Next Article in Journal
The Oxidative Fermentation of Ethanol in Gluconacetobacter diazotrophicus Is a Two-Step Pathway Catalyzed by a Single Enzyme: Alcohol-Aldehyde Dehydrogenase (ADHa)
Next Article in Special Issue
Expression and Diagnostic Value of HE4 in Pancreatic Adenocarcinoma
Previous Article in Journal
Mechanisms of Hyperhomocysteinemia Induced Skeletal Muscle Myopathy after Ischemia in the CBS/+ Mouse Model
Previous Article in Special Issue
Gene Mutation Analysis in EGFR Wild Type NSCLC Responsive to Erlotinib: Are There Features to Guide Patient Selection?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular Targeting of the Oncoprotein PLK1 in Pediatric Acute Myeloid Leukemia: RO3280, a Novel PLK1 Inhibitor, Induces Apoptosis in Leukemia Cells

by Na-Na Wang 1,†, Zhi-Heng Li 1,†, He Zhao 1,†, Yan-Fang Tao 1, Li-Xiao Xu 1, Jun Lu 1, Lan Cao 1, Xiao-Juan Du 2, Li-Chao Sun 3, Wen-Li Zhao 1, Pei-Fang Xiao 1, Fang Fang 1, Guang-Hao Su 1, Yan-Hong Li 1, Gang Li 1, Yi-Ping Li 1, Yun-Yun Xu 1, Hui-Ting Zhou 1, Yi Wu 1, Mei-Fang Jin 1, Lin Liu 1, Jian Ni 4, Jian Wang 1, Shao-Yan Hu 1, Xue-Ming Zhu 1, Xing Feng 1,* and Jian Pan 1,*add Show full author list remove Hide full author list
1
Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou 215003, China
2
Department of Gastroenterology, the 5th Hospital of Chinese People's Liberation Army (PLA), Yinchuan 750000, China
3
Department of Cell and Molecular Biology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
4
Translational Research Center, Second Hospital, The Second Clinical School, Nanjing Medical University, Nanjing 210000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2015, 16(1), 1266-1292; https://doi.org/10.3390/ijms16011266
Submission received: 15 October 2014 / Accepted: 29 December 2014 / Published: 7 January 2015

Abstract

:
Polo-like kinase 1 (PLK1) is highly expressed in many cancers and therefore a biomarker of transformation and potential target for the development of cancer-specific small molecule drugs. RO3280 was recently identified as a novel PLK1 inhibitor; however its therapeutic effects in leukemia treatment are still unknown. We found that the PLK1 protein was highly expressed in leukemia cell lines as well as 73.3% (11/15) of pediatric acute myeloid leukemia (AML) samples. PLK1 mRNA expression was significantly higher in AML samples compared with control samples (82.95 ± 110.28 vs. 6.36 ± 6.35; p < 0.001). Kaplan-Meier survival analysis revealed that shorter survival time correlated with high tumor PLK1 expression (p = 0.002). The 50% inhibitory concentration (IC50) of RO3280 for acute leukemia cells was between 74 and 797 nM. The IC50 of RO3280 in primary acute lymphocytic leukemia (ALL) and AML cells was between 35.49 and 110.76 nM and 52.80 and 147.50 nM, respectively. RO3280 induced apoptosis and cell cycle disorder in leukemia cells. RO3280 treatment regulated several apoptosis-associated genes. The regulation of DCC, CDKN1A, BTK, and SOCS2 was verified by western blot. These results provide insights into the potential use of RO3280 for AML therapy; however, the underlying mechanisms remain to be determined.

1. Introduction

Acute leukemia is a family of serious medical conditions relating with an original diagnosis of leukemia. Acute leukemia is known as cancer of the blood which can cause death if not medicated [1]. Multiple small-molecule kinase inhibitors are currently being developed for leukemia treatment [2,3,4,5]. For example, selective FLT3 (fms-related tyrosine kinase 3) inhibitors, such as AC220 (quizartinib), have been proven clinically effective in acute myeloid leukemia (AML) patients with internal tandem duplications (ITD) in the gene of FLT3 [6,7]. Other inhibitors in development target mediators of downstream signaling pathways, such as mTOR [8] and MEK [9], or cell cycle machinery, such as aurora kinases [3,10] or cyclin-dependent kinases [11]. Recently, the polo-like kinase 1 (PLK1) inhibitor has showed promising effects in the treatment of acute leukemia. Polo-like kinases (PLKs) are a set of regulators that control mitotic progression [12]. PLK1, the best-characterized mammalian PLK, is a particularly attractive target for cancer drug development because most cancers require its activity [12,13,14,15].
PLK1 is a serine/threonine-protein kinase expressed during mitosis and overexpressed in multiple cancers, including acute leukemia [13]. PLK1 expression is increased in several types of cancers including urothelial carcinoma of bladder [16], renal cancer [17], breast cancer [18], prostate cancer [19], neuroblastoma [20], hepatocellular carcinoma [21], cervical carcinoma [22], and non-melanoma skin cancers [23]. PLK1 is also highly expressed in leukemic cell lines and over expressed in a majority of samples from patients with acute myeloid leukemia compared with normal progenitors [24]. And PLK1 knockdown by small interfering RNA also blocked proliferation of leukemic cell lines and the clonogenic potential of primary cells from patients [24]. PLK1 inhibition is a promising strategy for the treatment of AML. Furthermore, the expression of PLK1 in untransformed cells is much lower, which makes PLK1 a suitable target for the development of cancer-specific small molecule drugs. Supporting evidence of oncogenic properties of PLK1 comes from the overexpression studies of PLK1 in an NIH3T3 cell line [25,26]. These cells become capable of forming foci in soft agar due to PLK1 overexpression [27]. Depletion or inhibition of kinase activity of PLK1 is sufficient to induce cell-cycle arrest and apoptosis in cancer cell lines and in xenograft tumor models [28,29,30,31].
Inhibitors of PLK1 have been developed for potential human cancer therapy. NMS-P937 is a novel PLK1-specific inhibitor and shows high potency in proliferation inhibition on a large number of cell lines [28]. Another PLK1 inhibitor, BI2536, was tested against a panel of nine ALL cell lines at nanomolar concentrations. BI2536 strongly diminished colony formation capacity and increased apoptosis rates of these cells [32]. Co-treatment with BI2536 and vorinostat synergistically induced cell death in parental or imatinib mesylate-resistant BCR/ABL+ cells and primary CD34+ bone marrow cells but was minimally toxic to normal cells [33]. TAK-960 is another novel PLK1 inhibitor that has shown activity in several tumor cell lines, including those cells highly expressed multidrug-resistant protein 1 (MDR1) [29]. GW843682X is also a novel selective PLK1 inhibitor which caused accumulation of cells in the G2/M phase and mediated apoptosis of human leukemia cells [34].
Wovkulich and colleagues [30] have developed a novel PLK1 inhibitor, RO3280; RO3280 has potent inhibitory activity against PLK1, good selectivity against other kinases, and excellent in vitro cellular potency. However, the molecular function of this drug in leukemia is still unknown [30]. In the present study, RO3280 has been evaluated to further characterize its preclinical antitumor efficacy, and the molecular mechanism of action was explored with real-time PCR arrays.

2. Results and Discussion

2.1. Expression of PLK1 Is Upregulated in AML Cells and Pediatric AML Patients

As reported previously, PLK1 is highly expressed in a broad set of cancer cell lines and overexpressed in a majority of cancer patient samples compared with normal progenitor cells. However, the expression of PLK1 in AML, and specifically pediatric AML, has not been clearly defined. We demonstrate that the expression of PLK1 is very high in AML cell lines, with the highest levels observed in CCRF, NB4, and K562 cells (Figure 1A). To examine the expression of PLK1 in pediatric AML samples, we obtained samples from 15 patients with pediatric AML and 12 control patients. High protein expression of PLK1 was observed in 73.3% (11/15) of the pediatric AML samples compared to 0% (0/12) of the normal bone marrow (NBM) control samples (Figure 1B). Real-time PCR was also used to examine the mRNA transcript levels of PLK1 in 105 pediatric AML samples and 30 NBM/ITP (idiopathic thrombocytopenic purpura) (control samples (Figure 1C)). PLK1 expression was significantly higher in the AML samples compared to the control samples (82.95 ± 110.28 vs. 6.36 ± 6.35; p < 0.001). Bone marrow specimens were obtained from 105 pediatric patients with AML at the time of diagnosis, who presented at Children’s Hospital of Soochow University between 2000 and 2011. We suppose the high SD (standard deviation) values are related to the cDNA quality of samples. Examination of pediatric AML patient clinicopathology revealed that expression of PLK1 is related with FAB (French-American-Britain) and MRD (Minimal Residual Disease, Table 1). However, there were no significant differences in other clinical features such as sex, age, initial hemoglobin level, white blood cell counts, platelet counts, or chromosomal abnormalities between individuals with high and low PLK1 expression (Table 1). The prognostic significance of PLK1 expression was assessed in 105 Chinese pediatric AML patients with clinical follow-up records. Kaplan-Meier survival analysis revealed shorter survival times for patients with high PLK1 expression in tumors (p = 0.002, Table 2 and Figure 1C). Furthermore, multivariate analysis revealed that PLK1 expression is an independent prognostic factor in pediatric AML (p = 0.041, Table 3). In summary, our results demonstrate that PLK1 expression is heightened in patients with pediatric AML and in human myeloid leukemia cell lines. This indicates that PLK1 may be a suitable oncogene target for pediatric AML therapy.
Figure 1. Expression of PLK1 is upregulated in AML cells and pediatric AML patients (A) Western blot analysis showing PLK1 protein expression in nine leukemia cell lines; (B) Western blot analysis showing PLK1 protein expression in 15 pediatric AML samples and 12 NBM samples; (C) Real-time PCR analysis of the PLK1 mRNA transcript levels in 105 pediatric AML samples and 30 NBM/ITP (normal bone marrow/idiopathic thrombocytopenic purpura) control samples; and (D) Kaplan-Meier survival analysis of 105 pediatric AML patients comparing high and low PLK1 expression (p = 0.002).
Figure 1. Expression of PLK1 is upregulated in AML cells and pediatric AML patients (A) Western blot analysis showing PLK1 protein expression in nine leukemia cell lines; (B) Western blot analysis showing PLK1 protein expression in 15 pediatric AML samples and 12 NBM samples; (C) Real-time PCR analysis of the PLK1 mRNA transcript levels in 105 pediatric AML samples and 30 NBM/ITP (normal bone marrow/idiopathic thrombocytopenic purpura) control samples; and (D) Kaplan-Meier survival analysis of 105 pediatric AML patients comparing high and low PLK1 expression (p = 0.002).
Ijms 16 01266 g001aIjms 16 01266 g001b
Table 1. Association of polo-like kinase 1 (PLK1) expression with clinico-pathological characteristics in 105 pediatric acute myeloid leukemia (AML) samples.
Table 1. Association of polo-like kinase 1 (PLK1) expression with clinico-pathological characteristics in 105 pediatric acute myeloid leukemia (AML) samples.
Clinical VariablesNo. of PatientsPLK1 Expression (n)p
LowHigh
Gender
Male4227150.373
Female633528
Age (year)
<66035250.864
≥6452718
Leukocyte (/μL)
>10,0006137240.693
≤10,000442519
FAB
M1–M69359340.011
M71239
Cytogenetics
Favorable5028220.160
Intermediate27207
Unfavorable281414
MRD
<0.25%4936130.005
≥0.25%562630
FAB: French-American-Britain; MRD: Minimal Residual Disease.
Table 2. Association of PLK1 expression with Kaplan-Meier survival in 105 pediatric AML samples.
Table 2. Association of PLK1 expression with Kaplan-Meier survival in 105 pediatric AML samples.
VariableNo. of PatientsOver Survivalp
Median ± SE
Cytogenetics
Favorable5046.664 ± 3.717<0.001
Intermediate2729.220 ± 3.188
Unfavorable2811.161 ± 1.827
FAB
M1–M69336.113 ± 2.885<0.001
M7128.542 ± 1.820
Leukocyte (/μL)
>10,0006130.220 ± 2.9740.803
≤10,0004433.631 ± 4.063
MRD
<0.25%4953.627 ± 3.151<0.001
≥0.25%5618.893 ± 2.425
PLK1 expression
Low (<12.420)6239.319 ± 3.5390.002
High (≥12.420)4324.054 ± 3.709
Table 3. Cox multivariate analysis of PLK1 expression and clinico-pathological features in pediatric AML.
Table 3. Cox multivariate analysis of PLK1 expression and clinico-pathological features in pediatric AML.
VariableOdds RatioEXP (B) 95% CIp
Cytogenetics
Favorable vs. Intermediate and Unfavorable6.1642.477 (1.210–5.068)0.013
MRD
<0.25% vs. ≥0.25%14.0845.176 (2.193–12.214)0.000
Leukocyte (/uL)
>10,000 vs. ≤10,0000.2001.138 (0.646–2.055)0.655
FAB classification
M7 vs. M1–M67.1482.683 (1.301–5.533)0.008
PLK1 Expression
Low vs. High4.1951.806 (1.026–3.179)0.041
EXP (B) 95% CI: 95% CI (confidence intervals) of relative risk.

2.2. RO3280 Inhibits the Growth of Acute Leukemia Cells

The novel PLK1 inhibitor RO3280 decreased leukemia cell viability in a dose-dependent manner (Figure 2A,B). The RO3280 IC50 measurement was determined in several acute leukemia cell lines: U937 186 nM, HL60 175 nM, NB4 74 nM, K562 797 nM, MV4-11 120 nM, and CCRF 162 nM. RO3280 treatment could also dramatically impact cell morphology as observed in NB4 cells (Figure 2C).
In order to better understand the effective of RO3280, we compared it with other PLK1 inhibitors: Rigosertib (ON 01910. Na) and BI2536. The IC50 of these PLK1 inhibitors was analyzed in both NB4 and K562 cells (Figure 2D). In NB4 cells the following IC50 concentrations were determined: RO3280 13.45 nM, ON 01910. Na 13.02 nM, and BI2536 87.65 nM. In K562 cells the following IC50 concentrations were determined: RO3280 301 nM, ON 01910. Na 1606 nM, and BI2536 448 nM. To determine the effectiveness of RO3280 in primary leukemia, we determined the IC50 in ALL and AML cells. In primary ALL the IC50 of RO3280 is 35.49–110.76 nM (Figure 2E, Table 4) and in primary AML the IC50 of RO3280 is 52.80–147.50 nM (Figure 2E, Table 5). These results demonstrate that the PLK1 inhibitor RO3280 effectively inhibits the proliferation of leukemia cells.

2.3. RO3280 Induced Apoptosis and Cell Cycle Disorder in Leukemia Cells

To determine if RO3280 induces apoptosis in leukemia cells, we assessed Annexin V staining, cell cycle disorder and caspase activation in leukemia cells after treatment. Cells treated with RO3280 for 24 h showed higher Annexin V staining compared with untreated cells (Figure 3). This indicates that RO3280 induces apoptosis in leukemia cells. Cell cycle disorder was determined by a cell cycle assay, which showed that RO3280 significantly induces cell cycle disorder in nine leukemia cell lines (Figure 4). Hoechst 33342 staining showed that 24 h RO3280 treatment induced DNA fragmentation and the formation of abnormal nuclear cells (Figure 5A). When either HL-60 or NB4 cells were treated with RO3280, there was a significant increase in abnormal nuclear cell formation compared with control DMSO treated cells (Figure 5B). Moreover, to clearly demonstrate that RO3280 causes apoptosis in leukemia cells, we assessed the following well-recognized markers of apoptosis: PARP, caspase-3, and caspase-9. After 24 h of treatment with RO3280 (50 or 100 nM), cleaved PARP and caspase-9 were observed in NB4 and HL-60 cells (Figure 5C). These results are consistent with the Annexin V staining and cell cycle analysis, demonstrating that RO3280 induces apoptosis in leukemia cells. This further suggests that RO3280 may have promising antitumor therapeutic applications.
Figure 2. RO3280 inhibits the growth of acute leukemia cells. (A) Molecular structure of RO3280; (B) Viability and IC50 analysis of RO3280 in six leukemia cells. The following are the determined RO3280 IC50s: U937 186 nM, HL60 175 nM, NB4 74 nM, K562 797 nM, MV4-11 120 nM and CCRF 162 nM; (C) Micrographs were taken of NB4 cells treated with RO3280 100 nM or DMSO (scale bar = 50 μm); (D) The IC50 of three PLK1 inhibitors, RO3280, ON 01910.Na and BI2536, was analyzed in NB4 and K562 cells. IC50 in NB4 cells: RO3280 13.45 nM, ON 01910. Na 13.02 nM, and BI2536 87.65 nM. IC50 in K562 cells: RO3280 301 nM, ON 01910. Na 1606 nM, and BI2536 is 448 nM; and (E) The IC50 of RO3280 in primary acute lymphocytic leukemia (ALL) and AML cells was also analyzed: ALL 35.49–110.76 nM and AML 52.80–147.50 nM. Cell proliferation was calculated as a percentage of the DMSO treated control wells. The IC50 values were derived after plotting proliferation values on a logarithmic curve. Experiments were performed in quadruplicate and repeated twice.
Figure 2. RO3280 inhibits the growth of acute leukemia cells. (A) Molecular structure of RO3280; (B) Viability and IC50 analysis of RO3280 in six leukemia cells. The following are the determined RO3280 IC50s: U937 186 nM, HL60 175 nM, NB4 74 nM, K562 797 nM, MV4-11 120 nM and CCRF 162 nM; (C) Micrographs were taken of NB4 cells treated with RO3280 100 nM or DMSO (scale bar = 50 μm); (D) The IC50 of three PLK1 inhibitors, RO3280, ON 01910.Na and BI2536, was analyzed in NB4 and K562 cells. IC50 in NB4 cells: RO3280 13.45 nM, ON 01910. Na 13.02 nM, and BI2536 87.65 nM. IC50 in K562 cells: RO3280 301 nM, ON 01910. Na 1606 nM, and BI2536 is 448 nM; and (E) The IC50 of RO3280 in primary acute lymphocytic leukemia (ALL) and AML cells was also analyzed: ALL 35.49–110.76 nM and AML 52.80–147.50 nM. Cell proliferation was calculated as a percentage of the DMSO treated control wells. The IC50 values were derived after plotting proliferation values on a logarithmic curve. Experiments were performed in quadruplicate and repeated twice.
Ijms 16 01266 g002aIjms 16 01266 g002b
Table 4. Pathologic features and inhibition of cell growth by RO3280 in primary culture cells of pediatric ALL.
Table 4. Pathologic features and inhibition of cell growth by RO3280 in primary culture cells of pediatric ALL.
NumberGenderAge (Years)DiagnosisALL TypingChromosome AnalysisFusion GenePrednisone SensitivityPLK1IC50 (nM)
1Female5ALLB46, XYNot detectedSensitivepositive35.39
2Male4ALLB46, XYTEL/AML1+Sensitivepositive30.79
3Male3ALLBALL/53–54, XY, +4, +6, +10, 12p+, +14, +17, +18, +20, +21Not detectedSensitivepositive64.43
4Female4ALLB46, XXNot detectedSensitivepositive8.27
5Female4ALLBALL/53–55, XX, +X, 1q+, +4, +6, +10, +11, +15, +17, +21Not detectedSensitivepositive36.01
Table 5. Pathologic features and inhibition of cell growth by RO3280 in primary culture cells of pediatric AML.
Table 5. Pathologic features and inhibition of cell growth by RO3280 in primary culture cells of pediatric AML.
NumberGenderAge (Years)DiagnosisAML TypingChromosome AnalysisFusion GenePLK1IC50 nM
1Female9AMLM446, XXFLT3-ITDpositive88.65
2Female3AMLM446, XX, inv (16) (p13;q22)CBF/MYH11positive47.02
3Male4AMLM5b46, XY, −2,+10, t (10;10) (p13;q23)MLL/AF10positive28.57
4Male12AMLM2a45, X, −Y, t (8;21) (q22;q22)AML/ETOpositive32.92
5Female1AMLM446, XX, inv (16) (p13;q22)Not detectedpositive19.20
Figure 3. Annexin V analysis of apoptosis induced by RO3280 in acute leukemia cells. Annexin V staining of cells following a 24 h treatment with RO3280 at 50 or 100 nM compared with DMSO control mock treatment. All these analyses were repeated three times. ** p < 0.01.
Figure 3. Annexin V analysis of apoptosis induced by RO3280 in acute leukemia cells. Annexin V staining of cells following a 24 h treatment with RO3280 at 50 or 100 nM compared with DMSO control mock treatment. All these analyses were repeated three times. ** p < 0.01.
Ijms 16 01266 g003aIjms 16 01266 g003b
Figure 4. Cell cycle analysis of RO3280 induced cell cycle disorder in acute leukemia cells. Cell cycle analysis of nine leukemia cells treated for 24 h with RO3280 at 50 or 100 nM compared with DMSO control mock treatment. G2 phase of each group was analyzed and presented. All these analyses were repeated three times. * p < 0.05; ** p < 0.01. Left purple peak mean the G1 phase and right purple peak mean the G2 phase, red peak means the S phase; Pink, yellow and green peak mean the disorder of cell cycle.
Figure 4. Cell cycle analysis of RO3280 induced cell cycle disorder in acute leukemia cells. Cell cycle analysis of nine leukemia cells treated for 24 h with RO3280 at 50 or 100 nM compared with DMSO control mock treatment. G2 phase of each group was analyzed and presented. All these analyses were repeated three times. * p < 0.05; ** p < 0.01. Left purple peak mean the G1 phase and right purple peak mean the G2 phase, red peak means the S phase; Pink, yellow and green peak mean the disorder of cell cycle.
Ijms 16 01266 g004aIjms 16 01266 g004b
Figure 5. RO3280 induced DNA fragmentation and cleavage of apoptotic markers in acute leukemia cells. (A) Micrographs following Hoechst 33342 staining of cells treated with RO3280 (50 and 100 nM) for 24 h. This demonstrates the induction of DNA fragmentation and abnormal nuclear cell formation; (B) The abnormal nuclear cells were quantified and increased significantly with RO3280 treatment compared with mock treatment in both HL-60 and NB4 cells. ** p < 0.01; and (C) Western blot analysis of PARP, caspase-3, and caspase-9. After 24 h treatment with 50 or 100 nM RO3280, cleaved PARP and caspase-9 were detected in lysates from NB4 and HL-60 cells.
Figure 5. RO3280 induced DNA fragmentation and cleavage of apoptotic markers in acute leukemia cells. (A) Micrographs following Hoechst 33342 staining of cells treated with RO3280 (50 and 100 nM) for 24 h. This demonstrates the induction of DNA fragmentation and abnormal nuclear cell formation; (B) The abnormal nuclear cells were quantified and increased significantly with RO3280 treatment compared with mock treatment in both HL-60 and NB4 cells. ** p < 0.01; and (C) Western blot analysis of PARP, caspase-3, and caspase-9. After 24 h treatment with 50 or 100 nM RO3280, cleaved PARP and caspase-9 were detected in lysates from NB4 and HL-60 cells.
Ijms 16 01266 g005aIjms 16 01266 g005b

2.4. Real-Time PCR Array Identifies Genes Implicated in the Effect of RO3280 Treatment

In order to identify apoptosis and/or programmed cell death molecules implicated in the effects of RO3280, we used the SABioscience Human Apoptosis PCR Array PAHS-3012. With this real-time PCR array we analyzed and clustered the expression of 370 genes associated with apoptosis in DMSO or RO3280 treated cells (Figure 6A). The genes most significantly upregulated or downregulated are shown in Figure 6B,C, respectively. Examination of the array data revealed that 32 genes were significantly upregulated and 16 genes were significantly downregulated in RO3280 treatment group compared with DMSO control group (Table 6 and Table 7, respectively). The upregulated genes included protein kinase C zeta, receptor-interacting serine-threonine kinase 3, harakiri BCL2 interacting protein, DCC netrin 1 receptor, and cyclin-dependent kinase inhibitor 1A. The downregulated genes included interleukin 1α, nucleotide-binding oligomerization domain containing 2, caspase-1, apoptosis-related cysteine peptidase, serpin peptidase inhibitor clade B, B-cell CLL/lymphoma 2, and Bruton agammaglobulinemia tyrosine kinase. The RO3280-dependent upregulation of DCC and CDKN1A and downregulation of BTK and SOCS2 was verified by western blot analysis (Figure 7).
Figure 6. Real-time PCR array identifies genes implicated in the effect of RO3280 treatment. (A) Gene expression clustering of 370 key apoptosis genes in 50 nM RO3280-treated NB4 cells compared to DMSO-treated cells; (B) Relative expression of the most upregulated genes in RO3280-treated NB4 cells compared to DMSO-treated cells; and (C) Relative expression of the most downregulated genes in RO3280-treated NB4 cells compared to DMSO-treated cells.
Figure 6. Real-time PCR array identifies genes implicated in the effect of RO3280 treatment. (A) Gene expression clustering of 370 key apoptosis genes in 50 nM RO3280-treated NB4 cells compared to DMSO-treated cells; (B) Relative expression of the most upregulated genes in RO3280-treated NB4 cells compared to DMSO-treated cells; and (C) Relative expression of the most downregulated genes in RO3280-treated NB4 cells compared to DMSO-treated cells.
Ijms 16 01266 g006aIjms 16 01266 g006b
Figure 7. Western blot verification of real-time PCR array results. Western blot analysis of cells following a 24 h treatment with RO3280 at 50 or 100 nM compared with DMSO control mock treatment. The upregulation of DCC and CDKN1A and down regulation of BTK and SOCS2 in RO3280 treated cells. Protein lysates from treated cells were tested for expression levels by Western blot analysis.
Figure 7. Western blot verification of real-time PCR array results. Western blot analysis of cells following a 24 h treatment with RO3280 at 50 or 100 nM compared with DMSO control mock treatment. The upregulation of DCC and CDKN1A and down regulation of BTK and SOCS2 in RO3280 treated cells. Protein lysates from treated cells were tested for expression levels by Western blot analysis.
Ijms 16 01266 g007
Table 6. Genes upregulated in NB4 cells treated with RO3280 compared with the DMSO control group.
Table 6. Genes upregulated in NB4 cells treated with RO3280 compared with the DMSO control group.
GeneDescriptionDMSORO3280Fold Changep Value
GDNFglial cell derived neurotrophic factor0.00299.41723262.86320.0162
PRKCZprotein kinase C, zeta0.004212.76793065.98480.0457
RIPK3receptor-interacting serine-threonine kinase 30.00254.52491802.24330.0126
HRKharakiri, BCL2 interacting protein0.1471150.65031023.92420.0246
GRM4glutamate receptor, metabotropic 40.3625335.0571924.18890.0225
TDGF1teratocarcinoma-derived growth factor 10.045127.3203605.16730.0218
DCCDCC netrin 1 receptor3.4002425.7855125.22470.0225
BDNFbrain-derived neurotrophic factor44.17391023.202323.16300.0007
EEF1A2eukaryotic translation elongation factor 1 α 2633,377.635412,905,456.296820.37560.0083
NUPR1nuclear protein, transcriptional regulator, 111.4562232.134320.26290.0001
CIDECcell death-inducing DFFA-like effector c33.4482667.698119.96210.0187
CDKN1Acyclin-dependent kinase inhibitor 1A2113.736236,484.299717.26060.0113
BNIP-2BCL2/adenovirus E1B 19 kDa interacting protein 218.2415306.748316.81600.0145
BIRC5baculoviral IAP repeat containing 560,340.7297981,719.564816.26960.0164
TIMP3TIMP metallopeptidase inhibitor 315.1670201.909013.31240.0031
CASP14caspase 14, apoptosis-related cysteine peptidase32.1570410.516612.76600.0226
CARD10caspase recruitment domain family, member 1028,245.9365329,683.720911.67190.0292
ALOX15Barachidonate 15-lipoxygenase, type B68.4589796.173511.62990.0488
APOEapolipoprotein E411.81144632.265411.24850.0119
BCL3B-cell CLL/lymphoma 3950.20977763.83368.17070.0013
BNIP3LBCL2/adenovirus E1B 19 kDa interacting protein 3-like 16,818.9831130,854.42877.78020.0001
DAPK1death-associated protein kinase 140.6424287.79477.08110.0050
AIFM2apoptosis-inducing factor, mitochondrion-associated, 21759.571812,454.04717.07790.0156
BCL6B-cell CLL/lymphoma 6385.75592574.12016.67290.0061
AIFM3apoptosis-inducing factor, mitochondrion-associated, 3764.96985057.62176.61150.0500
ZNF443zinc finger protein 4432001.533312,744.64206.36740.0009
DAPK3death-associated protein kinase 35607.952830,206.62195.38640.0020
BCL2L11BCL2-like 11 (apoptosis facilitator)7094.660934,581.36504.87430.0000
BIRC-2baculoviral IAP repeat containing 28366.595435,580.91214.25270.0007
BAG3BCL2-associated athanogene 310,487.792334,964.82593.33390.0002
Bcl-10B-cell CLL/lymphoma 1013,170.601336,171.78652.74640.0000
BFARbifunctional apoptosis regulator14,398.643431,271.85622.17190.0243
Table 7. Genes downregulated in NB4 cells treated with RO3280 compared with the DMSO control group.
Table 7. Genes downregulated in NB4 cells treated with RO3280 compared with the DMSO control group.
GeneDescriptionDMSORO3280Fold Changep Value
IL1Ainterleukin 1α39.87030.00000.00000.0316
NOD2nucleotide-binding oligomerization domain containing 2135.50590.00000.00000.0000
CASP1caspase 1, apoptosis-related cysteine peptidase161.06680.00110.00000.0001
NME5NME/NM23 family member 5 26.46500.00060.00000.0000
SERPINB2serpin peptidase inhibitor, clade B168.24870.25040.00150.001
SOCS2suppressor of cytokine signaling 2150.34832.50120.01660.0006
MPOmyeloperoxidase1,652,217.420442,599.17760.02580.0000
TNFSF15tumor necrosis factor superfamily, member 1515.20091.28030.08420.0360
Bcl2A1BCL2-related protein A1619.9195152.66330.24630.0019
Bcl-2B-cell CLL/lymphoma 231,688.95168210.59050.25910.0026
BTKBruton’s agammaglobulinemia tyrosine kinase 37,873.98169837.20220.25970.0001
IL31RAinterleukin 31 receptor A823.6783230.44150.27980.0009
PROK2prokineticin 213,280.37334801.48890.36150.0004
AZU1azurocidin 129,361.934210,755.37690.36630.0003
LTBRlymphotoxin β receptor 6702.95362753.68370.41080.0004
HSPA9heat shock 70 kDa protein 9318,280.1754141,258.38870.44380.0000
Serine/threonine-protein kinase PLK1, also known as polo-like kinase 1 (PLK-1) or serine/threonine-protein kinase 13 (STPK13). PLK1 is an early trigger for G2/M transition. PLK1 supports the functional maturation of the centrosome in late G2/early prophase and establishment of the bipolar spindle. PLK1 is an interesting molecule both as a biomarker and as a target for highly specific cancer therapy for several reasons. Firstly, it is over-expressed in many cancers and can serve as a biomarker. PLK1 is highly expressed in a broad range of tumors. Secondly, the expression of PLK1 in untransformed cells is not nearly as high, which makes it a discriminating target for the development of cancer-specific small molecule drugs. Medical experts are now paying more attention to the role of PLK1 in acute myeloid leukemia [35]. Here we show that PLK1 is highly expressed in all AML cell lines tested and in 73.3% (11/15) of the pediatric AML samples compared to 0% (0/12) of the NBM control sample. Real-time PCR analysis of 105 pediatric AML samples demonstrated significantly upregulated PLK1 expression in the AML samples compared to the control samples. Sometimes the results of the RT-PCR were not very accurate when the cDNA quality of samples was not very high. Thus, it is best to utilize Western blot analysis in future studies. Kaplan-Meier survival analysis of 105 pediatric AML patients revealed shorter survival time correlated with high PLK1 expression in tumors. Furthermore, multivariate analysis revealed that PLK1 expression is an independent prognostic factor in pediatric AML. In summary, our results are indicate for the first time that PLK1 may be a good oncogene target in samples from pediatric AML patients and human myeloid leukemia cell lines.
Several PLK1 inhibitors have been discovered and show significant anticancer effects. ATP-competitive inhibitor BI 6727 [36,37] is currently in clinical trials for cancer treatment. PLK1 inhibition with BI2536 leads to apoptotic effects in SCCHN cell lines [38]. BI2536 is a potent PLK1 inhibitor with IC50 of 0.83 nM. BI2536 also shows 4- and 11-fold greater selectivity against PLK2 and PLK3, and its effects in leukemia have been reported by others [32]. TAK-960 is currently undergoing Phase I evaluation in adult patients with advanced solid malignancies [39]. ON 01910. Na inhibits mitotic progression and induces apoptosis in most cancer cell lines [40], is currently in a phase I study for treating adult patients with advanced solid malignancies [41]. Rigosertib (ON-01910) is a PLK1 inhibitor with IC50 of 9 nM. As presented here, in leukemia cells, RO3280 is more effective than Rigosertib (ON-01910). Wovkulich and colleagues recently developed RO3280, a novel PLK1 inhibitor [30]. RO3280 is a potent, highly selective inhibitor of PLK1 with IC50 of 3 nM. Until now, we know that PLK1 is a target of RO3280 and RO3280 has nearly no effect on PLK2 and PLK3. However, the effectiveness of RO3280 in leukemia treatment remains an open question. In this report we demonstrate that RO3280 inhibits leukemia cell proliferation in a dose-dependent manner and causes apoptosis. This suggests that RO3280 may be a promising antitumor treatment for leukemia. Indeed the potential of PLK1 inhibitors in treatment is promising, and combinations of PLK1 inhibitors with other anticancer drugs might offer a larger range of opportunities for cancer therapy [42].
Real-time PCR Array System is ideal tool for analyzing the expression of a focused panel of genes [43]. In this study, we used real-time PCR arrays to analyze the effect of RO3280 treatment on apoptotic gene regulation. Compared to an untreated control group, 32 genes related to apoptosis were significantly upregulated and 16 genes were significantly downregulated after RO3280 treatment. This confirmed previous reports that CDKN1A, CASP1, and BCL2 are regulated by PLK1 inhibition. PLK1 is a checkpoint protein whose role spans all of mitosis and includes DNA repair. PLK1 has been reported as a negative regulator of CDKN1A (p21) [44]. When cancer cells are treated with PLK1 inhibitors, p21 is increased in the cytoplasm. By contrast, deficiency of p21 renders tumor cells more susceptible to Polo-like kinase 1 inhibition [45]. A rrecent study showed that PLK1 inhibition causes post-mitotic DNA damage and senescence in a range of human tumor cell lines [46]. Small-molecule PLK1 inhibitors and PLK1 genetic knock-down resulted in the induction of DNA double-strand breaks and senescence in a range of human tumor cell lines. Molecular function of PLK1 is very complicated and in metastatic alveolar rhabdomyosarcoma (aRMS), PLK1 interacted with and phosphorylated PAX3-FOXO1 leading to protein stabilization. PLK1 inhibition led to elevated ubiquitination and rapid proteasomal degradation of this PAX3-FOXO1 chimeric oncoprotein [47]. Our study also identified some novel genes such as RIPK3, HRK, DCC, and BTK regulated by PLK1 inhibition. While each of these genes has a function related to apoptosis, many are additionally involved in separate cellular processes. RIPK3 is a component of the tumor necrosis factor (TNF) receptor-I signaling complex and a key regulator of TNF-induced necrosis [48]. The DCC gene encodes a netrin 1 receptor that partially localizes to lipid rafts and induces apoptosis in the absence of ligand. The receptor functions as a tumor suppressor and is frequently mutated or down regulated in colorectal [49] and ovarian cancers [50]. HRK promotes apoptosis in mammalian cells by interacting with the apoptotic inhibitors BCL-2 and BCL-XL via its BH3 domain [51]. Recent research indicates that PLK1 is an important regulator of PTEN during cell cycle progression [52]. Our research now indicates that PLK1 may also regulate RIPK3, DCC, and HRK expression. Until now, the regulation mechanism of PLK1 is still unclear. Thus, our results may provide new clues into the molecular mechanism of apoptosis induced by RO3280 in leukemia cells.

3. Experimental Section

3.1. Cell and Culture Conditions

Leukemia cell lines HL-60, MV4-11, U937, DAMI and K562 were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). CCRF, Raji, Jurkat, 697 and SHI-1 cell lines (gifts from Wang Jian-Rong, The Cyrus Tang Hematology center of Soochow University, Suzhou, China). All cell lines were maintained at 37 °C in RPMI 1640 (Gibco, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (Invitrogen, Carlsbad, CA, USA). RO3280 (Cat: S7248; Selleck Chemicals, West Paterson, NJ, USA) was dissolved in DMSO (Cat: D4540; Sigma-Aldrich, St. Louis, MO, USA).

3.2. Patients and Samples

Bone marrow specimens were obtained from 105 pediatric patients with AML at the time of diagnosis, who presented at Children’s Hospital of Soochow University between 2000 and 2011. Ethical approval was provided by the Children’s Hospital of Soochow University Ethics Committee (Nos. SUEC2000-021 and SUEC2011-037), and written informed consent was obtained from the parents or guardians. AML diagnosis was made in accordance with the revised French-American-British (FAB) classification. Patients’ clinical and laboratory features are summarized in Table 1. Additionally, 23 healthy donors’ bone marrow samples and seven patients with ITP were analyzed as controls. Bone marrow mononuclear cells (BMNCs) from patients were isolated within 2 h using Ficoll solution when bone marrow samples were harvested.

3.3. CD34+ Cell Purification

For CD34+ cell selection, the Miltenyi immunoaffinity device (VarioMACS 130-046-703) was used according to the manufacturer’s instructions (Miltenyi Biotech, Auburn, CA, USA). Briefly, the CD34+ cells are magnetically labeled with CD34 MicroBeads. Then, the cell suspension is loaded onto a MACSR column which is placed in the magnetic field of a MACS Separator. The magnetically labeled CD34+ cells are retained within the column. The unlabeled cells run through; CD34+ cells were adsorbed on the magnetic poles. After removing the column from the magnetic field, the magnetically retained CD34+ cells can be eluted as the positively selected cell fraction.

3.4. Quantitative Reverse-Transcription PCR for PLK1

Quantitative real-time PCR was performed to determine the expression levels of PLK1 genes. Total RNA was reverse transcribed using the Reverse Transcription Kit, according to the manufacturer’s protocol (Applied Biosystems Inc., Foster City, CA, USA). The real time PCR primers used to quantify GAPDH expression were: Forward (F): 5'-AGAAGGCTGGGGCTCATTTG-3' and Reverse (R): 5'-AGGGGCCATCCACAGTCTTC-3' and for PLK1 were: F: 5'-AGTCGACCACCTCACCTGTC-3' and R: 5'-GCCCCTCACAGTCCTCAATA-3'. Expression of PLK1 was normalized to endogenous GAPDH expression. Real-time PCR array analysis was according to the MIQE Guidelines [53] and performed in a total volume of 20 µL including 1 µL of cDNA, primers (0.2 mM each) and 10 µL of SYBR Green mix (Roche, Basel, Switzerland). Reactions were run on a Lightcycler 480 (Roche) using universal thermal cycling parameters (95 °C for 5 min, 45 cycles of 10 s at 95 °C, 20 s at 60 °C and 15 s at 72 °C; followed by a melting curve: 10 s at 95 °C, 60 s at 60 °C and continued melting). The results were obtained using the sequence detection software of the Lightcycler 480 and analyzed using Microsoft Excel. For quality control purposes, melting curves were acquired for all samples. The comparative Ct method was used to quantify gene expression. The target gene expression level was normalized to expression of the housekeeping gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH) within the same sample (−ΔCt), and then the relative expression of each gene was calculated using Log 2 (−ΔCt).

3.5. Cell Proliferation and Viability Assay

Leukemia cells or primary leukemia cells (2 × 104) were seeded in 96-well plates overnight and incubated with DMSO, or increasing concentrations of RO3280 (0.05–120 μM) for 24 h. The same volume of DMSO added to the vehicle treated wells. Each drug concentration was replicated four times. Then, 10 μL CCK8 (Dojindo Molecular Technologies, Tokyo, Japan) solution was added to each well, incubated at 37 °C for 2–4 h and the optical density (OD) values were measured at 450 nm using a scanning multi-well spectrophotometer (BioRad Model 550, Hercules, CA, USA). Relative survival rate was calculated from the absorbance values compared with the control group. The proliferation of cells was calculated as a percentage of the DMSO-treated control wells with 50% inhibitory concentration (IC50) values derived after plotting proliferation values on a logarithmic curve. The IC50 of PLK1 inhibitor was calculated by Graph Prism software (GraphPad-Prism Software Inc., San Diego, CA, USA).

3.6. Cell Cycle Analysis

Firstly, leukemia cells were collected and washed with PBS for 5 min by centrifugation at 125× g. Then, cells were fixed with paraformaldehyde and transparented with 0.5% Triton X-100 for 10 min. After that, cells were resuspended and incubated for 30 min at 37 °C in staining solution containing 1.5 μmol/L propidium iodide (P4170; Sigma-Aldrich, St. Louis, MO, USA) and 25 μg/mL RNase A The samples (1 × 104 cells) were analyzed with a Beckman Gallios™ Flow Cytometer (Beckman, Krefeld, Germany).

3.7. Apoptosis Assay

Apoptosis assay was according to the manual operation of BD Annexin V Staining Kit (Cat: 556420; BD Biosciences, Franklin Lakes, NJ, USA). Briefly, cells were washed two times with cold PBS and then cells were resuspended in 1× Binding Buffer at a concentration of ~1 × 106 cells/mL. Then 100 µL of the solution (~1 × 105 cells) was transferred to a 5 mL culture tube. Annexin V and PI (Propidium Iodide) 5 µL/test was added. Cells were gently mixed and incubated for 15 min at RT in the dark. 400 µL of 1× Binding Buffer was added to each tube and the cells were analyzed by flow cytometry within one hour.

3.8. Western Blot Analysis

Cellular proteins were extracted in 40 mM Tris-HCl (pH 7.4) containing 150 mM NaCl and 1% (v/v) Triton X-100, supplemented with protease inhibitors for western blot analysis. Equal amounts of protein were resolved on 12% SDS-PAGE gels, and then transferred to a PVDF membrane (Millipore, Bedford, MA, USA). Blots were blocked and probed with antibodies against Caspase 3 (Cat: 9661S, 1:1000; Cell Signaling Technology Inc., Danvers, MA, USA), Caspase 9 (Cat: 4501S 1:1000, Cell Signaling Technology Inc.), PARP (Cat: 9542S, 1:1000; Cell Signaling Technology Inc.), PLK1 (Cat: 4535S, 1:1000; Cell Signaling Technology Inc.), RIPK3 (Cat: 13526S,1:1000; Cell Signaling Technology Inc.), HRK (Cat: ab45419, 1:1000; Abcam Trading (Shanghai) Company Ltd., Shanghai, China), DCC (Cat: 3858S, 1:1000; Cell Signaling Technology Inc.), CDKN1A(Cat: 2947S, 1:1000; Cell Signaling Technology Inc.), CASP1 (Cat: 2225S, 1:1000; Cell Signaling Technology Inc.), BCL2 (Cat: ab7973, 1:1000; Abcam Trading (Shanghai) Company Ltd.), BTK(Cat: 3533S, 1:1000; Cell Signaling Technology Inc.), SOCS2 (Cat: 2779S, 1:1000; Cell Signaling Technology Inc.), GAPDH (1:5000; Sigma). After washing three times, the blots were incubated with horseradish peroxidase-conjugated secondary antibodies for 1 h and visualized using an enhanced chemiluminescence kit (Pierce, Rockford, IL, USA). Finally, the protein bands were visualized after exposure of the membrane to Kodak X-ray film (Kodak, Rochester, NY, USA).

3.9. Hoechst 33342 Staining Analysis

Cells were seeded into 6-well plates, and then treated with RO3280 (50 or 100 nM) and cultured at 37 °C for 24 h, stained with 0.1 µg/mL Hoechst 33342 (Sigma) for 5 min, then observed with filters for blue fluorescence under fluorescence microscopy (OLYMPUS IX71; Olympus Corporation, Tokyo, Japan). Abnormal nuclear cells were counted between the RO3280 treatment group and DMSO control group.

3.10. Real-Time PCR Array Analysis

Samples from each group were submerged in 2 mL Trizol (Invitrogen) for RNA extraction, and stored at −80 °C until further processed. cDNA synthesis was performed on 4 μg of RNA in a 10 μL sample volume using SuperScript II reverse transcriptase (Invitrogen) as recommended by the manufacturer. Real-time PCR array (SABioscience Human Apoptosis PCR Array PAHS-3012, Frederick, MD, USA) analysis was performed in a total volume of 20 μL including 2 μL of cDNA, primers (0.2 mM each) and 10 μL of SYBR Green mix (Roche). For gene expression quantification a comparative Ct method was used. Gene expression levels for each sample were normalized to the expression level of the housekeeping gene encoding Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) within a given sample (−ΔCt); the relative expression of each gene was calculated with 106 × Log 2 (−ΔCt). Statistical significance of gene expression was calculated with the t-test using SPSS 11.5 software (Chicago, IL, USA).

3.11. Statistical Analysis

Each experimental condition was performed three times, and these replicates were presented in results. All values are presented as means ± SEM. Student’s paired t-test was applied to reveal statistical significances. p values less than 0.05 were considered significant. Statistical analyses were performed using SPSS Software for Windows.

4. Conclusions

In this study, we identified PLK1 as a possible oncoprotein target in pediatric AML patient samples and human myeloid leukemia cell lines. Kaplan-Meier survival analysis revealed shorter survival time correlated with high PLK1 expression in tumors. Our findings also showed for the first time that RO3280 treatment inhibits cell proliferation and induces apoptosis in leukemia cells. Real time PCR array analysis demonstrated that 32 genes are significantly upregulated and 16 genes are significantly downregulated after RO3280 treatment. This is the first indication of RIPK3, HRK, DCC, and BTK gene regulation reported with RO3280 treatment. These results may provide new insights into the molecular mechanism of RO3280-induced apoptosis; however, further research will be required to determine the underlying details. Taken together, our findings suggest that RO3280 may be a suitable drug candidate for the treatment of pediatric AML.

Acknowledgments

This work was supported by grants from the National Key Basic Research Program (No. 2010CB933902), grants from key medical subjects of Jiangsu province (XK201120), Innovative team of Jiangsu Province (Nos. LJ201114 and LJ201126), Special Clinical Medical Science and Technology of Jiangsu province (BL2012050, BL2013014), Key Laboratory of Suzhou (SZS201108, SZS201307), National Natural Science Foundation (Nos. 81100371, 81370627, 81300423 and 81272143), Natural Science Foundation of Jiangsu Province (No. BK2011308), Universities Natural Science Foundation of Jiangsu Province (No. 11KJB320014), Talent’s Subsidy Project in Science and Education of the Department of Public Health of Suzhou City (No. SWKQ1020) and Major Scientific and Technological Special Project for “Significant New Drugs Creation” (No. 2012ZX09103301-040).

Author Contributions

Jian Pan and Xing Feng designed and directed the study; Na-Na Wang, Zhi-Heng Li and He Zhao performed most of the experiments; Yan-Fang Tao and Li-Xiao Xu performed the real-time PCR array analysis; Jun Lu, Wen-Li Zhao and Lan Cao collected the leukemia sample; Pei-Fang Xiao, Yi-Ping Li and Yun-Yun Xu collected the clinical information of samples; Xiao-Juan Du and Li-Chao Sun supported the design of primers for PCR; Fang Fang, Gang Li, Guang-Hao Su, Hui-Ting Zhou and Yan-Hong Li drafted the manuscript; Yi Wu, Mei-Fang Jin, Lin Liu and Xue-Ming Zhu performed the apoptosis analysis; Jian Wang, Shao-Yan Hu and Jian Ni participated in study design and coordination, data analysis and interpretation, and drafted the manuscript; and all authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jemal, A.; Thomas, A.; Murray, T.; Thun, M. Cancer statistics. CA Cancer J. Clin. 2002, 52, 23–47. [Google Scholar] [CrossRef] [PubMed]
  2. Jabbour, E.; Cortes, J.; Ravandi, F.; O’Brien, S.; Kantarjian, H. Targeted therapies in hematology and their impact on patient care: Chronic and acute myeloid leukemia. Semin. Hematol. 2013, 50, 271–283. [Google Scholar] [CrossRef] [PubMed]
  3. Miano, M.; Micalizzi, C.; Calvillo, M.; Dufour, C. New targets in pediatric acute myeloid leukemia. Immunol. Lett. 2013, 155, 47–50. [Google Scholar] [CrossRef] [PubMed]
  4. Zhao, Y.; Huang, H.; Wei, G. Novel agents and biomarkers for acute lymphoid leukemia. J. Hematol. Oncol. 2013, 6, 40. [Google Scholar] [CrossRef] [PubMed]
  5. Moore, A.S.; Kearns, P.R.; Knapper, S.; Pearson, A.D.; Zwaan, C.M. Novel therapies for children with acute myeloid leukaemia. Leukemia 2013, 27, 1451–1460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lancet, J.E. New agents: Great expectations not realized. Best Pract. Res. Clin. Haematol. 2013, 26, 269–274. [Google Scholar] [CrossRef] [PubMed]
  7. Ostronoff, F.; Estey, E. The role of quizartinib in the treatment of acute myeloid leukemia. Expert Opin. Investig. Drugs 2013, 22, 1659–1669. [Google Scholar] [CrossRef] [PubMed]
  8. Hales, E.C.; Taub, J.W.; Matherly, L.H. New insights into Notch1 regulation of the PI3K-AKT-mTOR1 signaling axis: Targeted therapy of gamma-secretase inhibitor resistant T-cell acute lymphoblastic leukemia. Cell. Signal. 2014, 26, 149–161. [Google Scholar] [CrossRef] [PubMed]
  9. Vitagliano, O.; Addeo, R.; D’Angelo, V.; Indolfi, C.; Indolfi, P.; Casale, F. The Bcl-2/Bax and Ras/Raf/MEK/ERK signaling pathways: Implications in pediatric leukemia pathogenesis and new prospects for therapeutic approaches. Expert Rev. Hematol. 2013, 6, 587–597. [Google Scholar] [CrossRef] [PubMed]
  10. Peloquin, G.L.; Chen, Y.B.; Fathi, A.T. The evolving landscape in the therapy of acute myeloid leukemia. Protein Cell 2013, 4, 735–746. [Google Scholar] [CrossRef] [PubMed]
  11. Smith, C.C.; Shah, N.P. The role of kinase inhibitors in the treatment of patients with acute myeloid leukemia. Am. Soc. Clin. Oncol. Educ. Book 2013. [Google Scholar] [CrossRef]
  12. Hatzimichael, E.; Georgiou, G.; Benetatos, L.; Briasoulis, E. Gene mutations and molecularly targeted therapies in acute myeloid leukemia. Am. J. Blood Res. 2013, 3, 29–51. [Google Scholar] [PubMed]
  13. Gjertsen, B.T.; Schoffski, P. Discovery and development of the Polo-like kinase inhibitor volasertib in cancer therapy. Leukemia 2014. [Google Scholar] [CrossRef]
  14. Tsykunova, G.; Reikvam, H.; Ahmed, A.B.; Nepstad, I.; Gjertsen, B.T.; Bruserud, O. Targeting of polo-like kinases and their cross talk with Aurora kinases—Possible therapeutic strategies in human acute myeloid leukemia? Expert Opin. Investig. Drugs 2012, 21, 587–603. [Google Scholar] [CrossRef] [PubMed]
  15. Wasch, R.; Hasskarl, J.; Schnerch, D.; Lubbert, M. BI2536—Targeting the mitotic kinase Polo-like kinase 1 (PLK1). In Small Molecules in Oncology; Springer Berlin Heidelberg: Berlin, Germany, 2010; Volume 184, pp. 215–218. [Google Scholar]
  16. Zhang, Z.; Zhang, G.; Kong, C. High expression of Polo-like kinase 1 is associated with the metastasis and recurrence in urothelial carcinoma of bladder. Urol. Oncol. 2013, 31, 1222–1230. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, G.; Zhang, Z.; Liu, Z. Polo-like kinase 1 is overexpressed in renal cancer and participates in the proliferation and invasion of renal cancer cells. Tumour Biol. 2013, 34, 1887–1894. [Google Scholar] [CrossRef] [PubMed]
  18. Maire, V.; Nemati, F.; Richardson, M.; Vincent-Salomon, A.; Tesson, B.; Rigaill, G.; Gravier, E.; Marty-Prouvost, B.; de Koning, L.; Lang, G.; et al. Polo-like kinase 1: A potential therapeutic option in combination with conventional chemotherapy for the management of patients with triple-negative breast cancer. Cancer Res. 2013, 73, 813–823. [Google Scholar] [CrossRef] [PubMed]
  19. Deeraksa, A.; Pan, J.; Sha, Y.; Liu, X.D.; Eissa, N.T.; Lin, S.H.; Yu-Lee, L.Y. PLK1 is upregulated in androgen-insensitive prostate cancer cells and its inhibition leads to necroptosis. Oncogene 2013, 32, 2973–2983. [Google Scholar] [CrossRef] [PubMed]
  20. Ackermann, S.; Goeser, F.; Schulte, J.H.; Schramm, A.; Ehemann, V.; Hero, B.; Eggert, A.; Berthold, F.; Fischer, M. Polo-like kinase 1 is a therapeutic target in high-risk neuroblastoma. Clin. Cancer Res. 2011, 17, 731–741. [Google Scholar] [CrossRef] [PubMed]
  21. Pellegrino, R.; Calvisi, D.F.; Ladu, S.; Ehemann, V.; Staniscia, T.; Evert, M.; Dombrowski, F.; Schirmacher, P.; Longerich, T. Oncogenic and tumor suppressive roles of Polo-like kinases in human hepatocellular carcinoma. Hepatology 2010, 51, 857–868. [Google Scholar] [PubMed]
  22. Zhang, Y.; Liu, Y.; Yang, Y.X.; Xia, J.H.; Zhang, H.X.; Li, H.B.; Yu, C.Z. The expression of PLK-1 in cervical carcinoma: A possible target for enhancing chemosensitivity. J. Exp. Clin. Cancer Res. 2009, 28, 130. [Google Scholar] [CrossRef] [PubMed]
  23. Schmit, T.L.; Zhong, W.; Nihal, M.; Ahmad, N. Polo-like kinase 1 (PLK1) in non-melanoma skin cancers. Cell Cycle 2009, 8, 2697–2702. [Google Scholar] [CrossRef] [PubMed]
  24. Renner, A.G.; Dos Santos, C.; Recher, C.; Bailly, C.; Creancier, L.; Kruczynski, A.; Payrastre, B.; Manenti, S. Polo-like kinase 1 is overexpressed in acute myeloid leukemia and its inhibition preferentially targets the proliferation of leukemic cells. Blood 2009, 114, 659–662. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, Y.; Du, X.L.; Wang, C.J.; Lin, D.C.; Ruan, X.; Feng, Y.B.; Huo, Y.Q.; Peng, H.; Cui, J.L.; Zhang, T.T.; et al. Reciprocal activation between PLK1 and STAT3 contributes to survival and proliferation of esophageal cancer cells. Gastroenterology 2012, 142, 521–530. [Google Scholar] [CrossRef] [PubMed]
  26. Behren, A.; Muhlen, S.; Acuna Sanhueza, G.A.; Schwager, C.; Plinkert, P.K.; Huber, P.E.; Abdollahi, A.; Simon, C. Phenotype-assisted transcriptome analysis identifies FOXM1 downstream from Ras-MKK3-p38 to regulate in vitro cellular invasion. Oncogene 2010, 29, 1519–1530. [Google Scholar] [CrossRef] [PubMed]
  27. Malumbres, M. Preclinical models for cell cycle-targeted therapies. Adv. Exp. Med. Biol. 2006, 587, 139–147. [Google Scholar] [PubMed]
  28. 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] [PubMed]
  29. 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] [PubMed]
  30. Chen, S.; Bartkovitz, D.; Cai, J.; Chen, Y.; Chen, Z.; Chu, X.J.; Le, K.; Le, N.T.; Luk, K.C.; Mischke, S.; et al. Identification of novel, potent and selective inhibitors of Polo-like kinase 1. Bioorgan. Med. Chem. Lett. 2012, 22, 1247–1250. [Google Scholar] [CrossRef]
  31. Gumireddy, K.; Reddy, M.V.; Cosenza, S.C.; Boominathan, R.; 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] [PubMed]
  32. Oliveira, J.C.; Pezuk, J.A.; Brassesco, M.S.; Morales, A.G.; Queiroz, R.G.; Scrideli, C.A.; Tone, L.G. PLK1 expression and BI2536 effects in childhood acute lymphoblastic leukemia. Pediatr. Blood Cancer 2014, 61, 1227–1231. [Google Scholar] [CrossRef] [PubMed]
  33. Dasmahapatra, G.; Patel, H.; Nguyen, T.; Attkisson, E.; Grant, S. PLK1 inhibitors synergistically potentiate HDAC inhibitor lethality in imatinib mesylate-sensitive or resistant BCR/ABL+ leukemia cells in vitro and in vivo. Clin. Cancer Res. 2013, 19, 404–414. [Google Scholar] [CrossRef] [PubMed]
  34. Ikezoe, T.; Yang, J.; Nishioka, C.; Takezaki, Y.; Tasaka, T.; Togitani, K.; Koeffler, H.P.; Yokoyama, A. A novel treatment strategy targeting Polo-like kinase 1 in hematological malignancies. Leukemia 2009, 23, 1564–1576. [Google Scholar] [CrossRef] [PubMed]
  35. Berg, T.; Bug, G.; Ottmann, O.G.; Strebhardt, K. Polo-like kinases in AML. Expert Opin. Investig. Drugs 2012, 21, 1069–1074. [Google Scholar] [CrossRef] [PubMed]
  36. 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 (BI6727) in patients with locally advanced or metastatic urothelial cancer. Cancer 2014, 120, 976–982. [Google Scholar] [CrossRef] [PubMed]
  37. 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.; Fritsch, H.; et al. A phase I study of two dosing schedules of volasertib (BI6727), an intravenous Polo-like kinase inhibitor, in patients with advanced solid malignancies. Br. J. Cancer 2014, 110, 2434–2440. [Google Scholar] [CrossRef] [PubMed]
  38. Wagenblast, J.; Hirth, D.; Thron, L.; Arnoldner, C.; Diensthuber, M.; Stover, T.; Hambek, M. Effects of the Polo-like-kinase-1-inhibitor BI2536 in squamous cell carcinoma cell lines of the head and neck. Oncol. Lett. 2012, 4, 175–177. [Google Scholar] [PubMed]
  39. 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). Bioorg. Med. Chem. Lett. 2013, 23, 3662–3666. [Google Scholar] [CrossRef] [PubMed]
  40. Oussenko, I.A.; Holland, J.F.; Reddy, E.P.; Ohnuma, T. Effect of ON 01910. Na, an anticancer mitotic inhibitor, on cell-cycle progression correlates with RanGAP1 hyperphosphorylation. Cancer Res. 2011, 71, 4968–4976. [Google Scholar] [CrossRef] [PubMed]
  41. Bowles, D.W.; Diamond, J.R.; Lam, E.T.; Weekes, C.D.; Astling, D.P.; Anderson, R.T.; Leong, S.; Gore, L.; Varella-Garcia, M.; Vogler, B.W.; et al. Phase I study of oral rigosertib (ON 01910. Na), a dual inhibitor of the PI3K and PLK1 pathways, in adult patients with advanced solid malignancies. Clin. Cancer Res. 2014, 20, 1656–1665. [Google Scholar] [CrossRef] [PubMed]
  42. Garuti, L.; Roberti, M.; Bottegoni, G. Polo-like kinases inhibitors. Curr. Med. Chem. 2012, 19, 3937–3948. [Google Scholar] [CrossRef] [PubMed]
  43. Schmittgen, T.D.; Lee, E.J.; Jiang, J. High-throughput real-time PCR. Methods Mol. Biol. 2008, 429, 89–98. [Google Scholar] [PubMed]
  44. Liu, X. PLK1, a negative regulator of p21. Cell Cycle 2009, 8, 336–337. [Google Scholar] [PubMed]
  45. Kreis, N.N.; Louwen, F.; Zimmer, B.; Yuan, J. Loss of p21Cip1/CDKN1A renders cancer cells susceptible to Polo-like kinase 1 inhibition. Oncotarget 2014, in press. [Google Scholar]
  46. Driscoll, D.L.; Chakravarty, A.; Bowman, D.; Shinde, V.; Lasky, K.; Shi, J.; Vos, T.; Stringer, B.; Amidon, B.; D’Amore, N.; et al. PLK1 inhibition causes post-mitotic DNA damage and senescence in a range of human tumor cell lines. PLoS One 2014, 9, e111060. [Google Scholar] [CrossRef] [PubMed]
  47. Thalhammer, V.; Lopez-Garcia, L.A.; Herrero Martin, D.; Hecker, R.; Laubscher, D.; Gierisch, M.E.; Wachtel, M.; Bode, P.; Nanni, P.; Blank, B.; et al. PLK1 phosphorylates PAX3-FOXO1 the inhibition of which triggers regression of alveolar rhabdomyosarcoma. Cancer Res. 2014. [Google Scholar] [CrossRef]
  48. Zhao, J.; Jitkaew, S.; Cai, Z.; Choksi, S.; Li, Q.; Luo, J.; Liu, Z.G. Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis. Proc. Natl. Acad. Sci. USA 2012, 109, 5322–5327. [Google Scholar] [CrossRef] [PubMed]
  49. Haddick, P.C.; Tom, I.; Luis, E.; Quinones, G.; Wranik, B.J.; Ramani, S.R.; Stephan, J.P.; Tessier-Lavigne, M.; Gonzalez, L.C. Defining the ligand specificity of the deleted in colorectal cancer (DCC) receptor. PLoS One 2014, 9, e84823. [Google Scholar] [CrossRef] [PubMed]
  50. Meimei, L.; Peiling, L.; Baoxin, L.; Changmin, L.; Rujin, Z.; Chunjie, H. Lost expression of DCC gene in ovarian cancer and its inhibition in ovarian cancer cells. Med. Oncol. 2011, 28, 282–289. [Google Scholar] [CrossRef] [PubMed]
  51. Inohara, N.; Ding, L.; Chen, S.; Nunez, G. harakiri, A novel regulator of cell death, encodes a protein that activates apoptosis and interacts selectively with survival-promoting proteins BCL-2 and BCL-XL. EMBO J. 1997, 16, 1686–1694. [Google Scholar] [CrossRef] [PubMed]
  52. Choi, B.; Pagano, M.; Dai, W. PLK1 phosphorylates PTEN and regulates its mitotic activity during the cell cycle. J. Biol. Chem. 2014, 20, 14066–14074. [Google Scholar] [CrossRef]
  53. Bustin, S.A.; Benes, V.; Garson, J.A.; Hellemans, J.; Huggett, J.; Kubista, M.; Mueller, R.; Nolan, T.; Pfaffl, M.W.; Shipley, G.L.; et al. The MIQE guidelines: Minimum information for publication of quantitative real-time PCR experiments. Clin. Chem. 2009, 55, 611–622. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Wang, N.-N.; Li, Z.-H.; Zhao, H.; Tao, Y.-F.; Xu, L.-X.; Lu, J.; Cao, L.; Du, X.-J.; Sun, L.-C.; Zhao, W.-L.; et al. Molecular Targeting of the Oncoprotein PLK1 in Pediatric Acute Myeloid Leukemia: RO3280, a Novel PLK1 Inhibitor, Induces Apoptosis in Leukemia Cells. Int. J. Mol. Sci. 2015, 16, 1266-1292. https://doi.org/10.3390/ijms16011266

AMA Style

Wang N-N, Li Z-H, Zhao H, Tao Y-F, Xu L-X, Lu J, Cao L, Du X-J, Sun L-C, Zhao W-L, et al. Molecular Targeting of the Oncoprotein PLK1 in Pediatric Acute Myeloid Leukemia: RO3280, a Novel PLK1 Inhibitor, Induces Apoptosis in Leukemia Cells. International Journal of Molecular Sciences. 2015; 16(1):1266-1292. https://doi.org/10.3390/ijms16011266

Chicago/Turabian Style

Wang, Na-Na, Zhi-Heng Li, He Zhao, Yan-Fang Tao, Li-Xiao Xu, Jun Lu, Lan Cao, Xiao-Juan Du, Li-Chao Sun, Wen-Li Zhao, and et al. 2015. "Molecular Targeting of the Oncoprotein PLK1 in Pediatric Acute Myeloid Leukemia: RO3280, a Novel PLK1 Inhibitor, Induces Apoptosis in Leukemia Cells" International Journal of Molecular Sciences 16, no. 1: 1266-1292. https://doi.org/10.3390/ijms16011266

Article Metrics

Back to TopTop