mg/m2 of Docetaxel; prolonged neutropenia and febrile neutropenia were the DLTs observed, although the safety profile was considered acceptable and manageable [187]. Similar results were obtained with Carboplatin and Capecitabine, in which the best response was SD, and the DLTs observed were thrombocytopenia and neutropenia, respectively [188,189]. The efficacy outcomes with Ispinesib as monotherapy in patients with liver cancer (NCT00095992), metastatic prostate cancer (NCT00096499), recurrent or metastatic squamous cell carcinoma of the head and neck (NCT00095628), melanoma (NCT00095953), and metastatic kidney cancer (NCT00354250) were also disappointing, with SD being the best response [190,191,192,193,194]. Better results, although modest, were reported from trials against ovarian cancer (NCT00097409) and breast cancer (NCT00607841), in which PRs were observed in 5% and 6.7% of patients, respectively [195,196]. Additionally, a phase I trial on pediatric patients with relapsed or refractory solid tumors has also been initiated with Ispinesib as monotherapy (NCT00363272). Similarly to the previous studies, Ispinesib was well tolerated, but no objective responses were observed, and only three SDs (12.5%) were reported [197].
Meanwhile, another small molecule derived from Ispinesib, SB-743921, was discovered, which exhibited a five-fold increase in potency against Eg-5 compared to Ispinesib, and reached two phase I/II trials, both already completed [198]. In the first-in-human trial (NCT00136513) in patients with solid tumors as monotherapy, the MTD was established as 4 mg/m2, and neutropenia was the most common DLT registered. Six patients (15%) showed SD as the best response and a PR was reported in a patient (2.3%) with cholangiocarcinoma [199]. In another phase I/II trial with non-Hodgkin lymphoma and Hodgkin lymphoma patients (NCT00343564), it was reported that DLT and MTD were significantly increased when SB-743921 was co-administered with G-CSF [200]. In this study, four PRs (7.1%) were reported: in three patients with Hodgkin lymphoma, and in one patient with non-Hodgkin lymphoma [200].

6.2. ARRY-250 (Filanesib)

Filanesib is a potent Eg-5 inhibitor with an IC50 of 6 nM in vitro [201]. Eight phase I/II studies have been completed in patients with solid and hematological tumors. In the phase I trial (NCT00462358) with patients with solid tumors as monotherapy or with G-CSF support (Filgrastim), the MTD was defined as 1.25 mg/m2 without prophylactic Filgrastim, and as 1.60 mg/m2 in combination with Filgrastim [202]. Filanesib was observed to cause myelosuppression, with the most common treatment-related AEs being febrile neutropenia, neutropenia, leukopenia, and thrombocytopenia [202]. No objective responses were reported and 18% of patients achieved SD as best response [202]. In another monotherapy phase I/II trial in AML patients (NCT00637052), hematological toxicities were the most common AEs. SD was observed in 10% of patients and one PR (3%) was reported [203]. The most promising outcomes were reported in patients with multiple myeloma. The first phase I/II trial with patients with multiple myeloma started in 2009 (NCT00821249) to assess the safety profile and efficacy of Filanesib. Based on the myelosuppression reported in previous studies, G-CSF (Filgrastim) was added to the treatment regimen during phase I and II [204]. In phase II, patients were treated with Filanesib as monotherapy (including G-CSF support), or in combination with low doses of Dexamethasone. As a single agent, 39% of patients had SD and five PRs (16%) were reported, whereas in combination with Dexamethasone, 41% of patients achieved SD and eight PRs (15%) were observed [204]. These results prompted the use of Filanesib in more trials in patients with multiple myeloma in combination with the proteasome inhibitors Bortezomib and Carfilzomib. In a phase I trial (NCT01248923), Filanesib was administrated in combination with Bortezomib and Dexamethasone (including G-CSF support). The safety profile was considered favorable and the hematological toxicities were manageable through G-CSF support [205]. The overall response rate (ORR) was 20%, including one CR (2%) and ten PRs (18%) [205]. In another phase I trial (NCT01372540), Filanesib was administrated in combination with Carfilzomib and Dexamethasone, and G-CSF support was also included. Overall, no CR was reported, although twenty-three PRs (37%) were observed [206]. Additionally, in another phase I/II trial (NCT02384083), Filanesib was combined with Pomalidomide and Dexamethasone, and G-CSF support was also included. Despite the G-CSF support, more than 60% of the patients developed grade 3/4 neutropenia; nevertheless, the efficacy results were promising with an ORR of 51%, including two CRs (4%) and twenty-one PRs (47%) [207]. Considering these good results, Filanesib will likely enter a phase III trial against multiple myeloma.

6.3. ALN-VSP02

ALN-VSP02 is unique among the anti-Eg-5 as it is an siRNA lipid nanoparticle formulation targeting the expression of Eg-5 and vascular endothelial growth factor (VEGF) [208]. Two phase I trials were completed with patients with solid tumors as monotherapy (NCT01158079 and NCT00882180). Patients were enrolled sequentially on one of seven dose levels, ranging from 0.1 to 1.5 mg/kg. ALN-VSP02 demonstrated a safety profile at multiple doses, with fatigue, asthenia, nausea, vomiting, and fever being the most common AEs. The best response was a CR (2.7%) observed in a patient with endometrial cancer with multiple hepatic metastases [209].

6.4. Litronesib

Litronesib, developed by Kyowa Kirin and Eli Lilly and Company, was demonstrated to inhibit Eg-5 with an IC50 of 26 nM in vitro [210]. At least seven phase I/II trials were completed, involving solid tumor patients treated with Litronesib as monotherapy or with G-CSF support (Pegfilgrastim and Filgrastim). Generally, the efficacy results were disappointing. In two phase I trials in advanced tumor patients (NCT01214629 and NCT01214642), Litronesib was administrated as single agent or in combination with Pegfilgrastim. In the first-in-human study with Litronesib (NCT01214629), the MTD was 4 mg/m2 for Litronesib without G-CSF support, and 6 mg/m2 with Pegfilgrastim. Neutropenia and leukopenia were the most common AEs observed. In total, 26% of patients had SD as the best response, and two PRs (3.7%) were reported in patients with platinum-sensitive ovarian carcinoma and neuroendocrine carcinoma [211], whereas in the other trial (NCT01214642), the safety profile was similar, but no objective responses were observed, with 36.5% of patients achieving SD as the best response [211]. Additionally, another study with Litronesib as monotherapy in patients with solid tumors was initiated in 2011 (NCT01358019). Again, the best response was SD observed in two patients (16.7%) [212]. No further development is planned for this Eg-5 inhibitor, since Kyowa Kirin and Eli Lilly and Company decided to discontinue Litronesib.

6.5. EMD 534085

EMD 534085 was developed by Merck-KGaA and was shown to inhibit Eg-5 with an IC50 of 8 nM in vitro [213]. EMD 534085 entered a phase I trial in patients with solid tumors or lymphoma as a single agent. The MTD was defined as 108 mg/m2 and neutropenia was the most common DLT observed. No objective responses were reported, with 52% of patients achieving SD [214]. No further development is planned with this inhibitor.

6.6. SC-205

4SC-205, developed by 4SC, has entered a phase I study against advanced malignancies as monotherapy (NCT01065025). Patients received 4SC-205 once a week or twice weekly. The MTD was defined as 150 mg/m2 (once weekly) and 75 mg/m2 (twice weekly). Neutropenia was the most common DLT, similarly to the results obtained with the other Eg-5 inhibitors. No CR or PR were reported, and 28% of patients had SD as the best response [215].

6.7. AZD4877

AZD4877, from AstraZeneca, inhibits Eg-5 with an IC50 of 2 nM in vitro, and has entered six phase I/II trials against solid and hematological tumors [216]. Overall, the results were disappointing. In the phase I trials in solid tumor patients (NCT00613652 and NCT00389389), neutropenia was the most common DLT [217,218]. No objective responses were observed, with 31% and 27% of patients achieving SD, respectively [217,218]. Other trials with AML (NCT00486265) and urothelial cancer (NCT00661609) patients demonstrated similar efficacy. In the phase I trial, all AML patients had treatment failure, whereas in the phase II study with urothelial cancer patients, 18% of patients had SD, and no objective responses were reported [219,220]. No further development is planned for AZ4877.

6.8. ARQ 621

ARQ 621, from ArQule, demonstrated a broad spectrum toxicity against a panel of human cancer cell lines [221]. In 2009, ARQ 621 entered a phase I trial with solid tumor patients as monotherapy (NCT00825487). ARQ 621 appeared to be well tolerated at a dose of 280 mg/m2, with the most common AEs being fatigue, nausea, and anemia. No objective responses were observed, with 12.5% of patients achieving SD as the best response [222]. No further development is planned for this compound.

6.9. MK-0731

MK-0731, developed by Merck Sharp & Dohme, inhibits Eg-5 with an IC50 of 2.2 nM in vitro [223]. MK-0731 has entered a phase I trial against advanced solid tumors as monotherapy in 2005 (NCT00104364). The MTD was determined as 17 mg/m2, neutropenia being the major DLT observed [224]. No objective responses were reported and only four patients (4.3%) achieved SD [224]. Development of MK-0731 has been halted.
The safety outcomes of the Eg-5 inhibitors were similar to those of Mps1, Plk1, and Aurora kinase inhibitors, with neutropenia as the most common AE. Generally, Eg-5 inhibitors exhibited poor or modest efficacy in patients with solid tumors, either as monotherapy or in combinational treatments, except for a CR observed with ALN-VSP02 as a single agent in a patient with endometrial cancer with multiple hepatic metastases. The most promising efficacy results were achieved with Filanesib in multiple myeloma patients, especially when combined with the proteasome inhibitors Bortezomib and Carfilzomib. It is expected that Filanesib will enter phase III trials. One possible explanation for the promising results of Filanesib is its better pharmacokinetic profile, primarily a higher half-life compared to the other Eg-5 inhibitors. More studies must be performed to assess which drugs demonstrate synergistic effects with Eg-5 inhibitors, as well as to identify novel and more potent inhibitors with a better pharmacokinetic profile, in order to improve the efficacy of the Eg-5 inhibition-based therapeutic approach.
Table 7. Eg-5 inhibitors in clinical trials 1.
Table 7. Eg-5 inhibitors in clinical trials 1.
CompoundClinical TrialsCurrent StatusConditionsInterventionsOutcomes 2Refs.
SB-715992 (Ispinesib)
Pharmaceutics 13 01011 i036
16 Clinical trialsPhase I/II
14 Completed
2 Terminated
Metastatic Prostate, Kidney and Colorectal Cancer, Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck, Breast, Ovarian and Liver Cancer, Melanoma, Hodgkin’s or Non-Hodgkin’s Lymphoma, Acute Leukemia
Advanced Myelodysplastic Syndromes
MonotherapyBest response was a PR for ovarian and breast cancer[190,191,192,193,194,195,196,197]
Combination with Docetaxel, Capecitabine and CarboplatinBest response was SD
SB-743921
Pharmaceutics 13 01011 i037
NCT00136513Phase I
Completed
Solid TumorsMonotherapyBest response was PR for cholangiocarcinoma[199]
NCT00343564Phase I
Phase II
Completed
Non-Hodgkin Lymphoma and Hodgkin LymphomaMonotherapyBest response was PR for non-Hodgkin lymphoma[200]
ARRY-520 (Filanesib)
Pharmaceutics 13 01011 i038
8 Clinical trialsPhase I/II
Completed
Multiple Myeloma, Advanced/Refractory Myeloid Leukemia, Advanced Solid TumorsMonotherapyBest response was a PR for multiple myeloma[202,203,204,205,206,207]
Combination with Pomalidomide, Dexamethasone, Carfilzomib, Filgrastim, Bortezomib, CarfilzomibBest responses were a CR for multiple myeloma in combination with Pomalidomide, Bortezomib, Dexamethasone, and Filgrastim; PR for multiple myeloma in combination with Bortezomib, Pomalidomide, Dexamethasone, Filgrastim, and Carfilzomib
ALN-VSP02
(siRNA)
NCT01158079Phase I
Completed
Solid TumorsMonotherapyBest response was CR for endometrial cancer with multiple hepatic metastases[209]
NCT00882180Phase I
Completed
LY2523355 (Litronesib)
Pharmaceutics 13 01011 i039
7 Clinical trialsPhase I/II
6 Completed
1 Terminated
Metastatic Breast Cancer, Metastatic and/or Advanced Cancer, Acute Leukemia,
Small Cell Lung Cancer
Monotherapy,
Combination with Pegfilgrastim, and Filgrastim
Best response was PR for non-small-cell lung cancer, ovarian and neuroendocrine carcinomas, and breast cancer in combination with Pegfilgrastim[211,212]
EMD 534085
Pharmaceutics 13 01011 i040
UnknownPhase IAdvanced Solid Tumors,
Lymphoma
MonotherapyBest response was SD[214]
4SC-205
(Structure Undisclosed)
NCT01065025Phase I
Completed
Advanced MalignanciesMonotherapyBest response was SD[215]
AZD4877
Pharmaceutics 13 01011 i041
6 Clinical trialsPhase I/II
3 Completed
3 Terminated
Bladder Cancer, Transitional Cell Bladder Cancer, Urethra Cancer, Ureter Cancer, Renal Pelvis Cancer, Acute Myelogenous Leukemia, Non-Hodgkin LymphomaMonotherapyBest response was SD for non-Hodgkin lymphoma, acute myeloid leukemia, and urothelial cancer[217,218,219,220,225]
ARQ 621
Pharmaceutics 13 01011 i042
NCT00825487Phase I
Completed
Metastatic Solid Tumors,
Refractory/Relapsed Hematologic Malignancies
MonotherapyBest response was SD[222]
MK-0731
Pharmaceutics 13 01011 i043
NCT00104364Phase I
Completed
Advanced Solid MalignanciesMonotherapyBest response was SD for non-small-cell lung cancer, cervical and ovarian cancer[224]
1 Data collected from clinicaltrials.gov. 2 CR, complete remission; PR, partial response; SD, stable disease.

7. Conclusions and Perspectives

MTAs were the first class of antimitotics that demonstrated clinical benefits in cancer patients. However, several tumors have developed resistance, which, together with the neurological and myeloid toxicity provoked by these agents, has led to the search for and development of alternative approaches to block mitosis. Our increased knowledge on mitotic events has provided an opportunity to identify key mitotic proteins that could be targeted for cancer therapy. As described in this review, several inhibitors, known as second-generation antimitotics (SGAs), were identified for Mps1, Aurora kinases, Plk1, Eg-5, and, to a lesser extent, CENP-E, and these have reached clinical trials.
Among all SGAs tested, the most promising molecules are (i) the Plk1 inhibitors Volasertib and Rigosertib, which demonstrated good efficacy results in AML and MDS patients, respectively; (ii) the AurB inhibitor Barasertib for patients with AML; (iii) the AurA inhibitor Alisertib, which has demonstrated antitumor activity against several solid and hematological tumors; (iv) the pan-Aurora inhibitors Ilorasertib and Cenisertib for patients with AML; and (v) the Eg-5 inhibitor Filanesib for patients with multiple myeloma. However, many AEs were reported. The most common AEs associated with these SGAs were hematological, primarily neutropenia. These results were somehow expected given the high proliferation rate of the bone marrow cells [226]. In the same line of thought, better responses were achieved in patients with hematological tumors than in those with solid cancers, in concordance with the lower proliferation rate observed in solid tumors, compared to hematological cancers [226].
Until now, no small molecule has been approved for clinical use, and MTAs still remain the antimitotic agents with the best therapeutic benefit. While SGAs target individual proteins, MTAs destabilize the huge microtubule/cytoskeleton network involved in the dynamics of several proteins and organelles, thereby indirectly affecting their function. This could be a plausible explanation for the MTAs’ therapeutic success, compared to SGAs. Interestingly, better responses were achieved in combinational treatments than with SGAs as monotherapy. Several studies reported the synergistic effects of different SGAs with many drugs such as MTAs or platinum-based agents. Therefore, combinational treatment represents a solid strategy for achieving better therapeutic effectiveness, while decreasing drug dosage and minimizing AEs and resistance.
From an SAC response point of view, the SGAs are divided into two groups [11,227]. The mitotic blockers, which include the inhibitors of CENP-E, Eg-5, and Plk1, activate the SAC, thereby inducing a prolonged mitotic delay that is expected to culminate in cell death. The mitotic drivers, which include the inhibitors of Mps1 and Aurora B kinase, override the SAC and induce premature mitotic exit with extensive chromosome missegregation, resulting in chromosome aberrations that are incompatible with the cell viability of daughter cells. Thus, it seems reasonable to use both approaches for cancer therapy. Yet, some challenges need to be overcome. There is profound inter- and an intra-variability in terms of cell fates following the prolonged mitotic arrest of cancer cells treated with mitotic blockers [228]. Prolonged mitotic arrest can result in cell death in mitosis, or mitotic slippage (also known as checkpoint adaptation), in which cells exit mitosis without cell division and return to the interphase as tetraploid cells. These cells can undergo cell cycle arrest, die, or re-replicate their genomes and endocycle [228]. Slippage is one of the resistance mechanisms against antimitotic drugs.
On the other hand, mitotic drivers can fuel genomic instability. In case of the incomplete inhibition of mitotic driver targets, which is more likely to occur in vivo than in vitro, chromosome segregation errors may be generated below the threshold required to kill cancer cells, which, theoretically, increases genomic instability, thereby fueling malignancy. This may explain the general failure of both mitotic blockers and mitotic drivers in clinical trials when used as monotherapy. Nonetheless, despite their poor clinical activity as single agents, SGAs may be valuable for synthetic lethal combinations intended to selectively target cancer cells, thus decreasing the risk of mitotic slippage, while enhancing the therapeutic window.
SGAs exhibited promising antitumor activity in pre-clinical studies, but failed in clinical trials. A possible reason for this differential antitumor activity is that two-dimensional (2D) cultures lack cell–cell contacts and a natural tumor microenvironment, which are important in tumor signaling and drug response [229]. Using preclinical models that better mimic the tumor microenvironment, such as patient-derived 3D tumors, should increase the predictive value of pre-clinical antimitotic research, helping in anticipating clinical outcomes [230].
The most common AEs observed in patients treated with SGA were related to hematologic dysfunction. The development of drug-delivery systems could be a valuable approach to overcome this issue, facilitating better targeting towards cancer cells and in-tumor drug retention, while increasing the therapeutic window [231].
Patients with the same tumor type respond differently to the same agents, probably due to different genetic and/or epigenetic modifications that alter the sensitivity to a specific drug [232,233]. Thus, patient stratification using predictive biomarkers, together with an in-depth understanding of the mechanisms by which SGAs kill cancer cells, should pave the way to their effective clinical use.

Author Contributions

Review topic and general structure, H.B.; data collection, P.N., writing the review draft, P.N. and H.B.; discussions, revising of text and generation of final version, P.N., P.M.A.S., I.A., and H.B.; preparation of the figures and tables, P.N. and P.M.A.S.; funding acquisition, H.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by CESPU—Cooperativa de Ensino Superior Politécnico e Universitário, under the projects “AntiMitoSphere_APSFCT_IINFACTS_2021” and “ActivCHIRAL_PI2RL_IINFACTS_2021”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Publicly available datasets were analyzed in this study. The data can be found here: [https://clinicaltrials.gov/].

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lim, S.; Kaldis, P. Cdks, cyclins and CKIs: Roles beyond cell cycle regulation. Development 2013, 140, 3079–3093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Compton, D.A. Mechanisms of Aneuploidy. Curr. Opin. Cell Biol. 2011, 23, 109–113. [Google Scholar] [CrossRef] [PubMed]
  4. Silva, P.; Barbosa, J.; Nascimento, A.V.; Faria, J.; Reis, R.; Bousbaa, H. Monitoring the fidelity of mitotic chromosome segregation by the spindle assembly checkpoint. Cell Prolif. 2011, 44, 391–400. [Google Scholar] [CrossRef]
  5. Van Vuuren, R.J.; Visagie, M.H.; Theron, A.E.; Joubert, A.M. Antimitotic drugs in the treatment of cancer. Cancer Chemother. Pharm. 2015, 76, 1101–1112. [Google Scholar] [CrossRef] [Green Version]
  6. Jordan, M.A.; Wilson, L. Microtubules as a target for anticancer drugs. Nat. Rev. Cancer 2004, 4, 253–265. [Google Scholar] [CrossRef]
  7. Matson, D.R.; Stukenberg, P.T. Spindle Poisons and Cell Fate: A Tale of Two Pathways. Mol. Interv. 2011, 11, 141. [Google Scholar] [CrossRef]
  8. Brito, D.A.; Rieder, C.L. Mitotic Checkpoint Slippage in Humans Occurs via Cyclin B Destruction in the Presence of an Active Checkpoint. Curr. Biol. 2006, 16, 1194–1200. [Google Scholar] [CrossRef] [Green Version]
  9. Nakayama, Y.; Inoue, T. Antiproliferative fate of the tetraploid formed after mitotic slippage and its promotion; a novel target for cancer therapy based on microtubule poisons. Molecules 2016, 21, 663. [Google Scholar] [CrossRef] [Green Version]
  10. Dumontet, C.; Jordan, M.A. Microtubule-binding agents: A dynamic field of cancer therapeutics. Nat. Rev. Drug Discov. 2010, 9, 790–803. [Google Scholar] [CrossRef] [Green Version]
  11. Henriques, A.C.; Ribeiro, D.; Pedrosa, J.; Sarmento, B.; Silva, P.M.A.; Bousbaa, H. Mitosis inhibitors in anticancer therapy: When blocking the exit becomes a solution. Cancer Lett. 2019, 440, 64–81. [Google Scholar] [CrossRef]
  12. Lauzé, E.; Stoelcker, B.; Luca, F.C.; Weiss, E.; Schutz, A.R.; Winey, M. Yeast spindle pole body duplication gene MPS1 encodes an essential dual specificity protein kinase. EMBO J. 1995, 14, 1655–1663. [Google Scholar] [CrossRef]
  13. Ji, Z.; Gao, H.; Jia, L.; Li, B.; Yu, H. A sequential multi-target Mps1 phosphorylation cascade promotes spindle checkpoint signaling. Elife 2017, 6. [Google Scholar] [CrossRef]
  14. Liu, X.; Winey, M. The MPS1 family of protein kinases. Annu. Rev. Biochem. 2012, 81, 561–585. [Google Scholar] [CrossRef] [Green Version]
  15. Yang, H.; Zhang, F.; Huang, C.-J.; Liao, J.; Han, Y.; Hao, P.; Chu, Y.; Lu, X.; Li, W.; Yu, H.; et al. Mps1 regulates spindle morphology through MCRS1 to promote chromosome alignment. Mol. Biol. Cell 2019, 30, 1060–1068. [Google Scholar] [CrossRef]
  16. Maciejowski, J.; Drechsler, H.; Grundner-Culemann, K.; Ballister, E.R.; Rodriguez-Rodriguez, J.-A.; Rodriguez-Bravo, V.; Jones, M.J.K.; Foley, E.; Lampson, M.A.; Daub, H.; et al. Mps1 Regulates Kinetochore-Microtubule Attachment Stability via the Ska Complex to Ensure Error-Free Chromosome Segregation. Dev. Cell 2017, 41, 143–156. [Google Scholar] [CrossRef] [Green Version]
  17. Xie, Y.; Wang, A.; Lin, J.; Wu, L.; Zhang, H.; Yang, X.; Wan, X.; Miao, R.; Sang, X.; Zhao, H. Mps1/TTK: A novel target and biomarker for cancer. J. Drug Target. 2017, 25, 112–118. [Google Scholar] [CrossRef]
  18. Jemaà, M.; Galluzzi, L.; Kepp, O.; Senovilla, L.; Brands, M.; Boemer, U.; Koppitz, M.; Lienau, P.; Prechtl, S.; Schulze, V.; et al. Characterization of novel MPS1 inhibitors with preclinical anticancer activity. Cell Death Differ. 2013, 20, 1532–1545. [Google Scholar] [CrossRef]
  19. Kwiatkowski, N.; Jelluma, N.; Filippakopoulos, P.; Soundararajan, M.; Manak, M.S.; Kwon, M.; Choi, H.G.; Sim, T.; Deveraux, Q.L.; Rottmann, S.; et al. Small-molecule kinase inhibitors provide insight into Mps1 cell cycle function. Nat. Chem. Biol. 2010, 6, 359–368. [Google Scholar] [CrossRef] [Green Version]
  20. Janssen, A.; Kops, G.J.P.L.; Medema, R.H. Elevating the frequency of chromosome mis-segregation as a strategy to kill tumor cells. Proc. Natl. Acad. Sci. USA 2009, 106, 19108–19113. [Google Scholar] [CrossRef] [Green Version]
  21. Wengner, A.M.; Siemeister, G.; Koppitz, M.; Schulze, V.; Kosemund, D.; Klar, U.; Stoeckigt, D.; Neuhaus, R.; Lienau, P.; Bader, B.; et al. Novel Mps1 Kinase Inhibitors with Potent Antitumor Activity. Mol. Cancer Ther. 2016, 15, 583–592. [Google Scholar] [CrossRef] [Green Version]
  22. Lorusso, P.; Chawla, S.P.; Bendell, J.; Shields, A.F.; Shapiro, G.; Rajagopalan, P.; Cyris, C.; Bruns, I.; Mei, J.; Souza, F.; et al. First-in-human study of the monopolar spindle 1 (Mps1) kinase inhibitor BAY 1161909 in combination with paclitaxel in subjects with advanced malignancies. Ann. Oncol. 2018, 29, viii138. [Google Scholar] [CrossRef]
  23. Colombo, R.; Caldarelli, M.; Giorgini, M.L.; Degrassi, A.; Ciomei, M.; Pezzetta, D.; Ballinari, D.; Montagnoli, A.; Pesenti, E.; Donati, D.; et al. Abstract 2097: Targeting aneuploidy with NMS-P153, a tight binder inhibitor of the spindle assembly checkpoint MPS1 (TTK) kinase. Cancer Res. 2013, 73, 2097. [Google Scholar] [CrossRef]
  24. Colombo, R.; Burbridge, M.; Rodriguez, M.; Cantero, F.; Caldarelli, M.; Giorgini, M.L.; Sola, F.; Ballinari, D.; Ciomei, M.; Bosotti, R.; et al. Abstract 1638: Preclinical characterization of the novel TTK kinase inhibitor S81694 for the treatment of triple negative breast cancer. Cancer Res. 2015, 75, 1638. [Google Scholar] [CrossRef]
  25. Woodward, H.L.; Innocenti, P.; Cheung, K.-M.J.; Hayes, A.; Roberts, J.; Henley, A.T.; Faisal, A.; Mak, G.W.-Y.; Box, G.; Westwood, I.M.; et al. Introduction of a Methyl Group Curbs Metabolism of Pyrido[3,4- d]pyrimidine Monopolar Spindle 1 (MPS1) Inhibitors and Enables the Discovery of the Phase 1 Clinical Candidate N 2-(2-Ethoxy-4-(4-methyl-4 H-1,2,4-triazol-3-yl)phenyl)-6-methyl- N 8-neopentylp. J. Med. Chem. 2018, 61, 8226–8240. [Google Scholar] [CrossRef]
  26. Anderhub, S.J.; Mak, G.W.-Y.; Gurden, M.D.; Faisal, A.; Drosopoulos, K.; Walsh, K.; Woodward, H.L.; Innocenti, P.; Westwood, I.M.; Naud, S.; et al. High Proliferation Rate and a Compromised Spindle Assembly Checkpoint Confers Sensitivity to the MPS1 Inhibitor BOS172722 in Triple-Negative Breast Cancers. Mol. Cancer Ther. 2019, 18, 1696–1707. [Google Scholar] [CrossRef] [Green Version]
  27. Mason, J.M.; Wei, X.; Fletcher, G.C.; Kiarash, R.; Brokx, R.; Hodgson, R.; Beletskaya, I.; Bray, M.R.; Mak, T.W. Functional characterization of CFI-402257, a potent and selective Mps1/TTK kinase inhibitor, for the treatment of cancer. Proc. Natl. Acad. Sci. USA 2017, 114, 3127–3132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Zheng, L.; Chen, Z.; Kawakami, M.; Chen, Y.; Roszik, J.; Mustachio, L.M.; Kurie, J.M.; Villalobos, P.; Lu, W.; Behrens, C.; et al. Tyrosine threonine kinase inhibition eliminates lung cancers by augmenting apoptosis and polyploidy. Mol. Cancer. Ther. 2019, 18, 1775–1786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Szymiczek, A.; Carbone, M.; Pastorino, S.; Napolitano, A.; Tanji, M.; Minaai, M.; Pagano, I.; Mason, J.M.; Pass, H.I.; Bray, M.R.; et al. Inhibition of the spindle assembly checkpoint kinase Mps-1 as a novel therapeutic strategy in malignant mesothelioma. Oncogene 2017, 36, 6501–6507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. de Boussac, H.; Bruyer, A.; Jourdan, M.; Maes, A.; Robert, N.; Gourzones, C.; Vincent, L.; Seckinger, A.; Cartron, G.; Hose, D.; et al. Kinome expression profiling to target new therapeutic avenues in multiple myeloma. Haematologica 2020, 105, 784–795. [Google Scholar] [CrossRef] [PubMed]
  31. Bruinsma, W.; Raaijmakers, J.A.; Medema, R.H. Switching Polo-like kinase-1 on and off in time and space. Trends Biochem. Sci. 2012, 37, 534–542. [Google Scholar] [CrossRef]
  32. Colicino, E.G.; Hehnly, H. Regulating a key mitotic regulator, polo-like kinase 1 (PLK1). Cytoskeleton 2018, 75, 481–494. [Google Scholar] [CrossRef] [Green Version]
  33. Combes, G.; Alharbi, I.; Braga, L.G.; Elowe, S. Playing polo during mitosis: PLK1 takes the lead. Oncogene 2017, 36, 4819–4827. [Google Scholar] [CrossRef]
  34. Wang, H.; Tao, Z.; Feng, M.; Li, X.; Deng, Z.; Zhao, G.; Yin, H.; Pan, T.; Chen, G.; Feng, Z.; et al. Dual PLK1 and STAT3 inhibition promotes glioblastoma cells apoptosis through MYC. Biochem. Biophys. Res. Commun. 2020, 533, 368–375. [Google Scholar] [CrossRef]
  35. Ueda, A.; Oikawa, K.; Fujita, K.; Ishikawa, A.; Sato, E.; Ishikawa, T.; Kuroda, M.; Kanekura, K. Therapeutic potential of PLK1 inhibition in triple-negative breast cancer. Lab. Investig. 2019, 99, 1275–1286. [Google Scholar] [CrossRef]
  36. Pajtler, K.W.; Sadowski, N.; Ackermann, S.; Althoff, K.; Schönbeck, K.; Batzke, K.; Sch, S.; Odersky, A.; Heukamp, L.; Astrahantseff, K.; et al. The GSK461364 PLK1 inhibitor exhibits strong antitumoral activity in preclinical neuroblastoma models. Oncotarget 2016, 8, 6730–6741. [Google Scholar] [CrossRef]
  37. Gutteridge, R.E.A.; Ndiaye, M.A.; Liu, X.; Ahmad, N. Plk1 inhibitors in cancer therapy: From laboratory to clinics. Mol. Cancer Ther. 2016, 15, 1427–1435. [Google Scholar] [CrossRef] [Green Version]
  38. Liu, Z.; Sun, Q.; Wang, X. PLK1, A Potential Target for Cancer Therapy. Transl. Oncol. 2016, 10, 22–32. [Google Scholar] [CrossRef] [Green Version]
  39. Lénárt, P.; Petronczki, M.; Steegmaier, M.; Di Fiore, B.; Lipp, J.J.; Hoffmann, M.; Rettig, W.J.; Kraut, N.; Peters, J.-M. The Small-Molecule Inhibitor BI 2536 Reveals Novel Insights into Mitotic Roles of Polo-like Kinase 1. Curr. Biol. 2007, 17, 304–315. [Google Scholar] [CrossRef] [Green Version]
  40. Sebastian, M.; Reck, M.; Waller, C.F.; Kortsik, C.; Frickhofen, N.; Schuler, M.; Fritsch, H.; Gaschler-Markefski, B.; Hanft, G.; Munzert, G.; et al. The efficacy and safety of BI 2536, a novel Plk-1 inhibitor, in patients with stage IIIB/IV non-small cell lung cancer who had relapsed after, or failed, chemotherapy: Results from an open-label, randomized phase ii clinical trial. J. Thorac. Oncol. 2010, 5, 1060–1067. [Google Scholar] [CrossRef]
  41. Ellis, P.M.; Chu, Q.S.; Leighl, N.; Laurie, S.A.; Fritsch, H.; Gaschler-Markefski, B.; Gyorffy, S.; Munzert, G. A phase i open-label dose-escalation study of intravenous BI 2536 together with pemetrexed in previously treated patients with non-small-cell lung cancer. Clin. Lung Cancer 2013, 14, 19–27. [Google Scholar] [CrossRef]
  42. Mross, K.; Dittrich, C.; Aulitzky, W.E.; Strumberg, D.; Schutte, J.; Schmid, R.M.; Hollerbach, S.; Merger, M.; Munzert, G.; Fleischer, F.; et al. A randomised phase II trial of the Polo-like kinase inhibitor BI 2536 in chemo-nave patients with unresectable exocrine adenocarcinoma of the pancreas-a study within the Central European Society Anticancer Drug Research (CESAR) collaborative network. Br. J. Cancer 2012, 107, 280–286. [Google Scholar] [CrossRef] [Green Version]
  43. Müller-Tidow, C.; Bug, G.; Lübbert, M.; Krämer, A.; Krauter, J.; Valent, P.; Nachbaur, D.; Berdel, W.E.; Ottmann, O.G.; Fritsch, H.; et al. A randomized, open-label, phase I/II trial to investigate the maximum tolerated dose of the Polo-like kinase inhibitor BI 2536 in elderly patients with refractory/relapsed acute myeloid leukaemia. Br. J. Haematol. 2013, 163, 214–222. [Google Scholar] [CrossRef]
  44. Vose, J.M.; Friedberg, J.W.; Waller, E.K.; Cheson, B.D.; Juvvigunta, V.; Fritsch, H.; Petit, C.; Munzert, G.; Younes, A. The Plk1 inhibitor BI 2536 in patients with refractory or relapsed non-Hodgkin lymphoma: A phase I, open-label, single dose-escalation study. Leuk. Lymphoma 2013, 54, 708–713. [Google Scholar] [CrossRef]
  45. Rudolph, D.; Steegmaier, M.; Hoffmann, M.; Grauert, M.; Baum, A.; Quant, J.; Haslinger, C.; Garin-Chesa, P.; Adolf, G.R. BI 6727, a polo-like kinase inhibitor with improved pharmacokinetic profile and broad antitumor activity. Clin. Cancer Res. 2009, 15, 3094–3102. [Google Scholar] [CrossRef] [Green Version]
  46. Schöffski, P.; Awada, A.; Dumez, H.; Gil, T.; Bartholomeus, S.; Wolter, P.; Taton, M.; Fritsch, H.; Glomb, P.; Munzert, G. A phase I, dose-escalation study of the novel Polo-like kinase inhibitor volasertib (BI 6727) in patients with advanced solid tumours. Eur. J. Cancer 2012, 48, 179–186. [Google Scholar] [CrossRef]
  47. Nokihara, H.; Yamada, Y.; Fujiwara, Y.; Yamamoto, N.; Wakui, H.; Nakamichi, S.; Kitazono, S.; Inoue, K.; Harada, A.; Taube, T.; et al. Phase I trial of volasertib, a Polo-like kinase inhibitor, in Japanese patients with advanced solid tumors. Investig. New Drugs 2016, 34, 66–74. [Google Scholar] [CrossRef]
  48. Lin, C.-C.; Su, W.-C.; Yen, C.-J.; Hsu, C.-H.; Su, W.-P.; Yeh, K.-H.; Lu, Y.-S.; Cheng, A.-L.; Huang, D.C.-L.; Fritsch, H.; et al. A phase I study of two dosing schedules of volasertib (BI 6727), an intravenous polo-like kinase inhibitor, in patients with advanced solid malignancies. Br. J. Cancer 2014, 110, 2434–2440. [Google Scholar] [CrossRef] [Green Version]
  49. Stadler, W.M.; Vaughn, D.J.; Sonpavde, G.; Vogelzang, N.J.; Tagawa, S.T.; Petrylak, D.P.; Rosen, P.; Lin, C.-C.; Mahoney, J.; Modi, S.; et al. An open-label, single-arm, phase 2 trial of the polo-like kinase inhibitor volasertib (BI 6727) in patients with locally advanced or metastatic urothelial cancer. Cancer 2014, 120, 976–982. [Google Scholar] [CrossRef] [Green Version]
  50. Pujade-Lauraine, E.; Selle, F.; Weber, B.; Ray-Coquard, I.-L.; Vergote, I.; Sufliarsky, J.; Del Campo, J.M.; Lortholary, A.; Lesoin, A.; Follana, P.; et al. Volasertib Versus Chemotherapy in Platinum-Resistant or -Refractory Ovarian Cancer: A Randomized Phase II Groupe des Investigateurs Nationaux pour l’Etude des Cancers de l’Ovaire Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 706–713. [Google Scholar] [CrossRef]
  51. Ellis, P.M.; Leighl, N.B.; Hirsh, V.; Reaume, M.N.; Blais, N.; Wierzbicki, R.; Sadrolhefazi, B.; Gu, Y.; Liu, D.; Pilz, K.; et al. A Randomized, Open-Label Phase II Trial of Volasertib as Monotherapy and in Combination With Standard-Dose Pemetrexed Compared With Pemetrexed Monotherapy in Second-Line Treatment for Non-Small-Cell Lung Cancer. Clin. Lung Cancer 2015, 16, 457–465. [Google Scholar] [CrossRef] [PubMed]
  52. De Braud, F.; Cascinu, S.; Spitaleri, G.; Pilz, K.; Clementi, L.; Liu, D.; Sikken, P.; De Pas, T. A phase I, dose-escalation study of volasertib combined with nintedanib in advanced solid tumors. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2015, 26, 2341–2346. [Google Scholar] [CrossRef] [PubMed]
  53. Machiels, J.-P.; Peeters, M.; Herremans, C.; Surmont, V.; Specenier, P.; De Smet, M.; Pilz, K.; Strelkowa, N.; Liu, D.; Rottey, S. A phase I study of volasertib combined with afatinib, in advanced solid tumors. Cancer Chemother. Pharm. Ther. 2015, 76, 843–851. [Google Scholar] [CrossRef] [PubMed]
  54. Awada, A.; Dumez, H.; Aftimos, P.G.; Costermans, J.; Bartholomeus, S.; Forceville, K.; Berghmans, T.; Meeus, M.-A.; Cescutti, J.; Munzert, G.; et al. Phase I trial of volasertib, a Polo-like kinase inhibitor, plus platinum agents in solid tumors: Safety, pharmacokinetics and activity. Investig. New Drugs 2015, 33, 611–620. [Google Scholar] [CrossRef]
  55. Kobayashi, Y.; Yamauchi, T.; Kiyoi, H.; Sakura, T.; Hata, T.; Ando, K.; Watabe, A.; Harada, A.; Taube, T.; Miyazaki, Y.; et al. Phase I trial of volasertib, a Polo-like kinase inhibitor, in Japanese patients with acute myeloid leukemia. Cancer Sci. 2015, 106, 1590–1595. [Google Scholar] [CrossRef]
  56. Gumireddy, K.; Reddy, M.V.R.; Cosenza, S.C.; Nathan, R.B.; Baker, S.J.; Papathi, N.; Jiang, J.; Holland, J.; Reddy, E.P. ON01910 a non-ATP-competitive small molecule inhibitor of Plk1, is a potent anticancer agent. Cancer Cell 2005, 7, 275–286. [Google Scholar] [CrossRef] [Green Version]
  57. Ma, W.W.; Messersmith, W.A.; Dy, G.K.; Weekes, C.D.; Whitworth, A.; Ren, C.; Maniar, M.; Wilhelm, F.; Eckhardt, S.G.; Adjei, A.A.; et al. Phase I study of Rigosertib, an inhibitor of the phosphatidylinositol 3-kinase and Polo-like kinase 1 pathways, combined with gemcitabine in patients with solid tumors and pancreatic cancer. Clin. Cancer Res. 2012, 18, 2048–2055. [Google Scholar] [CrossRef] [Green Version]
  58. O’Neil, B.H.; Scott, A.J.; Ma, W.W.; Cohen, S.J.; Aisner, D.L.; Menter, A.R.; Tejani, M.A.; Cho, J.K.; Granfortuna, J.; Coveler, L.; et al. A phase II/III randomized study to compare the efficacy and safety of rigosertib plus gemcitabine versus gemcitabine alone in patients with previously untreated metastatic pancreatic cancer. Ann. Oncol. 2015, 26, 1923–1929. [Google Scholar] [CrossRef]
  59. Garcia-Manero, G.; Fenaux, P.; Al-Kali, A.; Baer, M.R.; Sekeres, M.A.; Roboz, G.J.; Gaidano, G.; Scott, B.L.; Greenberg, P.; Platzbecker, U.; et al. Rigosertib versus best supportive care for patients with high-risk myelodysplastic syndromes after failure of hypomethylating drugs (ONTIME): A randomised, controlled, phase 3 trial. Lancet Oncol. 2016, 17, 496–508. [Google Scholar] [CrossRef]
  60. Al-Kali, A.; Hiwase, D.; Baer, M.R.; Greenberg, P.; Shortt, J.; Collins, R.; Steensma, D.P.; Verma, A.; Roboz, G.J.; Shammo, J.M.; et al. Relationship of bone marrow blast (BMBL) response to overall survival (OS) in a multicenter study of rigosertib (Rigo) in patients (pts) with myelodysplastic syndrome (MDS) with excess blasts progressing on or after treatment with a hypomethylating agent. JCO 2017, 35, 7056. [Google Scholar] [CrossRef]
  61. Navada, S.C.; Garcia-Manero, G.; OdchimarReissig, R.; Pemmaraju, N.; Alvarado, Y.; Ohanian, M.N.; John, R.B.; Demakos, E.P.; Zbyszewski, P.S.; Maniar, M.; et al. Rigosertib in combination with azacitidine in patients with myelodysplastic syndromes or acute myeloid leukemia: Results of a phase 1 study. Leuk Res. 2020, 94, 106369. [Google Scholar] [CrossRef]
  62. Gilmartin, A.G.; Bleam, M.R.; Richter, M.C.; Erskine, S.G.; Kruger, R.G.; Madden, L.; Hassler, D.F.; Smith, G.K.; Gontarek, R.R.; Courtney, M.P.; et al. Distinct concentration-dependent effects of the polo-like kinase 1-specific inhibitor GSK461364A, including differential effect on apoptosis. Cancer Res. 2009, 69, 6969–6977. [Google Scholar] [CrossRef] [Green Version]
  63. Olmos, D.; Barker, D.; Sharma, R.; Brunetto, A.T.; Yap, T.A.; Taegtmeyer, A.B.; Barriuso, J.; Medani, H.; Degenhardt, Y.Y.; Allred, A.J.; et al. Phase I study of GSK461364, a specific and competitive Polo-like kinase 1 inhibitor, in patients with advanced solid malignancies. Clin. Cancer Res. An. Off. J. Am. Assoc. Cancer Res. 2011, 17, 3420–3430. [Google Scholar] [CrossRef] [Green Version]
  64. Doi, T.; Murakami, H.; Wan, K.; Miki, M.; Kotani, H.; Sakamoto, N.; Yamamoto, N.; Ohtsu, A. A first-in-human phase I dose-escalation study of MK-1496, first-in-class orally available novel PLK1 inhibitor, in patients with advanced solid tumors. JCO 2011, 29, 3012. [Google Scholar] [CrossRef]
  65. Nie, Z.; Feher, V.; Natala, S.; McBride, C.; Kiryanov, A.; Jones, B.; Lam, B.; Liu, Y.; Kaldor, S.; Stafford, J.; et al. Discovery of TAK-960: An orally available small molecule inhibitor of polo-like kinase 1 (PLK1). Bioorganic. Med. Chem. Lett. 2013, 23, 3662–3666. [Google Scholar] [CrossRef]
  66. Hikichi, Y.; Honda, K.; Hikami, K.; Miyashita, H.; Kaieda, I.; Murai, S.; Uchiyama, N.; Hasegawa, M.; Kawamoto, T.; Sato, T.; et al. TAK-960, a novel, orally available, selective inhibitor of polo-like kinase 1, shows broad-spectrum preclinical antitumor activity in multiple dosing regimens. Mol. Cancer Ther. 2012, 11, 700–709. [Google Scholar] [CrossRef] [Green Version]
  67. Valsasina, B.; Beria, I.; Alli, C.; Alzani, R.; Avanzi, N.; Ballinari, D.; Cappella, P.; Caruso, M.; Casolaro, A.; Ciavolella, A.; et al. NMS-P937, an orally available, specific small-molecule polo-like kinase 1 inhibitor with antitumor activity in solid and hematologic malignancies. Mol. Cancer Ther. 2012, 11, 1006–1016. [Google Scholar] [CrossRef] [Green Version]
  68. Weiss, G.J.; Jameson, G.; Von Hoff, D.D.; Valsasina, B.; Davite, C.; Di Giulio, C.; Fiorentini, F.; Alzani, R.; Carpinelli, P.; Di Sanzo, A.; et al. Phase I dose escalation study of NMS-1286937, an orally available Polo-Like Kinase 1 inhibitor, in patients with advanced or metastatic solid tumors. Investig. New Drugs 2018, 36, 85–95. [Google Scholar] [CrossRef] [Green Version]
  69. Demeure, M.J.; Armaghany, T.; Ejadi, S.; Ramanathan, R.K.; Elfiky, A.; Strosberg, J.R.; Smith, D.C.; Whitsett, T.; Liang, W.S.; Sekar, S.; et al. A phase I/II study of TKM-080301, a PLK1 -targeted RNAi in patients with adrenocortical cancer (ACC). JCO 2016, 34, 2547. [Google Scholar] [CrossRef]
  70. El Dika, I.; Lim, H.Y.; Yong, W.P.; Lin, C.-C.; Yoon, J.-H.; Modiano, M.; Freilich, B.; Choi, H.J.; Chao, T.-Y.; Kelley, R.K.; et al. An Open-Label, Multicenter, Phase I, Dose Escalation Study with Phase II Expansion Cohort to Determine the Safety, Pharmacokinetics, and Preliminary Antitumor Activity of Intravenous TKM-080301 in Subjects with Advanced Hepatocellular Carcinoma. Oncologist 2019, 24, 747. [Google Scholar] [CrossRef] [Green Version]
  71. Available online: https://cyclacel.com/research_programs_oncology_cyc140.shtml (accessed on 8 May 2021).
  72. Navada, S.C.; Fruchtman, S.M.; Odchimar-Reissig, R.; Demakos, E.P.; Petrone, M.E.; Zbyszewski, P.S.; Holland, J.F.; Silverman, L.R. A phase 1/2 study of rigosertib in patients with myelodysplastic syndromes (MDS) and MDS progressed to acute myeloid leukemia. Leuk Res. 2018, 64, 10–16. [Google Scholar] [CrossRef]
  73. Komrokji, R.S.; Raza, A.; Lancet, J.E.; Ren, C.; Taft, D.; Maniar, M.; Wilhelm, F.; List, A.F. Phase I clinical trial of oral rigosertib in patients with myelodysplastic syndromes. Br. J. Haematol. 2013, 162, 517–524. [Google Scholar] [CrossRef]
  74. King, S.I.; Purdie, C.A.; Bray, S.E.; Quinlan, P.R.; Jordan, L.B.; Thompson, A.M.; Meek, D.W. Immunohistochemical detection of Polo-like kinase-1 (PLK1) in primary breast cancer is associated with TP53 mutation and poor clinical outcome. Breast Cancer Res. 2012, 14, R40. [Google Scholar] [CrossRef] [Green Version]
  75. Guan, R.; Tapang, P.; Leverson, J.D.; Albert, D.; Giranda, V.L.; Luo, Y. Small Interfering RNA–Mediated Polo-Like Kinase 1 Depletion Preferentially Reduces the Survival of p53-Defective, Oncogenic Transformed Cells and Inhibits Tumor Growth in Animals. Cancer Res. 2005, 65, 2698–2704. [Google Scholar] [CrossRef] [Green Version]
  76. Bolanos-Garcia, V.M. Aurora kinases. Int. J. Biochem. Cell Biol. 2005, 37, 1572–1577. [Google Scholar] [CrossRef]
  77. Willems, E.; Dedobbeleer, M.; Digregorio, M.; Lombard, A.; Lumapat, P.N.; Rogister, B. The functional diversity of Aurora kinases: A comprehensive review. Cell Div. 2018, 13, 7. [Google Scholar] [CrossRef] [Green Version]
  78. Girdler, F.; Gascoigne, K.E.; Eyers, P.A.; Hartmuth, S.; Crafter, C.; Foote, K.M.; Keen, N.J.; Taylor, S.S. Validating Aurora B as an anti-cancer drug target. J. Cell Sci. 2006, 119, 3664–3675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Hauf, S.; Cole, R.W.; LaTerra, S.; Zimmer, C.; Schnapp, G.; Walter, R.; Heckel, A.; van Meel, J.; Rieder, C.L.; Peters, J.-M. The small molecule Hesperadin reveals a role for Aurora B in correcting kinetochore-microtubule attachment and in maintaining the spindle assembly checkpoint. J. Cell Biol. 2003, 161, 281–294. [Google Scholar] [CrossRef] [PubMed]
  80. Du, J.; Yan, L.; Torres, R.; Gong, X.; Bian, H.; Marugán, C.; Boehnke, K.; Baquero, C.; Hui, Y.-H.; Chapman, S.C.; et al. Aurora A-Selective Inhibitor LY3295668 Leads to Dominant Mitotic Arrest, Apoptosis in Cancer Cells, and Shows Potent Preclinical Antitumor Efficacy. Mol. Cancer Ther. 2019, 18, 2207–2219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Kaestner, P.; Stolz, A.; Bastians, H. Determinants for the efficiency of anticancer drugs targeting either Aurora-A or Aurora-B kinases in human colon carcinoma cells. Mol. Cancer Ther. 2009, 8, 2046–2056. [Google Scholar] [CrossRef] [Green Version]
  82. Glaser, K.B.; Li, J.; Marcotte, P.A.; Magoc, T.J.; Guo, J.; Reuter, D.R.; Tapang, P.; Wei, R.-Q.; Pease, L.J.; Bui, M.H.; et al. Preclinical characterization of ABT-348, a kinase inhibitor targeting the aurora, vascular endothelial growth factor receptor/platelet-derived growth factor receptor, and Src kinase families. J. Pharm. Exp. Ther. 2012, 343, 617–627. [Google Scholar] [CrossRef]
  83. Maitland, M.L.; Piha-Paul, S.; Falchook, G.; Kurzrock, R.; Nguyen, L.; Janisch, L.; Karovic, S.; McKee, M.; Hoening, E.; Wong, S.; et al. Clinical pharmacodynamic/exposure characterisation of the multikinase inhibitor ilorasertib (ABT-348)in a phase 1 dose-escalation trial. Br. J. Cancer 2018, 118, 1042–1050. [Google Scholar] [CrossRef]
  84. Garcia-Manero, G.; Tibes, R.; Kadia, T.; Kantarjian, H.; Arellano, M.; Knight, E.A.; Xiong, H.; Qin, Q.; Munasinghe, W.; Roberts-Rapp, L.; et al. Phase 1 dose escalation trial of ilorasertib, a dual Aurora/VEGF receptor kinase inhibitor, in patients with hematologic malignancies. Investig. New Drugs 2015, 33, 870–880. [Google Scholar] [CrossRef] [Green Version]
  85. Peter, B.; Bibi, S.; Eisenwort, G.; Wingelhofer, B.; Berger, D.; Stefanzl, G.; Blatt, K.; Herrmann, H.; Hadzijusufovic, E.; Hoermann, G.; et al. Drug-induced inhibition of phosphorylation of STAT5 overrides drug resistance in neoplastic mast cells. Leukemia 2018, 32, 1016–1022. [Google Scholar] [CrossRef]
  86. Mita, M.; Gordon, M.; Rejeb, N.; Gianella-Borradori, A.; Jego, V.; Mita, A.; Sarantopoulos, J.; Sankhala, K.; Mendelson, D. A phase l study of three different dosing schedules of the oral aurora kinase inhibitor MSC1992371A in patients with solid tumors. Target. Oncol. 2014, 9, 215–224. [Google Scholar] [CrossRef]
  87. Raymond, E.; Alexandre, J.; Faivre, S.; Goldwasser, F.; Besse-Hammer, T.; Gianella-Borradori, A.; Jego, V.; Trandafir, L.; Rejeb, N.; Awada, A. A phase I schedule dependency study of the aurora kinase inhibitor MSC1992371A in combination with gemcitabine in patients with solid tumors. Investig. New Drugs 2014, 32, 94–103. [Google Scholar] [CrossRef]
  88. Graux, C.; Sonet, A.; Maertens, J.; Duyster, J.; Greiner, J.; Chalandon, Y.; Martinelli, G.; Hess, D.; Heim, D.; Giles, F.J.; et al. A phase I dose-escalation study of MSC1992371A, an oral inhibitor of aurora and other kinases, in advanced hematologic malignancie. Leuk Res. 2013, 37, 1100–1106. [Google Scholar] [CrossRef] [Green Version]
  89. Harrington, E.A.; Bebbington, D.; Moore, J.; Rasmussen, R.K.; Ajose-Adeogun, A.O.; Nakayama, T.; Graham, J.A.; Demur, C.; Hercend, T.; Diu-Hercend, A.; et al. VX-680, a potent and selective small-molecule inhibitor of the Aurora kinases, suppresses tumor growth in vivo. Nat. Med. 2004, 10, 262–267. [Google Scholar] [CrossRef]
  90. Traynor, A.M.; Hewitt, M.; Liu, G.; Flaherty, K.T.; Clark, J.; Freedman, S.J.; Scott, B.B.; Leighton, A.M.; Watson, P.A.; Zhao, B.; et al. Phase I dose escalation study of MK-0457, a novel Aurora kinase inhibitor, in adult patients with advanced solid tumors. Cancer Chemother. Pharm. 2011, 67, 305–314. [Google Scholar] [CrossRef] [Green Version]
  91. Giles, F.J.; Swords, R.T.; Nagler, A.; Hochhaus, A.; Ottmann, O.G.; Rizzieri, D.A.; Talpaz, M.; Clark, J.; Watson, P.; Xiao, A.; et al. MK-0457, an Aurora kinase and BCR-ABL inhibitor, is active in patients with BCR-ABL T315I leukemia. Leukemia 2013, 27, 113–117. [Google Scholar] [CrossRef]
  92. Sini, P.; Gürtler, U.; Zahn, S.K.; Baumann, C.; Rudolph, D.; Baumgartinger, R.; Strauss, E.; Haslinger, C.; Tontsch-Grunt, U.; Waizenegger, I.C.; et al. Pharmacological profile of BI 847325, an orally bioavailable, ATP-competitive inhibitor of MEK and Aurora kinases. Mol. Cancer Ther. 2016, 15, 2388–2398. [Google Scholar] [CrossRef] [Green Version]
  93. Schöffski, P.; Aftimos, P.; Dumez, H.; Deleporte, A.; De Block, K.; Costermans, J.; Billiet, M.; Meeus, M.-A.; Lee, C.; Schnell, D.; et al. A phase I study of two dosing schedules of oral BI 847325 in patients with advanced solid tumors. Cancer Chemother Pharm. 2016, 77, 99–108. [Google Scholar] [CrossRef]
  94. Howard, S.; Berdini, V.; Boulstridge, J.A.; Carr, M.G.; Cross, D.M.; Curry, J.; Devine, L.A.; Early, T.R.; Fazal, L.; Gill, A.L.; et al. Fragment-based discovery of the pyrazol-4-yl urea (AT9283), a multitargeted kinase inhibitor with potent aurora kinase activity. J. Med. Chem 2009, 52, 379–388. [Google Scholar] [CrossRef]
  95. Dent, S.F.; Gelmon, K.A.; Chi, K.N.; Jonker, D.J.; Wainman, N.; Capier, C.A.; Chen, E.X.; Lyons, J.F.; Seymour, L. NCIC CTG IND.181: Phase i study of AT9283 given as a weekly 24 hour infusion in advanced malignancies. Investig. New Drugs 2013, 31, 1522–1529. [Google Scholar] [CrossRef] [PubMed]
  96. Foran, J.; Ravandi, F.; Wierda, W.; Garcia-Manero, G.; Verstovsek, S.; Kadia, T.; Burger, J.; Yule, M.; Langford, G.; Lyons, J.; et al. A phase i and pharmacodynamic study of AT9283, a small-molecule inhibitor of aurora kinases in patients with relapsed/refractory leukemia or myelofibrosis. Clin. Lymphoma Myeloma Leuk. 2014, 14, 223–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Moreno, L.; Marshall, L.V.; Pearson, A.D.J.; Morland, B.; Elliott, M.; Campbell-Hewson, Q.; Makin, G.; Halford, S.E.R.; Acton, G.; Ross, P.; et al. A phase i trial of AT9283 (a selective inhibitor of aurora kinases) in children and adolescents with solid tumors: A cancer research UK study. Clin. Cancer Res. 2015, 21, 267–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Payton, M.; Bush, T.L.; Chung, G.; Ziegler, B.; Eden, P.; McElroy, P.; Ross, S.; Cee, V.J.; Deak, H.L.; Hodous, B.L.; et al. Preclinical evaluation of AMG 900, a novel potent and highly selective pan-aurora kinase inhibitor with activity in taxane-resistant tumor cell lines. Cancer Res. 2010, 70, 9846–9854. [Google Scholar] [CrossRef] [Green Version]
  99. Carducci, M.; Shaheen, M.; Markman, B.; Hurvitz, S.; Mahadevan, D.; Kotasek, D.; Goodman, O.B.; Rasmussen, E.; Chow, V.; Juan, G.; et al. A phase 1, first-in-human study of AMG 900, an orally administered pan-Aurora kinase inhibitor, in adult patients with advanced solid tumors. Investig. New Drugs 2018, 36, 1060–1071. [Google Scholar] [CrossRef]
  100. Kantarjian, H.M.; Schuster, M.W.; Jain, N.; Advani, A.; Jabbour, E.; Gamelin, E.; Rasmussen, E.; Juan, G.; Anderson, A.; Chow, V.F.; et al. A phase 1 study of AMG 900, an orally administered pan-aurora kinase inhibitor, in adult patients with acute myeloid leukemia. Am. J. Hematol. 2017, 92, 660–667. [Google Scholar] [CrossRef] [Green Version]
  101. Carpinelli, P.; Ceruti, R.; Giorgini, M.L.; Cappella, P.; Gianellini, L.; Croci, V.; Degrassi, A.; Texido, G.; Rocchetti, M.; Vianello, P.; et al. PHA-739358, a potent inhibitor of Aurora kinases with a selective target inhibition profile relevant to cancer. Mol. Cancer Ther. 2007, 6, 3158–3168. [Google Scholar] [CrossRef] [Green Version]
  102. Meulenbeld, H.J.; Bleuse, J.P.; Vinci, E.M.; Raymond, E.; Vitali, G.; Santoro, A.; Dogliotti, L.; Berardi, R.; Cappuzzo, F.; Tagawa, S.T.; et al. Randomized phase II study of danusertib in patients with metastatic castration-resistant prostate cancer after docetaxel failure. BJU Int. 2013, 111, 44–52. [Google Scholar] [CrossRef]
  103. Oslob, J.D.; Romanowski, M.J.; Allen, D.A.; Baskaran, S.; Bui, M.; Elling, R.A.; Flanagan, W.M.; Fung, A.D.; Hanan, E.J.; Harris, S.; et al. Discovery of a potent and selective aurora kinase inhibitor. Bioorg. Med. Chem Lett. 2008, 18, 4880–4884. [Google Scholar] [CrossRef]
  104. Robert, F.; Verschraegen, C.; Hurwitz, H.; Uronis, H.; Advani, R.; Chen, A.; Taverna, P.; Wollman, M.; Fox, J.; Michelson, G. A phase I trial of sns-314, a novel and selective pan-aurora kinase inhibitor, in advanced solid tumor patients. JCO 2009, 27, 2536. [Google Scholar] [CrossRef]
  105. Farrell, P.; Shi, L.; Matuszkiewicz, J.; Balakrishna, D.; Hoshino, T.; Zhang, L.; Elliott, S.; Fabrey, R.; Lee, B.; Halkowycz, P.; et al. Biological characterization of TAK-901, an investigational, novel, multitargeted Aurora B kinase inhibitor. Mol. Cancer Ther. 2013, 12, 460–470. [Google Scholar] [CrossRef] [Green Version]
  106. Wang, S.; Midgley, C.A.; Scaërou, F.; Grabarek, J.B.; Griffiths, G.; Jackson, W.; Kontopidis, G.; McClue, S.J.; McInnes, C.; Meades, C.; et al. Discovery of N-Phenyl-4-(thiazol-5-yl)pyrimidin-2-amine aurora kinase inhibitors. J. Med. Chem. 2010, 53, 4367–4378. [Google Scholar] [CrossRef] [Green Version]
  107. Adams, N.D.; Adams, J.L.; Burgess, J.L.; Chaudhari, A.M.; Copeland, R.A.; Donatelli, C.A.; Drewry, D.H.; Fisher, K.E.; Hamajima, T.; Hardwicke, M.A.; et al. Discovery of GSK1070916, a potent and selective inhibitor of aurora B/C kinase. J. Med. Chem. 2010, 53, 3973–4001. [Google Scholar] [CrossRef]
  108. Jani, J.P.; Arcari, J.; Bernardo, V.; Bhattacharya, S.K.; Briere, D.; Cohen, B.D.; Coleman, K.; Christensen, J.G.; Emerson, E.O.; Jakowski, A.; et al. PF-03814735, an orally bioavailable small molecule aurora kinase inhibitor for cancer therapy. Mol. Cancer Ther. 2010, 9, 883–894. [Google Scholar] [CrossRef] [Green Version]
  109. Schöffski, P.; Jones, S.F.; Dumez, H.; Infante, J.R.; Van Mieghem, E.; Fowst, C.; Gerletti, P.; Xu, H.; Jakubczak, J.L.; English, P.A.; et al. Phase I, open-label, multicentre, dose-escalation, pharmacokinetic and pharmacodynamic trial of the oral aurora kinase inhibitor PF-03814735 in advanced solid tumours. Eur. J. Cancer 2011, 47, 2256–2264. [Google Scholar] [CrossRef]
  110. Seymour, J.F.; Kim, D.W.; Rubin, E.; Haregewoin, A.; Clark, J.; Watson, P.; Hughes, T.; Dufva, I.; Jimenez, J.L.; Mahon, F.-X.; et al. 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. 2014, 4, e238. [Google Scholar] [CrossRef] [Green Version]
  111. Yang, J.; Ikezoe, T.; Nishioka, C.; Tasaka, T.; Taniguchi, A.; Kuwayama, Y.; Komatsu, N.; Bandobashi, K.; Togitani, K.; Koeffler, H.P.; et al. AZD1152, a novel and selective aurora B kinase inhibitor, induces growth arrest, apoptosis, and sensitization for tubulin depolymerizing agent or topoisomerase II inhibitor in human acute leukemia cells in vitro and in vivo. Blood 2007, 110, 2034–2040. [Google Scholar] [CrossRef] [Green Version]
  112. Schwartz, G.K.; Carvajal, R.D.; Midgley, R.; Rodig, S.J.; Stockman, P.K.; Ataman, O.; Wilson, D.; Das, S.; Shapiro, G.I. Phase i study of barasertib (AZD1152), a selective inhibitor of Aurora B kinase, in patients with advanced solid tumors. Investig. New Drugs 2013, 31, 370–380. [Google Scholar] [CrossRef] [PubMed]
  113. Boss, D.S.; Witteveen, P.O.; van der Sar, J.; Lolkema, M.P.; Voest, E.E.; Stockman, P.K.; Ataman, O.; Wilson, D.; Das, S.; Schellens, J.H. Clinical evaluation of AZD1152, an i.v. inhibitor of Aurora B kinase, in patients with solid malignant tumors. Ann. Oncol. 2011, 22, 431–437. [Google Scholar] [CrossRef] [PubMed]
  114. Löwenberg, B.; Muus, P.; Ossenkoppele, G.; Rousselot, P.; Cahn, J.-Y.; Ifrah, N.; Martinelli, G.; Amadori, S.; Berman, E.; Sonneveld, P.; et al. Phase 1/2 study to assess the safety, efficacy, and pharmacokinetics of barasertib (AZD1152) in patients with advanced acute myeloid leukemia. Blood 2011, 118, 6030–6036. [Google Scholar] [CrossRef] [PubMed]
  115. Tsuboi, K.; Yokozawa, T.; Sakura, T.; Watanabe, T.; Fujisawa, S.; Yamauchi, T.; Uike, N.; Ando, K.; Kihara, R.; Tobinai, K.; et al. A phase I study to assess the safety, pharmacokinetics and efficacy of barasertib (AZD1152), an Aurora B kinase inhibitor, in Japanese patients with advanced acute myeloid leukemia. Leuk. Res. 2011, 35, 1384–1389. [Google Scholar] [CrossRef]
  116. Kantarjian, H.M.; Martinelli, G.; Jabbour, E.J.; Quintás-Cardama, A.; Ando, K.; Bay, J.-O.; Wei, A.; Gröpper, S.; Papayannidis, C.; Owen, K.; et al. 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 2013, 119, 2611–2619. [Google Scholar] [CrossRef]
  117. Dennis, M.; Davies, M.; Oliver, S.; D’Souza, R.; Pike, L.; Stockman, P. Phase i study of the Aurora B kinase inhibitor barasertib (AZD1152) to assess the pharmacokinetics, metabolism and excretion in patients with acute myeloid leukemi. Cancer Chemother. Pharm. 2012, 70, 461–469. [Google Scholar] [CrossRef] [Green Version]
  118. Kantarjian, H.M.; Sekeres, M.A.; Ribrag, V.; Rousselot, P.; Garcia-Manero, G.; Jabbour, E.J.; Owen, K.; Stockman, P.K.; Oliver, S.D. Phase I study assessing the safety and tolerability of barasertib (AZD1152) with low-dose cytosine arabinoside in elderly patients with AML. Clin. Lymphoma Myeloma Leuk. 2013, 13, 559–567. [Google Scholar] [CrossRef] [Green Version]
  119. Collins, G.P.; Eyre, T.A.; Linton, K.M.; Radford, J.; Vallance, G.D.; Soilleux, E.; Hatton, C. A phase II trial of AZD1152 in relapsed/refractory diffuse large B-cell lymphoma. Br. J. Haematol. 2015, 170, 886–890. [Google Scholar] [CrossRef]
  120. Available online: https://opnme.com/molecules/aurb-bi831266 (accessed on 1 July 2021).
  121. Dittrich, C.; Fridrik, M.A.; Koenigsberg, R.; Lee, C.; Goeldner, R.-G.; Hilbert, J.; Greil, R. A phase 1 dose escalation study of BI 831266, an inhibitor of Aurora kinase B, in patients with advanced solid tumors. Investig. New Drugs 2015, 33, 409–422. [Google Scholar] [CrossRef] [Green Version]
  122. Tontsch-Grunt, U.; Gürtler, U.; Zahn, S.K.; Boehmelt, G.; Jarvis, M.; Adolf, G.R.; Solca, F. Abstract 1080: Molecular and cellular pharmacology of BI 811283, a potent inhibitor of Aurora B kinase. Cancer Res. 2010, 70, 1080. [Google Scholar] [CrossRef]
  123. Mross, K.; Richly, H.; Frost, A.; Scharr, D.; Nokay, B.; Graeser, R.; Lee, C.; Hilbert, J.; Goeldner, R.-G.; Fietz, O.; et al. A phase I study of BI 811283, an Aurora B kinase inhibitor, in patients with advanced solid tumors. Cancer Chemother. Pharm. 2016, 78, 405–417. [Google Scholar] [CrossRef] [Green Version]
  124. Döhner, H.; Müller-Tidow, C.; Lübbert, M.; Fiedler, W.; Krämer, A.; Westermann, J.; Bug, G.; Schlenk, R.F.; Krug, U.; Goeldner, R.-G.; et al. A phase I trial investigating the Aurora B kinase inhibitor BI 811283 in combination with cytarabine in patients with acute myeloid leukaemia. Br. J. Haematol. 2019, 185, 583–587. [Google Scholar] [CrossRef]
  125. Zhou, Y.; Shan, S.; Li, Z.; Xin, L.; Pan, D.; Yang, Q.; Liu, Y.; Yue, X.; Liu, X.; Gao, J.; et al. CS2164, a novel multi-target inhibitor against tumor angiogenesis, mitosis and chronic inflammation with anti-tumor potency. Cancer Sci. 2017, 108, 469–477. [Google Scholar] [CrossRef] [Green Version]
  126. Sun, Y.; Yang, L.; Hao, X.; Liu, Y.; Zhang, J.; Ning, Z.; Shi, Y. Phase I dose-escalation study of chiauranib, a novel angiogenic, mitotic, and chronic inflammation inhibitor, in patients with advanced solid tumors. J. Hematol. Oncol. 2019, 12, 9. [Google Scholar] [CrossRef]
  127. Manfredi, M.G.; Ecsedy, J.A.; Chakravarty, A.; Silverman, L.; Zhang, M.; Hoar, K.M.; Stroud, S.G.; Chen, W.; Shinde, V.; Huck, J.J.; et al. Characterization of alisertib (MLN8237), an investigational small-molecule inhibitor of Aurora A kinase using novel in vivo pharmacodynamic assays. Clin. Cancer Res. 2011, 17, 7614–7624. [Google Scholar] [CrossRef] [Green Version]
  128. Dees, E.C.; Cohen, R.B.; von Mehren, M.; Stinchcombe, T.E.; Liu, H.; Venkatakrishnan, K.; Manfredi, M.; Fingert, H.; Burris, H.A.; Infante, J.R. Phase I study of aurora A kinase inhibitor MLN8237 in advanced solid tumors: Safety, pharmacokinetics, pharmacodynamics, and bioavailability of two oral formulations. Clin. Cancer Res. 2012, 18, 4775–4784. [Google Scholar] [CrossRef] [Green Version]
  129. Matulonis, U.A.; Sharma, S.; Ghamande, S.; Gordon, M.S.; Del Prete, S.A.; Ray-Coquard, I.; Kutarska, E.; Liu, H.; Fingert, H.; Zhou, X.; et al. Phase II study of MLN8237 (alisertib), an investigational Aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol. Oncol. 2012, 127, 63–69. [Google Scholar] [CrossRef]
  130. Dickson, M.A.; Mahoney, M.R.; Tap, W.D.; D’Angelo, S.P.; Keohan, M.L.; Van Tine, B.A.; Agulnik, M.; Horvath, L.E.; Nair, J.S.; Schwartz, G.K. Phase II study of MLN8237 (Alisertib) in advanced/metastatic sarcoma. Ann. Oncol. 2016, 27, 1855–1860. [Google Scholar] [CrossRef]
  131. Beltran, H.; Oromendia, C.; Danila, D.C.; Montgomery, B.; Hoimes, C.; Szmulewitz, R.Z.; Vaishampayan, U.; Armstrong, A.J.; Stein, M.; Pinski, J.; et al. A Phase II Trial of the Aurora Kinase a Inhibitor Alisertib for Patients with Castration-resistant and Neuroendocrine Prostate Cancer: Efficacy and Biomarkers. Clin. Cancer Res. 2019, 25, 43–51. [Google Scholar] [CrossRef] [Green Version]
  132. Falchook, G.; Coleman, R.L.; Roszak, A.; Behbakht, K.; Matulonis, U.; Ray-Coquard, I.; Sawrycki, P.; Duska, L.R.; Tew, W.; Ghamande, S.; et al. Alisertib in Combination with Weekly Paclitaxel in Patients With Advanced Breast Cancer or Recurrent Ovarian Cancer: A Randomized Clinical Trial. JAMA Oncol. 2019, 5, e183773. [Google Scholar] [CrossRef] [Green Version]
  133. Necchi, A.; Lo Vullo, S.; Mariani, L.; Raggi, D.; Giannatempo, P.; Calareso, G.; Togliardi, E.; Crippa, F.; Di Genova, N.; Perrone, F.; et al. An open-label, single-arm, phase 2 study of the Aurora kinase a inhibitor alisertib in patients with advanced urothelial cancer. Investig. New Drugs 2016, 34, 236–242. [Google Scholar] [CrossRef]
  134. Owonikoko, T.K.; Niu, H.; Nackaerts, K.; Csoszi, T.; Ostoros, G.; Mark, Z.; Baik, C.; Joy, A.A.; Chouaid, C.; Jaime, J.C.; et al. Randomized Phase II Study of Paclitaxel plus Alisertib versus Paclitaxel plus Placebo as Second-Line Therapy for SCLC: Primary and Correlative Biomarker Analyses. J. Thorac. Oncol. 2020, 15, 274–287. [Google Scholar] [CrossRef] [Green Version]
  135. Graff, J.N.; Higano, C.S.; Hahn, N.M.; Taylor, M.H.; Zhang, B.; Zhou, X.; Venkatakrishnan, K.; Leonard, E.J.; Sarantopoulos, J. Open-label, multicenter, phase 1 study of alisertib (MLN8237), an aurora A kinase inhibitor, with docetaxel in patients with solid tumors. Cancer 2016, 122, 2524–2533. [Google Scholar] [CrossRef]
  136. Haddad, T.C.; D’Assoro, A.; Suman, V.; Opyrchal, M.; Peethambaram, P.; Liu, M.C.; Goetz, M.P.; Ingle, J.N. Phase I trial to evaluate the addition of alisertib to fulvestrant in women with endocrine-resistant, ER+ metastatic breast cancer. Breast Cancer Res. Treat. 2018, 168, 639–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Goff, L.W.; Azad, N.S.; Stein, S.; Whisenant, J.G.; Koyama, T.; Vaishampayan, U.; Hochster, H.; Connolly, R.; Weise, A.; LoRusso, P.M.; et al. Phase I study combining the aurora kinase a inhibitor alisertib with mFOLFOX in gastrointestinal cancer. Investig. New Drugs 2019, 37, 315–322. [Google Scholar] [CrossRef] [PubMed]
  138. DuBois, S.G.; Mosse, Y.P.; Fox, E.; Kudgus, R.A.; Reid, J.M.; McGovern, R.; Groshen, S.; Bagatell, R.; Maris, J.M.; Twist, C.J.; et al. Phase II Trial of Alisertib in Combination with Irinotecan and Temozolomide for Patients with Relapsed or Refractory Neuroblastoma. Clin. Cancer Res. 2018, 24, 6142–6149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Shah, H.A.; Fischer, J.H.; Venepalli, N.K.; Danciu, O.C.; Christian, S.; Russell, M.J.; Liu, L.C.; Zacny, J.P.; Dudek, A.Z. Phase I Study of Aurora A Kinase Inhibitor Alisertib (MLN8237) in Combination With Selective VEGFR Inhibitor Pazopanib for Therapy of Advanced Solid Tumors. Am. J. Clin. Oncol 2019, 42, 413–420. [Google Scholar] [CrossRef] [PubMed]
  140. Kelly, K.R.; Shea, T.C.; Goy, A.; Berdeja, J.G.; Reeder, C.B.; McDonagh, K.T.; Zhou, X.; Danaee, H.; Liu, H.; Ecsedy, J.A.; et al. Phase I study of MLN8237—investigational Aurora A kinase inhibitor—in relapsed/refractory multiple myeloma, non-Hodgkin lymphoma and chronic lymphocytic leukemia. Investig. New Drugs 2014, 32, 489–499. [Google Scholar] [CrossRef] [Green Version]
  141. Barr, P.M.; Li, H.; Spier, C.; Mahadevan, D.; LeBlanc, M.; Ul Haq, M.; Huber, B.D.; Flowers, C.R.; Wagner-Johnston, N.D.; Horwitz, S.M.; et al. Phase II Intergroup Trial of Alisertib in Relapsed and Refractory Peripheral T-Cell Lymphoma and Transformed Mycosis Fungoides: SWOG 1108. J. Clin. Oncol. 2015, 33, 2399–2404. [Google Scholar] [CrossRef]
  142. O’Connor, O.A.; Özcan, M.; Jacobsen, E.D.; Roncero, J.M.; Trotman, J.; Demeter, J.; Masszi, T.; Pereira, J.; Ramchandren, R.; Beaven, A.; et al. Randomized phase III study of alisertib or investigator’s choice (selected single agent) in patients with relapsed or refractory peripheral T-cell lymphoma. J. Clin. Oncol. 2019, 37, 613–623. [Google Scholar] [CrossRef]
  143. Friedberg, J.W.; Mahadevan, D.; Cebula, E.; Persky, D.; Lossos, I.; Agarwal, A.B.; Jung, J.; Burack, R.; Zhou, X.; Leonard, E.J.; et al. Phase II study of alisertib, a selective Aurora A kinase inhibitor, in relapsed and refractory aggressive B- and T-cell non-Hodgkin lymphomas. J. Clin. Oncol. 2014, 32, 44–50. [Google Scholar] [CrossRef]
  144. Kelly, K.R.; Friedberg, J.W.; Park, S.I.; McDonagh, K.; Hayslip, J.; Persky, D.; Ruan, J.; Puvvada, S.; Rosen, P.; Iyer, S.P.; et al. Phase I Study of the Investigational Aurora A Kinase Inhibitor Alisertib plus Rituximab or Rituximab/Vincristine in Relapsed/Refractory Aggressive B-cell Lymphoma. Clin. Cancer Res. 2018, 24, 6150–6159. [Google Scholar] [CrossRef] [Green Version]
  145. Rosenthal, A.; Kumar, S.; Hofmeister, C.; Laubach, J.; Vij, R.; Dueck, A.; Gano, K.; Stewart, A.K. A Phase Ib Study of the combination of the Aurora Kinase Inhibitor Alisertib (MLN8237) and Bortezomib in Relapsed Multiple Myeloma. Br. J. Haematol. 2016, 174, 323–325. [Google Scholar] [CrossRef] [Green Version]
  146. Brunner, A.M.; Blonquist, T.M.; DeAngelo, D.J.; McMasters, M.; Fell, G.; Hermance, N.M.; Winer, E.S.; Lindsley, R.C.; Hobbs, G.S.; Amrein, P.C.; et al. Alisertib plus induction chemotherapy in previously untreated patients with high-risk, acute myeloid leukaemia: A single-arm, phase 2 trial. Lancet Haematol. 2020, 7, e122–e133. [Google Scholar] [CrossRef]
  147. Mossé, Y.P.; Lipsitz, E.; Fox, E.; Teachey, D.T.; Maris, J.M.; Weigel, B.; Adamson, P.C.; Ingle, M.A.; Ahern, C.H.; Blaney, S.M. Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective small-molecule inhibitor of Aurora kinase A: A Children’s Oncology Group Phase I Consortium study. Clin. Cancer Res. 2012, 18, 6058–6064. [Google Scholar] [CrossRef] [Green Version]
  148. Mossé, Y.P.; Fox, E.; Teachey, D.T.; Reid, J.M.; Safgren, S.L.; Carol, H.; Lock, R.B.; Houghton, P.J.; Smith, M.A.; Hall, D.; et al. A Phase II Study of Alisertib in Children with Recurrent/Refractory Solid Tumors or Leukemia: Children’s Oncology Group Phase I and Pilot Consortium (ADVL0921). Clin. Cancer Res. 2019, 25, 3229–3238. [Google Scholar] [CrossRef] [Green Version]
  149. Fletcher, G.C.; Brokx, R.D.; Denny, T.A.; Hembrough, T.A.; Plum, S.M.; Fogler, W.E.; Sidor, C.F.; Bray, M.R. ENMD-2076 is an orally active kinase inhibitor with antiangiogenic and antiproliferative mechanisms of action. Mol. Cancer Ther. 2011, 10, 126–137. [Google Scholar] [CrossRef] [Green Version]
  150. Diamond, J.R.; Bastos, B.R.; Hansen, R.J.; Gustafson, D.L.; Eckhardt, S.G.; Kwak, E.L.; Pandya, S.S.; Fletcher, G.C.; Pitts, T.M.; Kulikowski, G.N.; et al. Phase I safety, pharmacokinetic, and pharmacodynamic study of ENMD-2076, a novel angiogenic and aurora kinase inhibitor, in patients with advanced solid tumors. Clin. Cancer Res. 2011, 17, 849–860. [Google Scholar] [CrossRef] [Green Version]
  151. Matulonis, U.A.; Lee, J.; Lasonde, B.; Tew, W.P.; Yehwalashet, A.; Matei, D.; Behbakht, K.; Grothusen, J.; Fleming, G.; Lee, N.K.; et al. ENMD-2076, an oral inhibitor of angiogenic and proliferation kinases, has activity in recurrent, platinum resistant ovarian cancer. Eur. J. Cancer 2013, 49, 121–131. [Google Scholar] [CrossRef]
  152. Lheureux, S.; Tinker, A.; Clarke, B.; Ghatage, P.; Welch, S.; Weberpals, J.I.; Dhani, N.C.; Butler, M.O.; Tonkin, K.; Tan, Q.; et al. A Clinical and Molecular Phase II Trial of Oral ENMD-2076 in Ovarian Clear Cell Carcinoma (OCCC): A Study of the Princess Margaret Phase II Consortium. Clin. Cancer Res. 2018, 24, 6168–6174. [Google Scholar] [CrossRef] [Green Version]
  153. Veitch, Z.; Zer, A.; Loong, H.; Salah, S.; Masood, M.; Gupta, A.; Bradbury, P.A.; Hogg, D.; Wong, A.; Kandel, R.; et al. A phase II study of ENMD-2076 in advanced soft tissue sarcoma (STS). Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
  154. Diamond, J.R.; Eckhardt, S.G.; Pitts, T.M.; van Bokhoven, A.; Aisner, D.; Gustafson, D.L.; Capasso, A.; Sams, S.; Kabos, P.; Zolman, K.; et al. A phase II clinical trial of the Aurora and angiogenic kinase inhibitor ENMD-2076 for previously treated, advanced, or metastatic triple-negative breast cancer. Breast Cancer Res. 2018, 20, 82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Abou-Alfa, G.K.; Mayer, R.; Venook, A.P.; O’Neill, A.F.; Beg, M.S.; LaQuaglia, M.; Kingham, P.T.; Kobos, R.; Basturk, O.; Brennan, C.; et al. Phase II Multicenter, Open-Label Study of Oral ENMD-2076 for the Treatment of Patients with Advanced Fibrolamellar Carcinoma. Oncologist 2020, 25, e1837–e1845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Yee, K.W.L.; Chen, H.-W.T.; Hedley, D.W.; Chow, S.; Brandwein, J.; Schuh, A.C.; Schimmer, A.D.; Gupta, V.; Sanfelice, D.; Johnson, T.; et al. A phase I trial of the aurora kinase inhibitor, ENMD-2076, in patients with relapsed or refractory acute myeloid leukemia or chronic myelomonocytic leukemia. Investig. New Drugs 2016, 34, 614–624. [Google Scholar] [CrossRef]
  157. Chu, Q.; Bouganim, N.; Fortier, C.; Zaknoen, S.; Stille, J.R.; Kremer, J.D.; Yuen, E.; Hui, Y.-H.; Peña, A.; de la Lithio, A.; et al. Abstract CT083: A Phase I/II study of aurora kinase A inhibitor, LY3295668 erbumine (AK-01): Safety as monotherapy in patients with locally advanced or metastatic solid tumors. Cancer Res. 2019, 79, CT083. [Google Scholar] [CrossRef]
  158. Manfredi, M.G.; Ecsedy, J.A.; Meetze, K.A.; Balani, S.K.; Burenkova, O.; Chen, W.; Galvin, K.M.; Hoar, K.M.; Huck, J.J.; LeRoy, P.J.; et al. Antitumor activity of MLN8054, an orally active small-molecule inhibitor of Aurora A kinase. Proc. Natl. Acad. Sci. USA 2007, 104, 4106–4111. [Google Scholar] [CrossRef] [Green Version]
  159. Dees, E.C.; Infante, J.R.; Cohen, R.B.; O’Neil, B.H.; Jones, S.; von Mehren, M.; Danaee, H.; Lee, Y.; Ecsedy, J.; Manfredi, M.; et al. Phase 1 study of MLN8054, a selective inhibitor of Aurora A kinase in patients with advanced solid tumors. Cancer Chemother. Pharm. 2011, 67, 945–954. [Google Scholar] [CrossRef] [Green Version]
  160. Macarulla, T.; Cervantes, A.; Elez, E.; Rodríguez-Braun, E.; Baselga, J.; Roselló, S.; Sala, G.; Blasco, I.; Danaee, H.; Lee, Y.; et al. Phase I study of the selective Aurora A kinase inhibitor MLN8054 in patients with advanced solid tumors: Safety, pharmacokinetics, and pharmacodynamics. Mol. Cancer Ther. 2010, 9, 2844–2852. [Google Scholar] [CrossRef] [Green Version]
  161. Shimomura, T.; Hasako, S.; Nakatsuru, Y.; Mita, T.; Ichikawa, K.; Kodera, T.; Sakai, T.; Nambu, T.; Miyamoto, M.; Takahashi, I.; et al. MK-5108, a highly selective Aurora-A kinase inhibitor, shows antitumor activity alone and in combination with docetaxel. Mol. Cancer Ther. 2010, 9, 157–166. [Google Scholar] [CrossRef] [Green Version]
  162. Amin, M.; Minton, S.E.; LoRusso, P.M.; Krishnamurthi, S.S.; Pickett, C.A.; Lunceford, J.; Hille, D.; Mauro, D.; Stein, M.N.; Wang-Gillam, A.; et al. A phase I study of MK-5108, an oral aurora a kinase inhibitor, administered both as monotherapy and in combination with docetaxel, in patients with advanced or refractory solid tumors. Investig. New Drugs 2016, 34, 84–95. [Google Scholar] [CrossRef] [Green Version]
  163. Miura, A.; Sootome, H.; Fujita, N.; Suzuki, T.; Fukushima, H.; Mizuarai, S.; Masuko, N.; Ito, K.; Hashimoto, A.; Uto, Y.; et al. TAS-119, a novel selective Aurora A and TRK inhibitor, exhibits antitumor efficacy in preclinical models with deregulated activation of the Myc, β-Catenin, and TRK pathways. Investig. New Drugs 2021. [Google Scholar] [CrossRef]
  164. Robbrecht, D.G.J.; Lopez, J.; Calvo, E.; He, X.; Hiroshi, H.; Soni, N.; Cook, N.; Dowlati, A.; Fasolo, A.; Moreno, V.; et al. A first-in-human phase 1 and pharmacological study of TAS-119, a novel selective Aurora A kinase inhibitor in patients with advanced solid tumours. Br. J. Cancer 2020, 1–8. [Google Scholar] [CrossRef]
  165. Cardin, D.B.; Park, H.; Diamond, J.R.; Drilon, A.E.; VerMeulen, W.L.; He, X.; Hirai, H.; Soni, N.; Berlin, J. Phase I study of the Aurora A kinase (AurA) inhibitor TAS-119 with paclitaxel (P) in advanced solid tumors. JCO 2019, 37, 3031. [Google Scholar] [CrossRef]
  166. Shiotsu, Y.; Kiyoi, H.; Ishikawa, Y.; Tanizaki, R.; Shimizu, M.; Umehara, H.; Ishii, K.; Mori, Y.; Ozeki, K.; Minami, Y.; et al. KW-2449, a novel multikinase inhibitor, suppresses the growth of leukemia cells with FLT3 mutations or T315I-mutated BCR/ABL translocation. Blood 2009, 114, 1607–1617. [Google Scholar] [CrossRef] [Green Version]
  167. Farag, S.; Zhang, S.; Suvannasankha, A.; Liang, J.; O’Bryant, R.; Lisa, W.; Gupta, S.; Bray, M.; Sidor, C.F.; Abonour, R. Clinical Activity of a Novel Multiple Tyrosine Kinase and Aurora Kinase Inhibitor, ENMD-2076, Against Multiple Myeloma: Interim Phase I Trial Results. Blood 2010, 116, 1957. [Google Scholar] [CrossRef]
  168. Warner, S.L.; Munoz, R.M.; Stafford, P.; Koller, E.; Hurley, L.H.; Hoff, D.D.; Von Han, H. Comparing Aurora A and Aurora B as molecular targets for growth inhibition of pancreatic cancer cells. Mol. Cancer Ther. 2006, 5, 2450–2458. [Google Scholar] [CrossRef] [Green Version]
  169. Carpinelli, P.; Moll, J. Aurora kinase inhibitors: Identification and preclinical validation of their biomarkers. Expert Opin. Targets 2008, 12, 69–80. [Google Scholar] [CrossRef]
  170. Ohashi, A.; Ohori, M.; Iwai, K. Motor activity of centromere-associated protein-E contributes to its localization at the center of the midbody to regulate cytokinetic abscission. Oncotarget 2016, 7, 79964–79980. [Google Scholar] [CrossRef] [Green Version]
  171. Yu, K.-W.; Zhong, N.; Xiao, Y.; She, Z.-Y. Mechanisms of kinesin-7 CENP-E in kinetochore-microtubule capture and chromosome alignment during cell division. Biol. Cell 2019, 111, 143–160. [Google Scholar] [CrossRef]
  172. Mao, Y.; Desai, A.; Cleveland, D.W. Microtubule capture by CENP-E silences BubR1-dependent mitotic checkpoint signaling. J. Cell Biol. 2005, 170, 873–880. [Google Scholar] [CrossRef] [Green Version]
  173. Kung, P.-P.; Martinez, R.; Zhu, Z.; Zager, M.; Blasina, A.; Rymer, I.; Hallin, J.; Xu, M.; Carroll, C.; Chionis, J.; et al. Chemogenetic evaluation of the mitotic kinesin CENP-E reveals a critical role in triple-negative breast cancer. Mol. Cancer Ther. 2014, 13, 2104–2115. [Google Scholar] [CrossRef] [Green Version]
  174. Ohashi, A.; Ohori, M.; Iwai, K.; Nambu, T.; Miyamoto, M.; Kawamoto, T.; Okaniwa, M. A novel time-dependent CENP-E inhibitor with potent antitumor activity. PLoS ONE 2015, 10. [Google Scholar] [CrossRef]
  175. Tanudji, M.; Shoemaker, J.; L’Italien, L.; Russell, L.; Chin, G.; Schebye, X.M. Gene silencing of CENP-E by small interfering RNA in HeLa cells leads to missegregation of chromosomes after a mitotic delay. Mol. Biol. Cell 2004, 15, 3771–3781. [Google Scholar] [CrossRef]
  176. Wood, K.W.; Lad, L.; Luo, L.; Qian, X.; Knight, S.D.; Nevins, N.; Brejc, K.; Sutton, D.; Gilmartin, A.G.; Chua, P.R.; et al. Antitumor activity of an allosteric inhibitor of centromere-associated protein-E. Proc. Natl. Acad. Sci. USA 2010, 107, 5839–5844. [Google Scholar] [CrossRef] [Green Version]
  177. Chung, V.; Heath, E.I.; Schelman, W.R.; Johnson, B.M.; Kirby, L.C.; Lynch, K.M.; Botbyl, J.D.; Lampkin, T.A.; Holen, K.D. First-time-in-human study of GSK923295, a novel antimitotic inhibitor of centromere-associated protein e (CENP-E), in patients with refractory cancer. Cancer Chemother. Pharm. 2012, 69, 733–741. [Google Scholar] [CrossRef]
  178. Sawin, K.E.; LeGuellec, K.; Philippe, M.; Mitchison, T.J. Mitotic spindle organization by a plus-end-directed microtubule motor. Nature 1992, 359, 540–543. [Google Scholar] [CrossRef]
  179. Blangy, A.; Lane, H.A.; D’Hérin, P.; Harper, M.; Kress, M.; Nigg, E.A. Phosphorylation by p34cdc2 regulates spindle association of human Eg5, a kinesin-related motor essential for bipolar spindle formation in vivo. Cell 1995, 83, 1159–1169. [Google Scholar] [CrossRef] [Green Version]
  180. Weil, D.; Garçon, L.; Harper, M.; Duménil, D.; Dautry, F.; Kress, M. Targeting the kinesin Eg5 to monitor siRNA transfection in mammalian cells. Biotechniques 2002, 33, 1244–1248. [Google Scholar] [CrossRef]
  181. Nakai, R.; Iida, S.; Takahashi, T.; Tsujita, T.; Okamoto, S.; Takada, C.; Akasaka, K.; Ichikawa, S.; Ishida, H.; Kusaka, H.; et al. K858, a novel inhibitor of mitotic kinesin Eg5 and antitumor agent, induces cell death in cancer cells. Cancer Res. 2009, 69, 3901–3909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Liu, M.; Yu, H.; Huo, L.; Liu, J.; Li, M.; Zhou, J. Validating the mitotic kinesin Eg5 as a therapeutic target in pancreatic cancer cells and tumor xenografts using a specific inhibitor. Biochem. Pharm. Ther. 2008, 76, 169–178. [Google Scholar] [CrossRef]
  183. Liu, L.; Liu, X.; Mare, M.; Dumont, A.S.; Zhang, H.; Yan, D.; Xiong, Z. Overexpression of Eg5 correlates with high grade astrocytic neoplasm. J. Neurooncol 2016, 126, 77–80. [Google Scholar] [CrossRef]
  184. Liu, C.; Zhou, N.; Li, J.; Kong, J.; Guan, X.; Wang, X. Eg5 Overexpression Is Predictive of Poor Prognosis in Hepatocellular Carcinoma Patients. Dis. Markers 2017, 2017. [Google Scholar] [CrossRef] [Green Version]
  185. Ding, S.; Xing, N.; Lu, J.; Zhang, H.; Nishizawa, K.; Liu, S.; Yuan, X.; Qin, Y.; Liu, Y.; Ogawa, O.; et al. Overexpression of Eg5 predicts unfavorable prognosis in non-muscle invasive bladder urothelial carcinoma. Int. J. Urol. 2011, 18, 432–438. [Google Scholar] [CrossRef]
  186. Liu, M.; Wang, X.; Yang, Y.; Li, D.; Ren, H.; Zhu, Q.; Chen, Q.; Han, S.; Hao, J.; Zhou, J. Ectopic expression of the microtubule-dependent motor protein Eg5 promotes pancreatic tumourigenesis. J. Pathol. 2010, 221, 221–228. [Google Scholar] [CrossRef]
  187. Blagden, S.P.; Molife, L.R.; Seebaran, A.; Payne, M.; Reid, A.H.M.; Protheroe, A.S.; Vasist, L.S.; Williams, D.D.; Bowen, C.; Kathman, S.J.; et al. A phase I trial of ispinesib, a kinesin spindle protein inhibitor, with docetaxel in patients with advanced solid tumours. Br. J. Cancer 2008, 98, 894–899. [Google Scholar] [CrossRef] [Green Version]
  188. Rodon, J.; Till, E.; Patnaik, A.; Takimoto, C.; Beeram, M.; Williams, D.; Bowen, C.; Hodge, J.; Dar, M.; Toclher, A. 640 POSTER Phase I study of ispinesib (SB-715992), a kinesin spindle protein inhibitor, in combination with capecitabine in patients with advanced solid tumors. Eur. J. Cancer Suppl. 2006, 4, 193. [Google Scholar] [CrossRef]
  189. Jones, S.F.; Plummer, E.R.; Burris, H.A.; Razak, A.R.; Meluch, A.A.; Bowen, C.J.; Williams, D.H.; Hodge, J.P.; Dar, M.M.; Calvert, A.H. Phase I study of ispinesib in combination with carboplatin in patients with advanced solid tumors. JCO 2006, 24, 2027. [Google Scholar] [CrossRef]
  190. Beer, T.M.; Goldman, B.; Synold, T.W.; Ryan, C.W.; Vasist, L.S.; Van Veldhuizen, P.J.; Dakhil, S.R.; Lara, P.N.; Drelichman, A.; Hussain, M.H.A.; et al. Southwest Oncology Group phase II study of ispinesib in androgen-independent prostate cancer previously treated with taxanes. Clin. Genitourin Cancer 2008, 6, 103–109. [Google Scholar] [CrossRef]
  191. Tang, P.A.; Siu, L.L.; Chen, E.X.; Hotte, S.J.; Chia, S.; Schwarz, J.K.; Pond, G.R.; Johnson, C.; Colevas, A.D.; Synold, T.W.; et al. Phase II study of ispinesib in recurrent or metastatic squamous cell carcinoma of the head and neck. Investig. New Drugs 2008, 26, 257–264. [Google Scholar] [CrossRef]
  192. Lee, C.W.; Bélanger, K.; Rao, S.C.; Petrella, T.M.; Tozer, R.G.; Wood, L.; Savage, K.J.; Eisenhauer, E.A.; Synold, T.W.; Wainman, N.; et al. A phase II study of ispinesib (SB-715992) in patients with metastatic or recurrent malignant melanoma: A National Cancer Institute of Canada Clinical Trials Group trial. Investig. New Drugs 2008, 26, 249–255. [Google Scholar] [CrossRef]
  193. Knox, J.J.; Gill, S.; Synold, T.W.; Biagi, J.J.; Major, P.; Feld, R.; Cripps, C.; Wainman, N.; Eisenhauer, E.; Seymour, L. A phase II and pharmacokinetic study of SB-715992, in patients with metastatic hepatocellular carcinoma: A study of the National Cancer Institute of Canada Clinical Trials Group (NCIC CTG IND.168). Investig. New Drugs 2008, 26, 265–272. [Google Scholar] [CrossRef]
  194. Lee, R.T.; Beekman, K.E.; Hussain, M.; Davis, N.B.; Clark, J.I.; Thomas, S.P.; Nichols, K.F.; Stadler, W.M. A University of Chicago consortium phase II trial of SB-715992 in advanced renal cell cance. Clin. Genitourin. Cancer 2008, 6, 21–24. [Google Scholar] [CrossRef] [Green Version]
  195. Shahin, M.S.; Braly, P.; Rose, P.; Malpass, T.; Bailey, H.; Alvarez, R.D.; Hodge, J.; Bowen, C.; Buller, R. A phase II, open-label study of ispinesib (SB-715992) in patients with platinum/taxane refractory or resistant relapsed ovarian cancer. JCO 2007, 25, 5562. [Google Scholar] [CrossRef]
  196. Gomez, H.L.; Philco, M.; Pimentel, P.; Kiyan, M.; Monsalvo, M.L.; Conlan, M.G.; Saikali, K.G.; Chen, M.M.; Seroogy, J.J.; Wolff, A.A.; et al. Phase I dose-escalation and pharmacokinetic study of ispinesib, a kinesin spindle protein inhibitor, administered on days 1 and 15 of a 28-day schedule in patients with no prior treatment for advanced breast cancer. Anticancer Drugs 2012, 23, 335–341. [Google Scholar] [CrossRef]
  197. Souid, A.-K.; Dubowy, R.L.; Ingle, A.M.; Conlan, M.G.; Sun, J.; Blaney, S.M.; Adamson, P.C. A pediatric phase I trial and pharmacokinetic study of ispinesib: A Children’s Oncology Group phase I consortium study. Pediatr. Blood Cancer 2010, 55, 1323–1328. [Google Scholar] [CrossRef] [Green Version]
  198. Jackson, J.R.; Gilmartin, A.; Dhanak, D.; Knight, S.; Parrish, C.; Luo, L.; Sutton, D.; Caulder, E.; Diamond, M.; Giardiniere, M.; et al. A second generation KSP inhibitor, SB-743921, is a highly potent and active therapeutic in preclinical models of cancer. Clin. Cancer Res. 2006, 12, B11. [Google Scholar]
  199. Holen, K.D.; Belani, C.P.; Wilding, G.; Ramalingam, S.; Volkman, J.L.; Ramanathan, R.K.; Vasist, L.S.; Bowen, C.J.; Hodge, J.P.; Dar, M.M.; et al. A first in human study of SB-743921, a kinesin spindle protein inhibitor, to determine pharmacokinetics, biologic effects and establish a recommended phase II dose. Cancer Chemother. Pharm. 2011, 67, 447–454. [Google Scholar] [CrossRef] [Green Version]
  200. O’Connor, O.A.; Gerecitano, J.; Van Deventer, H.; Hainsworth, J.; Zullo, K.M.; Saikali, K.; Seroogy, J.; Wolff, A.; Escandón, R. The addition of granulocyte-colony stimulating factor shifts the dose limiting toxicity and markedly increases the maximum tolerated dose and activity of the kinesin spindle protein inhibitor SB-743921 in patients with relapsed or refractory lymphoma: R. Leuk. Lymphoma 2015, 56, 2585–2591. [Google Scholar] [CrossRef]
  201. Lemieux, C.; Dewolf, W.; Voegtli, W.; Delisle, R.; Laird, E.; Wallace, E.; Woessner, R.; Corrette, C.; Allen, S.; Hans, J.; et al. ARRY-520, a novel, highly selective KSP inhibitor with potent anti-proliferative activity. Proc. Am. Assoc. Cancer Res. 2007, 48, 5590. [Google Scholar]
  202. LoRusso, P.M.; Goncalves, P.H.; Casetta, L.; Carter, J.A.; Litwiler, K.; Roseberry, D.; Rush, S.; Schreiber, J.; Simmons, H.M.; Ptaszynski, M.; et al. First-in-human phase 1 study of filanesib (ARRY-520), a kinesin spindle protein inhibitor, in patients with advanced solid tumors. Investig. New Drugs 2015, 33, 440–449. [Google Scholar] [CrossRef]
  203. Khoury, H.J.; Garcia-Manero, G.; Borthakur, G.; Kadia, T.; Foudray, M.C.; Arellano, M.; Langston, A.; Bethelmie-Bryan, B.; Rush, S.; Litwiler, K.; et al. A phase 1 dose-escalation study of ARRY-520, a kinesin spindle protein inhibitor, in patients with advanced myeloid leukemias. Cancer 2012, 118, 3556–3564. [Google Scholar] [CrossRef] [Green Version]
  204. Shah, J.J.; Kaufman, J.L.; Zonder, J.A.; Cohen, A.D.; Bensinger, W.I.; Hilder, B.; Rush, S.; Walker, D.; Tunquist, B.; Litwiler, K.; et al. A Phase 1/2 study of filanesib alone and in combination with low-dose dexamethasone in relapsed/refractory multiple myeloma. Cancer 2017, 123, 4617–4630. [Google Scholar] [CrossRef]
  205. Chari, A.; Htut, M.; Zonder, J.A.; Fay, J.W.; Jakubowiak, A.J.; Levy, J.B.; Lau, K.; Burt, S.M.; Tunquist, B.J.; Hilder, B.W.; et al. A phase 1 dose-escalation study of filanesib plus bortezomib and dexamethasone in patients with recurrent/refractory multiple myeloma. Cancer 2016, 122, 3327–3335. [Google Scholar] [CrossRef]
  206. Lee, H.C.; Shah, J.J.; Feng, L.; Manasanch, E.E.; Lu, R.; Morphey, A.; Crumpton, B.; Patel, K.K.; Wang, M.L.; Alexanian, R.; et al. A phase 1 study of filanesib, carfilzomib, and dexamethasone in patients with relapsed and/or refractory multiple myeloma. Blood Cancer J. 2019, 9, 80. [Google Scholar] [CrossRef] [Green Version]
  207. Ocio, E.M.; Motlló, C.; Rodríguez-Otero, P.; Martínez-López, J.; Cejalvo, M.J.; Martín-Sánchez, J.; Bladé, J.; García-Malo, M.D.; Dourdil, M.V.; García-Mateo, A.; et al. Filanesib in combination with pomalidomide and dexamethasone in refractory MM patients: Safety and efficacy, and association with alpha 1-acid glycoprotein (AAG) levels. Phase Ib/II Pomdefil clinical trial conducted by the Spanish MM group. Br. J. Haematol. 2021, 192, 522–530. [Google Scholar] [CrossRef]
  208. Toudjarska, I.; Judge, A.; Buck, T.; McClintock, K.; Jong, S.; de Ambegia, E.; Brodsky, J.; Akinc, A.; Racie, T.; Jeffs, L.; et al. Abstract B204: Development of ALN-VSP: An RNAi therapeutic for liver malignancies. Mol. Cancer Ther. 2009, 8, B204. [Google Scholar] [CrossRef] [Green Version]
  209. Tabernero, J.; Shapiro, G.I.; LoRusso, P.M.; Cervantes, A.; Schwartz, G.K.; Weiss, G.J.; Paz-Ares, L.; Cho, D.C.; Infante, J.R.; Alsina, M.; et al. First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement. Cancer Discov. 2013, 3, 406–417. [Google Scholar] [CrossRef] [Green Version]
  210. Ye, X.S.; Fan, L.; Van Horn, R.D.; Nakai, R.; Ohta, Y.; Akinaga, S.; Murakata, C.; Yamashita, Y.; Yin, T.; Credille, K.M.; et al. A novel Eg5 inhibitor (LY2523355) causes mitotic arrest and apoptosis in cancer cells and shows potent antitumor activity in xenograft tumor models. Mol. Cancer Ther. 2015, 14, 2463–2472. [Google Scholar] [CrossRef] [Green Version]
  211. Infante, J.R.; Patnaik, A.; Verschraegen, C.F.; Olszanski, A.J.; Shaheen, M.; Burris, H.A.; Tolcher, A.W.; Papadopoulos, K.P.; Beeram, M.; Hynes, S.M.; et al. Two Phase 1 dose-escalation studies exploring multiple regimens of litronesib (LY2523355), an Eg5 inhibitor, in patients with advanced cancer. Cancer Chemother. Pharm. 2017, 79, 315–326. [Google Scholar] [CrossRef]
  212. Wakui, H.; Yamamoto, N.; Kitazono, S.; Mizugaki, H.; Nakamichi, S.; Fujiwara, Y.; Nokihara, H.; Yamada, Y.; Suzuki, K.; Kanda, H.; et al. A phase 1 and dose-finding study of LY2523355 (litronesib), an Eg5 inhibitor, in Japanese patients with advanced solid tumors. Cancer Chemother. Pharm. 2014, 74, 15–23. [Google Scholar] [CrossRef]
  213. Schiemann, K.; Finsinger, D.; Zenke, F.; Amendt, C.; Knöchel, T.; Bruge, D.; Buchstaller, H.-P.; Emde, U.; Stähle, W.; Anzali, S. The discovery and optimization of hexahydro-2H-pyrano[3,2-c]quinolines (HHPQs) as potent and selective inhibitors of the mitotic kinesin-5. Bioorg. Med. Chem. Lett. 2010, 20, 1491–1495. [Google Scholar] [CrossRef]
  214. Hollebecque, A.; Deutsch, E.; Massard, C.; Gomez-Roca, C.; Bahleda, R.; Ribrag, V.; Bourgier, C.; Lazar, V.; Lacroix, L.; Gazzah, A.; et al. A phase I, dose-escalation study of the Eg5-inhibitor EMD 534085 in patients with advanced solid tumors or lymphoma. Investig. New Drugs 2013, 31, 1530–1538. [Google Scholar] [CrossRef]
  215. Mross, K.B.; Scharr, D.; Richly, H.; Frost, A.; Bauer, S.; Krauss, B.; Krauss, R.; Mais, A.; Hauns, B.; Hentsch, B.; et al. First-in-human study of 4SC-205 (AEGIS), a novel oral inhibitor of Eg5 kinesin spindle protein. JCO 2014, 32, 2564. [Google Scholar] [CrossRef]
  216. Theoclitou, M.-E.; Aquila, B.; Block, M.H.; Brassil, P.J.; Castriotta, L.; Code, E.; Collins, M.P.; Davies, A.M.; Deegan, T.; Ezhuthachan, J.; et al. Discovery of (+)-N-(3-aminopropyl)-N-[1-(5-benzyl-3-methyl-4-oxo-[1,2]thiazolo[5,4-d]pyrimidin-6-yl)-2-methylpropyl]-4-methylbenzamide (AZD4877), a kinesin spindle protein inhibitor and potential anticancer agent. J. Med. Chem. 2011, 54, 6734–6750. [Google Scholar] [CrossRef]
  217. Esaki, T.; Seto, T.; Ariyama, H.; Arita, S.; Fujimoto, C.; Tsukasa, K.; Kometani, T.; Nosaki, K.; Hirai, F.; Yagawa, K. Phase I study to assess the safety, Tolerability and pharmacokinetics of AZD4877 in japanese patients with solid tumors. Arch. Drug Inf. 2011, 4, 23–31. [Google Scholar] [CrossRef] [Green Version]
  218. Infante, J.R.; Kurzrock, R.; Spratlin, J.; Burris, H.A.; Eckhardt, S.G.; Li, J.; Wu, K.; Skolnik, J.M.; Hylander-Gans, L.; Osmukhina, A.; et al. A Phase I study to assess the safety, tolerability, and pharmacokinetics of AZD4877, an intravenous Eg5 inhibitor in patients with advanced solid tumors. Cancer Chemother. Pharm. 2012, 69, 165–172. [Google Scholar] [CrossRef]
  219. Jones, R.; Vuky, J.; Elliott, T.; Mead, G.; Arranz, J.A.; Chester, J.; Chowdhury, S.; Dudek, A.Z.; Müller-Mattheis, V.; Grimm, M.-O.; et al. Phase II study to assess the efficacy, safety and tolerability of the mitotic spindle kinesin inhibitor AZD4877 in patients with recurrent advanced urothelial cancer. Investig. New Drugs 2013, 31, 1001–1007. [Google Scholar] [CrossRef]
  220. Kantarjian, H.M.; Padmanabhan, S.; Stock, W.; Tallman, M.S.; Curt, G.A.; Li, J.; Osmukhina, A.; Wu, K.; Huszar, D.; Borthukar, G.; et al. Phase I/II multicenter study to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of AZD4877 in patients with refractory acute myeloid leukemia. Investig. New Drugs 2012, 30, 1107–1115. [Google Scholar] [CrossRef] [Green Version]
  221. Jeay, S.; Ali, S.; Chen, C.-R.; Uppalapati, U.; Senator, D.; Chan, T.; France, D.; Ashwell, M. Discovery of a novel Eg5 kinesin inhibitor, ARQ 621, with potent antitumor activity while sparing bone marrow-derived cells. Cancer Res. 2008, 68, 656. [Google Scholar]
  222. Chen, L.C.; Rosen, L.S.; Iyengar, T.; Goldman, J.W.; Savage, R.; Kazakin, J.; Chan, T.C.K.; Schwartz, B.E.; Abbadessa, G.; Von Hoff, D.D. First-in-human study with ARQ 621, a novel inhibitor of Eg5: Final results from the solid tumors cohort. JCO 2011, 29, 3076. [Google Scholar] [CrossRef]
  223. Cox, C.D.; Garbaccio, R.M. Discovery of allosteric inhibitors of kinesin spindle protein (KSP) for the treatment of taxane-refractory cancer: MK-0731 and analogs. Anticancer Agents Med. Chem. 2010, 10, 697–712. [Google Scholar] [CrossRef] [PubMed]
  224. Holen, K.; DiPaola, R.; Liu, G.; Tan, A.R.; Wilding, G.; Hsu, K.; Agrawal, N.; Chen, C.; Xue, L.; Rosenberg, E.; et al. A phase I trial of MK-0731, a kinesin spindle protein (KSP) inhibitor, in patients with solid tumors. Investig. New Drugs 2012, 30, 1088–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Gerecitano, J.F.; Stephenson, J.J.; Lewis, N.L.; Osmukhina, A.; Li, J.; Wu, K.; You, Z.; Huszar, D.; Skolnik, J.M.; Schwartz, G.K. A Phase I trial of the kinesin spindle protein (Eg5) inhibitor AZD4877 in patients with solid and lymphoid malignancies. Investig. New Drugs 2013, 31, 355–362. [Google Scholar] [CrossRef] [PubMed]
  226. Mitchison, T.J. The proliferation rate paradox in antimitotic chemotherapy. Mol. Biol. Cell 2012, 23, 1–6. [Google Scholar] [CrossRef] [PubMed]
  227. Bennett, A.; Sloss, O.; Topham, C.; Nelson, L.; Tighe, A.; Taylor, S.S. Inhibition of Bcl-xL sensitizes cells to mitotic blockers, but not mitotic drivers. Open Biol. 2016, 6, 160134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Gascoigne, K.E.; Taylor, S.S. Cancer Cells Display Profound Intra- and Interline Variation following Prolonged Exposure to Antimitotic Drugs. Cancer Cell 2008, 14, 111–122. [Google Scholar] [CrossRef] [Green Version]
  229. Fitzgerald, K.A.; Malhotra, M.; Curtin, C.M.; O’ Brien, F.J.; O’ Driscoll, C.M. Life in 3D is never flat: 3D models to optimise drug delivery. J. Control. Release 2015, 215, 39–54. [Google Scholar] [CrossRef]
  230. Pinto, B.; Henriques, A.C.; Silva, P.M.A.; Bousbaa, H. Three-Dimensional Spheroids as In vitro Preclinical Models for Cancer Research. Pharmaceutics 2020, 12, 1186. [Google Scholar] [CrossRef]
  231. Sgorla, D.; Bunhak, É.J.; Cavalcanti, O.A.; Fonte, P.; Sarmento, B. Exploitation of lipid-polymeric matrices at nanoscale for drug delivery applications. Expert Opin. Drug. Deliv. 2016, 13, 1301–1309. [Google Scholar] [CrossRef]
  232. Lv, J.-F.; Hu, L.; Zhuo, W.; Zhang, C.-M.; Zhou, H.-H.; Fan, L. Epigenetic alternations and cancer chemotherapy response. Cancer Chemother. Pharm. 2016, 77, 673–684. [Google Scholar] [CrossRef]
  233. Mountzios, G.; Dimopoulos, M.-A.; Soria, J.-C.; Sanoudou, D.; Papadimitriou, C.A. Histopathologic and genetic alterations as predictors of response to treatment and survival in lung cancer: A review of published data. Crit. Rev. Oncol. Hematol. 2010, 75, 94–109. [Google Scholar] [CrossRef]
Figure 1. Targeting mitosis for cancer treatment. (a,b) Representation of G2 interphase and the stages of mitosis. A description of the main cellular changes at each stage is presented. The progression throughout mitosis is monitored by the spindle assembly checkpoint activity (SAC ON and SAC OFF). (c) Activity of mitotic proteins during G2 and mitotic phases. MPS1, Aurora B, and PLK1 kinases are involved in several processes, being activated from G2 of interphase to telophase/cytokinesis. Aurora A and Eg-5 proteins ensure proper bipolar spindle shape, remaining activated from G2 to metaphase. CENP-E is required for accurate kinetochore–microtubule attachments, operating from prometaphase to metaphase. (d) SAC modulation by targeting mitotic proteins. MPS1 and Aurora B inhibition leads to SAC override, followed by massive chromosome missegregation and cell death. PLK1, Aurora A and Eg-5 inhibition induces spindle defects, while CENP-E inhibition promotes chromosome misalignment, leading to SAC activation, which in turn arrests cells in mitosis. Under mitotic arrest, the cell undergoes death or alternative pathways, namely G1 arrest/senescence, or continues cycling. (e) Second-generation antimitotics in clinical trials. Summary of antimitotic drug targets in different phases of clinical trials and current status. Created in BioRender.
Figure 1. Targeting mitosis for cancer treatment. (a,b) Representation of G2 interphase and the stages of mitosis. A description of the main cellular changes at each stage is presented. The progression throughout mitosis is monitored by the spindle assembly checkpoint activity (SAC ON and SAC OFF). (c) Activity of mitotic proteins during G2 and mitotic phases. MPS1, Aurora B, and PLK1 kinases are involved in several processes, being activated from G2 of interphase to telophase/cytokinesis. Aurora A and Eg-5 proteins ensure proper bipolar spindle shape, remaining activated from G2 to metaphase. CENP-E is required for accurate kinetochore–microtubule attachments, operating from prometaphase to metaphase. (d) SAC modulation by targeting mitotic proteins. MPS1 and Aurora B inhibition leads to SAC override, followed by massive chromosome missegregation and cell death. PLK1, Aurora A and Eg-5 inhibition induces spindle defects, while CENP-E inhibition promotes chromosome misalignment, leading to SAC activation, which in turn arrests cells in mitosis. Under mitotic arrest, the cell undergoes death or alternative pathways, namely G1 arrest/senescence, or continues cycling. (e) Second-generation antimitotics in clinical trials. Summary of antimitotic drug targets in different phases of clinical trials and current status. Created in BioRender.
Pharmaceutics 13 01011 g001
Figure 2. Spindle assembly checkpoint mechanism. In response to unattached or improperly attached kinetochores (Prometaphase), the SAC is turned ON and promotes the assembly of the mitotic checkpoint complex (MCC), made of Mad2, Bub3, BubR1 and Cdc20. At these kinetochores, MPS1 recruits Bub3, Bub1 and BubR1. The MCC inhibits the activity of anaphase-promoting complex/cyclosome (APC/C), leading to the stabilization of separase/securin and CDK1/cyclin B complexes, and consequent mitotic arrest. The Aurora B kinase (Aur B), associated with centromere heterochromatin, promotes proper kinetochore–microtubule attachments. Once all chromosomes are properly attached to spindle microtubules and are aligned at the metaphase plate (metaphase), the SAC is turned OFF, through MCC disassembly, and consequently Cdc20 can bind and activates the APC/C, resulting in the ubiquitylation (ub) of cyclin B and securin mitotic substracts. In turn, separase can cleave cohesins to promote sister chromatid separation (anaphase), while Cdk1 inactivation promotes exit from mitosis. Created in BioRender.
Figure 2. Spindle assembly checkpoint mechanism. In response to unattached or improperly attached kinetochores (Prometaphase), the SAC is turned ON and promotes the assembly of the mitotic checkpoint complex (MCC), made of Mad2, Bub3, BubR1 and Cdc20. At these kinetochores, MPS1 recruits Bub3, Bub1 and BubR1. The MCC inhibits the activity of anaphase-promoting complex/cyclosome (APC/C), leading to the stabilization of separase/securin and CDK1/cyclin B complexes, and consequent mitotic arrest. The Aurora B kinase (Aur B), associated with centromere heterochromatin, promotes proper kinetochore–microtubule attachments. Once all chromosomes are properly attached to spindle microtubules and are aligned at the metaphase plate (metaphase), the SAC is turned OFF, through MCC disassembly, and consequently Cdc20 can bind and activates the APC/C, resulting in the ubiquitylation (ub) of cyclin B and securin mitotic substracts. In turn, separase can cleave cohesins to promote sister chromatid separation (anaphase), while Cdk1 inactivation promotes exit from mitosis. Created in BioRender.
Pharmaceutics 13 01011 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Novais, P.; Silva, P.M.A.; Amorim, I.; Bousbaa, H. Second-Generation Antimitotics in Cancer Clinical Trials. Pharmaceutics 2021, 13, 1011. https://doi.org/10.3390/pharmaceutics13071011

AMA Style

Novais P, Silva PMA, Amorim I, Bousbaa H. Second-Generation Antimitotics in Cancer Clinical Trials. Pharmaceutics. 2021; 13(7):1011. https://doi.org/10.3390/pharmaceutics13071011

Chicago/Turabian Style

Novais, Pedro, Patrícia M. A. Silva, Isabel Amorim, and Hassan Bousbaa. 2021. "Second-Generation Antimitotics in Cancer Clinical Trials" Pharmaceutics 13, no. 7: 1011. https://doi.org/10.3390/pharmaceutics13071011

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop