Open Access

A drug targeting 5‑lipoxygenase enhances the activity of a JAK2 inhibitor in CD34+ bone marrow cells from patients with JAK2V617F‑positive polycythemia vera in vitro

  • Authors:
    • Yuan Chen
    • Hu Zhao
    • Jing Luo
    • Youping Liao
    • Kui Tan
    • Guoyu Hu
  • View Affiliations

  • Published online on: March 4, 2021     https://doi.org/10.3892/ol.2021.12612
  • Article Number: 351
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Janus kinase 2 (JAK2) inhibitors, the first targeted treatments for myeloproliferative neoplasms (MPNs), provide substantial benefits, including a marked reduction in splenomegaly and MPN‑associated symptoms. However, these drugs rarely induce molecular remission in patients with MPNs. Zileuton, a 5‑lipoxygenase (5‑LO) inhibitor, has been demonstrated to selectively deplete hematopoietic stem cells (HSCs) expressing a JAK2 point mutation (JAK2V617F) in mouse models of JAK2V617F‑induced polycythemia vera (PV). To determine the potential activity of 5‑LO inhibitors in combination with JAK inhibitors against human PV HSCs, the present study first analyzed 5‑LO expression in CD34+ bone marrow cells from patients with JAK2V617F‑positive PV using western blotting and reverse transcription‑quantitative PCR, and then examined the effect of zileuton combined with ruxolitinib on colony formation using a colony formation assay. Furthermore, cell cycle and apoptosis in CD34+ cells from patients with PV and healthy volunteers were determined by flow cytometry. In the present study, 5‑LO expression was upregulated in CD34+ cells from patients with PV compared with in CD34+ cells from healthy volunteers. Higher levels of leukotriene B4, a product of the 5‑LO signaling pathway, were detected in patients with PV compared with in healthy volunteers. Zileuton treatment suppressed the colony formation of CD34+ cells from patients with PV in a dose‑dependent manner. Furthermore, zileuton and ruxolitinib exerted their anticancer effects by suppressing hematopoietic colony formation, inducing apoptosis and arresting the cell cycle of human CD34+ cells from patients with PV. The combination of these two drugs exerted a more beneficial effect than either agent alone. Based on these data, zileuton enhanced the antitumor activity of low‑dose ruxolitinib in hematopoietic progenitor cells from patients with PV, providing conceptual validation for further clinical applications of combination treatment with ruxolitinib and zileuton for patients with PV.

Introduction

Polycythemia vera (PV), essential thrombocythemia and primary myelofibrosis (1) are Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) that originate from hematopoietic stem cells (HSCs). In 2005, an activating point mutation in Janus kinase 2 (JAK2V617F) was identified in the majority of patients with MPNs (25), which provided critical insights into MPN pathogenesis and promoted the development of Janus kinase 2 (JAK2) inhibitors. According to clinical studies, JAK2 inhibitors effectively decrease spleen size and relieve MPN-associated symptoms; however, their disease-modifying activity is limited (6,7).

5-Lipoxygenase (5-LO) is an important dioxygenase, since it is the key enzyme that catalyzes the transformation of arachidonic acid into inflammatory leukotrienes (LTs) (8). Accumulating evidence has suggested that the 5-LO signaling pathway is directly involved in cancer development by promoting cell proliferation, angiogenesis, migration and invasion, and inhibiting apoptosis (913). As demonstrated in a study by Chen et al (14), 5-LO is upregulated in a mouse model of JAK2V617F-induced PV, and inhibition of 5-LO by zileuton, a selective 5-LO inhibitor, attenuates PV development by blocking JAK2V617F-expressing HSCs in mice. Therefore, it may be hypothesized that zileuton could potentially eliminate persistent malignant HSCs in patients with PV. However, to the best of our knowledge, no previous reports have described the role of 5-LO in patients with JAK2V617F-positive PV.

Based on the aforementioned evidence, the combination of zileuton with a JAK2 inhibitor may be a promising treatment strategy for patients with PV. The present study first analyzed 5-LO expression in CD34+ cells from the bone marrow of patients with JAK2V617F-positive PV using western blotting and reverse transcription-quantitative PCR (RT-qPCR). Subsequently, the effects of zileuton combined with ruxolitinib on colony formation, apoptosis and the cell cycle of CD34+ cells from patients with PV were analyzed in vitro.

Materials and methods

Patient specimens and cell preparation

Bone marrow and peripheral blood were donated by 18 patients who were newly diagnosed with PV and 10 healthy adult volunteers at the Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Zhuzhou, China) between August 2017 and April 2019. All patients met the World Health Organization diagnostic criteria for PV (1). Patient characteristics are shown in Table I. The healthy volunteers were eligible if they were 18–69 years of age and in healthy condition without active infections, and serious liver, kidney, heart and other diseases. Bone marrow and peripheral blood from 10 healthy volunteers were used as normal controls. The volunteers included 6 women and 4 men. The mean age was 41.5 years, and the age ranged between 23 and 69 years. All participants provided written informed consent according to the protocol approved by the Medical Ethics Committees of the Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Zhuzhou, China) and in accordance with the principles outlined in the Declaration of Helsinki. Mononuclear cells were separated from bone marrow samples at 440 × g for 30 min at room temperature using Ficoll-Hypaque density gradient centrifugation (GE Healthcare). An EasySep™ CD34-positive selection kit (Stemcell Technologies, Inc.) was used to enrich the CD34+ cell population according to the manufacturer's protocol. CD34+ cells with a purity ≥85% were used in each experiment.

Table I.

Patient characteristics and experiments performed using patient samples.

Table I.

Patient characteristics and experiments performed using patient samples.

Experiments

CaseAge, yearsSexJAK2V617F allele burden, %Leukotriene B4 ELISAReverse transcription-quantitative PCRWestern blottingHematopoietic progenitor cell assaysFACS for apoptosis assayFACS for cell cycle analysis
PV168Female49YNNYYY
PV270Female26YYNNNY
PV343Male43YYNNYY
PV446Male75NYNYNN
PV567Male90YYNYNN
PV654Female80YNNYYY
PV769Male28YYYNYN
PV855Female70YNYYNN
PV972Female45YYYYYY
PV1067Male83NYYYYN
PV1166Female58YNYNNY
PV1241Female47YYYYYY
PV1361Male61YYYYNN
PV1448Female40YNYNYY
PV1567Male46NYNYYY
PV1668Male57YNNYNN
PV1756Female79NYNNNN
PV1866Male84YYNYYY

[i] PV, polycythemia vera; JAK2V617F, a mutation resulting in a substitution of valine for phenylalanine at amino acid 617 of the JAK2 protein; Y, yes; N, no.

Detection of leukotriene B4 (LTB4)

Plasma samples from patients with PV and healthy volunteers were collected to detect LTB4 levels using a leukotriene B4 Express ELISA kit (cat. no. 10009292; Cayman Chemical Company). Briefly, the standard or plasma sample, LTB4 AchE tracer and anti-LTB4 antibody were sequentially added to each well of a 96-well plate. The plates were then incubated for 60–90 min at room temperature before measuring the absorbance at 405 nm using an ELISA microplate reader (Thermo Fisher Scientific, Inc.).

RT-qPCR

Total RNA was extracted from the purified CD34+ cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). cDNA synthesis was performed using a PrimeScript™ RT reagent Kit with gDNA Eraser (Perfect Real Time) (cat. no. RR047A; Takara Bio, Inc.) according to the manufacturer's protocols. Reactions were incubated at 37°C for 15 min followed by heat inactivation for 5 sec at 85°C for reverse transcription. The PCR amplification was performed using TB Green™ Premix Ex Taq™ II (Tli RNaseH Plus) (cat. no. RR820A; Takara Bio, Inc.). The primer sequences of the human 5-LO gene and the GAPDH gene are listed in Table II. The thermocycling conditions were as follows: 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec and 60°C for 31 sec. Amplification was performed using an ABI 7300 system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The 2−ΔΔCq formula (15) was used to analyze the relative mRNA expression levels of 5-LO, which were normalized to the expression levels of GAPDH.

Table II.

Primer sequences used for reverse transcription-quantitative PCR.

Table II.

Primer sequences used for reverse transcription-quantitative PCR.

GeneProduct length, bpPrimerSequence (5′-3′)
5-lipoxygenase168Forward GCTGAATGACGACTGGTA
Reverse CGGTGTTGCTTGAGAATG
GAPDH263Forward ATGCTGGCGCTGAGTACGTC
Reverse GGTCATGAGTCCTTCCACGATA
Western blotting

RIPA lysis solution (cat. no. P0013B; Beyotime Institute of Biotechnology) was used to extract total proteins from purified CD34+ cells. BCA assays (cat. no. 23227; Pierce; Thermo Fisher Scientific, Inc.) were used to determine the protein concentration. An equal amount of total proteins (30 µg/lane) was separated by 10% SDS-PAGE and then transferred onto PVDF membrane (cat. no. IPVH00010; EMD Millipore). The membranes were blocked using TBS with 0.05% Tween-20 (TBST) containing 5% non-fat milk at room temperature for 2 h, and then incubated with primary antibodies against 5-LO (dilution, 1:500; cat. no. sc-136195; Santa Cruz Biotechnology, Inc.) and β-actin (dilution, 1:1,000; cat. no. ab8226; Abcam) overnight at 4°C. After washing with TBST, the membranes were incubated with secondary antibodies (dilution, 1:2,000; cat. no. 7076; Cell Signaling Technology, Inc.) conjugated to horseradish peroxidase at room temperature for 1 h. Finally, the protein blots were visualized using a Hypersensitive ECL chemiluminescence kit (cat. no. P10018FS; Beyotime Institute of Biotechnology). The results were semi-quantified using ImageJ software (v1.52; National Institutes of Health).

Colony formation assay (CFA)

The colony-forming ability of the cells was estimated by inoculating 500 CD34+ cells in MethoCult H4435 (Stemcell Technologies, Inc.). Various concentrations of zileuton (50, 100, 250 and 500 µM; Cayman Chemical Company) and/or 50 nM ruxolitinib (Cayman Chemical Company) were added for 14 days at 37°C. After 14 days, the presence of colonies (>40 cells) was scored under a light microscope (Zeiss LSM 800 with Airyscan; magnification, ×25; Zeiss AG).

Flow cytometry

CD34+ cells (1×106 cells/well) from patients with PV and healthy volunteers were seeded into a 6-well plate and treated with 100 µM zileuton, 50 nM ruxolitinib or the combination of 100 µM zileuton and 50 nM ruxolitinib for 48 h at 37°C. After 2 days, 5×105 cells were collected and applied to the flow cytometry detection. The apoptosis assay was performed using an Annexin V-FITC Apoptosis Detection Kit (cat. no. 556547; BD Biosciences). In brief, cells were stained with Annexin V-FITC and PI for 15 min at room temperature, and detected using a flow cytometer. Stained cells were analyzed using a FACSCalibur instrument (BD Biosciences) using a 488 nm excitation wavelength and emission was detected at 530 nm (for FITC) and 575 nm (for PI). The data were analyzed using the BD FACSuite™ version 1.01 (BD Biosciences). The phases of the cell cycle and DNA synthesis activity of CD34+ cells were determined using a FITC BrdU Flow kit (cat. no. 559619; BD Biosciences) according to the manufacturer's protocol. Briefly, cells were incubated with BrdU, a nucleoside analogue of thymidine, and then stained with anti-human CD34-allophycocyanin (dilution, 1:5; cat. no. 560940; BD Biosciences). After fixing in BD Cytofix/Cytoperm Buffer for 30 min on ice and permeabilizing in BD Cytoperm Permeabilization Buffer Plus for 10 min on ice, cells were treated with DNase to expose BrdU epitopes and then incubated with a FITC-conjugated anti-BrdU antibody (provided in kit; BD Biosciences) for 20 min at room temperature. DNA was counterstained with 7-aminoactinomycin D (7AAD; provided in kit; BD Biosciences) for 15 min at room temperature. Stained cells were then analyzed using a FACSCalibur instrument using a 488 nm excitation wavelength and emission was detected at 530 nm (for FITC-conjugated anti-BrdU) and 610 nm (for 7AAD). The data were analyzed using BD FACSuite™ version 1.01.

Statistical analysis

All experiments were repeated three times. Data are presented as medians and interquartile ranges. Differences in 5-LO expression and LTB4 levels between groups were compared using a two-tailed Mann-Whitney test, while data derived from the same samples after different treatments were analyzed using the Friedman test, and the Nemenyi post hoc test was subsequently used for pairwise comparisons between groups. GraphPad Prism 7 software (GraphPad Software, Inc.) and SPSS 26.0 software (IBM Corp.) were utilized for statistical analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

5-LO expression is increased in CD34+ cells from patients with PV

To determine the potential effects of 5-LO inhibitor zileuton in the treatment of human PV, the present study first assessed the basal protein and mRNA expression levels of 5-LO in CD34+ cells from the bone marrow of patients with PV and healthy volunteers. Western blot analysis revealed that CD34+ cells from patients with PV exhibited higher protein expression levels of 5-LO than those from healthy volunteers (Fig. 1A and B; Table SI). Consistent with the results observed for protein expression, 5-LO mRNA expression was also increased in CD34+ cells from patients with PV compared with in those from healthy volunteers, as demonstrated by the results of RT-qPCR (Fig. 1C; Table SII).

LTB4 levels are elevated in patients with PV

Plasma levels of LTB4, a metabolite of the 5-LO signaling pathway, were measured in 14 patients with PV and 10 healthy volunteers using ELISA. Higher LTB4 levels were observed in patients with PV compared with those in healthy volunteers (Fig. 2; Table SIII).

Zileuton suppresses the colony formation of CD34+ cells from patients with PV in a dose-dependent manner

A CFA was performed to assess the effect of zileuton treatment on the colony formation of primary CD34+ cells in vitro. As shown in Fig. 3, zileuton treatment inhibited granulocytes and monocytes (CFU-GM)- and burst-forming unit-erythroid (BFU-E)-derived colony formation by PV CD34+ cells in a dose-dependent manner, with an IC50 of 460.4 µM for CFU-GM and 233.5 µM for BFU-E (Fig. 3B and C; Table SIV). By contrast, zileuton treatment did not markedly alter the colony formation of the CD34+ cells from healthy volunteers (Fig. 3A and C; Table SIV).

Reduction of colony formation by zileuton treatment in PV CD34+ cells occurs through a reduction in 5-LO expression

To explore whether the effect of zileuton on colony formation of hematopoietic cells was associated with the levels of 5-LO, 5-LO protein expression in CD34+ cells from patients with PV and healthy volunteers was measured with and without treatment with increasing concentrations of zileuton using western blotting. As shown in the Fig. 4, zileuton dose-dependently decreased 5-LO protein expression in CD34+ cells from a patient with PV, which exhibited high 5-LO expression, but had no significant effects on 5-LO expression in CD34+ cells from a healthy volunteer.

Combination treatment with zileuton and ruxolitinib synergistically inhibits the colony formation of hematopoietic cells from patients with PV

The present study investigated the effect of combination treatment with zileuton and ruxolitinib on the formation of colonies of primary CD34+ cells from 10 patients with PV and 6 healthy volunteers in vitro. The doses selected for the present study were 100 µM zileuton and 50 nM ruxolitinib. In the CD34+ cells from healthy volunteers, treatment with 100 µM zileuton, 50 nM ruxolitinib or 100 µM zileuton combined with 50 nM ruxolitinib did not alter colony formation (Fig. 5A and C; Table SV). By contrast, the yields of CFU-GM and BFU-E in CD34+ cells from patients with PV treated with 50 nM ruxolitinib were decreased by 19 and 35%, respectively, and the yields of CFU-GM and BFU-E in CD34+ cells from patients with PV treated with 100 µM zileuton were decreased by 12 and 20%, respectively (Fig. 5B and C; Table SV). However, the combination of ruxolitinib with zileuton had a greater effect on the colony formation of CD34+ cells from patients with PV, with a 36% a reduction in CFU-GM and a 55% reduction in BFU-E (Fig. 5B and C; Table SV).

Combination treatment with zileuton and ruxolitinib induces apoptosis in CD34+ cells from patients with PV

The apoptosis rate of CD34+ cells from patients with PV and healthy volunteers after treatment with zileuton and/or ruxolitinib was detected using flow cytometry. The apoptosis rate of CD34+ cells from patients with PV was slightly increased following zileuton or ruxolitinib treatment (Fig. 6; Table SVI). However, combination treatment with zileuton and ruxolitinib induced apoptosis in a greater number of CD34+ cells from patients with PV than treatment with either individual drug. By contrast, neither ruxolitinib nor zileuton alone or in combination induced apoptosis of CD34+ cells from healthy volunteers (Fig. 6; Table SVI).

Treatment with zileuton and ruxolitinib arrests CD34+ cells from patients with PV at the G0/G1 phase of the cell cycle

Cell cycle arrest is an important effect of numerous anticancer agents. The present study investigated the effects of zileuton and/or ruxolitinib on the cell cycle of CD34+ cells from patients with PV in vitro. As shown in Fig. 7A and C, treatment with zileuton and ruxolitinib alone slightly increased the number of CD34+ cells from patients with PV that were arrested in the G0/G1 phase of the cell cycle, with a concomitant decrease in the percentage of cells in S phase (Table SVII). However, combination treatment with zileuton and ruxolitinib caused more CD34+ cells from patients with PV to be arrested in the G0/G1 phase and a significant decrease in the proportion of cells in S phase compared with either monotherapy. By contrast, the percentage of CD34+ cells from healthy volunteers in each phase of the cell cycle did not markedly change, regardless of whether they were treated with zileuton, ruxolitinib or the combination treatment (Fig. 7A and B; Table SVII).

Discussion

PV is the most common Philadelphia chromosome-negative MPN (16). Thrombosis and hemorrhage, as well as myelofibrotic and/or leukemic transformation (17,18), are potential complications occurring in patients with PV. The JAK2V617F mutation in the pseudokinase domain leads to constitutive phosphorylation of JAK2 and overactivation of downstream signaling pathways, ultimately resulting in uncontrolled myeloid cell proliferation (19,20). These findings motivated the clinical development of JAK kinase inhibitors for patients with MPNs. Ruxolitinib was the first selective JAK1/2 inhibitor that was demonstrated to be effective in patients with PV and myelofibrosis (2123), and it provides substantial benefits, including a marked decrease in splenomegaly and disease symptoms (24). However, treatment with ruxolitinib rarely induces molecular or pathological remission in patients with MPNs (2527). Furthermore, the dose-dependent hematological toxicity of ruxolitinib represents a major concern for a number of patients (28). Researchers have expressed an interest in identifying novel agents with different mechanisms of action other than targeting the JAK-STAT signaling pathway.

5-LO expression is upregulated in numerous different types of cancer, including pancreatic, breast, prostate, esophageal and colon cancer (2933). The products of 5-LO, such as 5-hydroxyeicosatetraenoic acid (5-HETE) and LTs, are able to promote cell proliferation, suppress apoptosis, promote angiogenesis and enhance tumor cell invasion (3436). According to previous studies, epidermal growth factors and neurotensin are involved in the 5-LO-mediated tumor progression in individuals with prostate cancer (37,38). A study of patients with colorectal cancer revealed that 5-HETE stimulates angiogenesis by inducing the expression of VEGF (39,40). Furthermore, increased activities of 5-LO and matrix metalloproteinases are associated with extracellular matrix stiffness (41) and enhance the invasiveness of cancer cells (42,43). The enzyme 5-LO is involved in the development of not only solid malignancies but also certain forms of leukemia (44,45). Recently, the 5-LO gene was shown to be a critical regulator for mouse leukemic stem cells (LSCs) in BCR-ABL-induced chronic myeloid leukemia (CML), and the combination of zileuton and imatinib extends the survival time of mice with CML (46). This finding motivated an initial clinical trial combining zileuton with imatinib as a treatment for CML (clinicaltrials.com; NCT02047149, NCT01130688) with no available efficacy data at present. Zileuton has been revealed to selectively deplete JAK2V617F-expressing HSCs, thereby preventing PV development in mice (14). The molecular mechanism of 5-LO in PV is associated with the β-catenin signaling pathway, which is required for the maintenance of both HSCs (47) and LSCs in individuals with CML (4850). Based on these results, drugs targeting the 5-LO signaling pathway can eradicate human JAK2V617F+ malignant HSCs and thus are potentially curative treatments for MPNs. However, a previous study reported low expression levels of 5-LO and LTB4 receptor 1 in CD34+ cells from patients with BCR-ABL-positive CML compared with healthy donors (51). Another study reported that 5-LO expression was undetectable in more primitive CML LSCs (52). Therefore, 5-LO has distinct and important regulatory and functional roles in human and murine CML. To the best of our knowledge, no previous reports have described the role of 5-LO in human PV.

In the present study, 5-LO mRNA and protein expression was increased in CD34+ cells from patients with PV compared with in CD34+ cells from healthy volunteers. Higher LTB4 levels were detected in patients with PV compared with healthy volunteers. Based on these results, the 5-LO signaling pathway was upregulated in patients with JAK2V617F-positive PV, consistent with previous findings from PV mice (14), indicating that 5-LO may be involved in the pathogenesis of human JAK2V617F-positive PV. Zileuton treatment decreased the colony formation of CD34+ cells from patients with PV in a dose-dependent manner by reducing 5-LO expression in PV CD34+ cells. Furthermore, zileuton and ruxolitinib exerted their anticancer effects by suppressing the colony formation of hematopoietic cells, inducing apoptosis and blocking the cell cycle of CD34+ cells from patients with PV. The combination of the two drugs was more effective than either agent alone. Similar effects were not observed in CD34+ cells from healthy volunteers after treatment with zileuton or ruxolitinib, either alone or in combination. Therefore, zileuton enhanced the antitumor activity of ruxolitinib against hematopoietic progenitor cells from patients with PV, suggesting that zileuton and ruxolitinib may represent an effective therapeutic combination. Notably, activities of both 100 µM zileuton and 50 nM ruxolitinib were relatively specific for hematopoietic progenitor cells from patients with PV, while sparing CD34+ cells from healthy volunteers. The latter finding suggests that the combination treatment may exhibit improved safety and low hematological toxicity.

In conclusion, zileuton exerted a synergistic effect with ruxolitinib on CD34+ cells from patients with PV by suppressing cell proliferation, inducing apoptosis and arresting the cell cycle, which provides conceptual validation for further clinical applications of combination therapy with ruxolitinib and zileuton for patients with PV. However, because CD34+ cells collected from some patients were not amplified enough to complete all downstream experiments, only a portion of the samples were analyzed in some experiments. This was a limitation of the present study. Future studies should focus on investigating the molecular mechanisms by which 5-LO inhibitors block colony formation, and induce apoptosis and cell cycle arrest, in patients with PV.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was funded by the Clinical Medical Technology Innovation Guidance Project of Hunan Province (grant no. 2018SK52803).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

GH and YC proposed and designed the current study. JL and YL selected patients and collected samples/clinical data. YC and HZ conducted the experiments. YC, HZ and KT were responsible for acquiring, analyzing and interpreting the data. YC and HZ drafted the initial manuscript. YC, HZ, JL and GH reviewed and edited the manuscript. GH, YC and HZ assessed the authenticity of all the raw data and ensured its legitimacy. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The experimental protocol was established according to the ethical guidelines of the Helsinki Declaration and was approved by the Medical Ethics Committees of The Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Zhuzhou, China). Written informed consent was obtained from each individual.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M and Vardiman JW: The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 127:2391–2405. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, et al: Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 365:1054–1061. 2005. View Article : Google Scholar : PubMed/NCBI

3 

James C, Ugo V, Le Couédic JP, Staerk J, Delhommeau F, Lacout C, Garçon L, Raslova H, Berger R, Bennaceur-Griscelli A, et al: A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 434:1144–1148. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Jones AV, Kreil S, Zoi K, Waghorn K, Curtis C, Zhang L, Score J, Seear R, Chase AJ, Grand FH, et al: Widespread occurrence of the JAK2 V617F mutation in chronic myeloproliferative disorders. Blood. 106:2162–2168. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, Tichelli A, Cazzola M and Skoda RC: A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 352:1779–1790. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Deininger M, Radich J, Burn TC, Huber R and Verstovsek S: The effect of long-term ruxolitinib treatment on JAK2p.V617F allele burden in patients with myelofibrosis. Blood. 126:1551–1554. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Cervantes F and Pereira A: Does ruxolitinib prolong the survival of patients with myelofibrosis? Blood. 129:832–837. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Needleman P, Turk J, Jakschik BA, Morrison AR and Lefkowith JB: Arachidonic acid metabolism. Ann Rev Biochem. 55:69–102. 1986. View Article : Google Scholar : PubMed/NCBI

9 

Peters-Golden M and Henderson WR Jr: Leukotrienes. N Engl J Med. 357:1841–1854. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Pidgeon GP, Lysaght J, Krishnamoorthy S, Reynolds JV, O'Byrne K, Nie D and Honn KV: Lipoxygenase metabolism: Roles in tumor progression and survival. Cancer Metastasis Rev. 26:503–524. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Wang D and Dubois RN: Eicosanoids and cancer. Nat Rev Cancer. 10:181–193. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Bishayee K and Khuda-Bukhsh AR: 5-lipoxygenase antagonist therapy: A new approach towards targeted cancer chemotherapy. Acta Biochim Biophys Sin (Shanghai). 45:709–719. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Moore GY and Pidgeon GP: Cross-talk between cancer cells and the tumour microenvironment: The role of the 5-lipoxygenase pathway. Int J Mol Sci. 18:2362017. View Article : Google Scholar

14 

Chen Y, Shan Y, Lu M, DeSouza N, Guo Z, Hoffman R, Liang A and Li S: Alox5 blockade eradicates JAK2V617F-induced polycythemia Vera in mice. Cancer Res. 77:164–174. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Mehta J, Wang H, Iqbal SU and Mesa R: Epidemiology of myeloproliferative neoplasms in the United States. Leuk Lymphoma. 55:595–600. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Hoffman R, Prchal JT, Samuelson S, Ciurea SO and Rondelli D: Philadelphia chromosome-negative myeloproliferative disorders: Biology and treatment. Biol Blood Marrow Transplant. 13 (Suppl 1):S64–S72. 2007. View Article : Google Scholar

18 

Mascarenhas J: A concise update on risk factors, therapy, and outcome of leukemic transformation of myeloproliferative neoplasms. Clin Lymphoma Myeloma Leuk. 16 (Suppl):S124–S129. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Silvennoinen O and Hubbard SR: Molecular insights into regulation of JAK2 in myeloproliferative neoplasms. Blood. 125:3388–3392. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, Boggon TJ, Wlodarska I, Clark JJ, Moore S, et al: Activating mutation in the tyrosine kinase JAK2 in polycythemia Vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 7:387–397. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, et al: A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 366:799–807. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Hasselbalch HC and Bjørn ME: Ruxolitinib versus standard therapy for the treatment of polycythemia Vera. N Engl J Med. 372:16702015. View Article : Google Scholar : PubMed/NCBI

23 

Pardanani A, Harrison C, Cortes JE, Cervantes F, Mesa RA, Milligan D, Masszi T, Mishchenko E, Jourdan E, Vannucchi AM, et al: Safety and efficacy of fedratinib in patients with primary or secondary myelofibrosis: A randomized clinical trial. JAMA Oncol. 1:643–651. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Kiladjian J, Verstovsek S, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Kirito K, et al: Results from the 208-week (4-year) Follow-up of RESPONSE Trial, a Phase 3 study comparing ruxolitinib (Rux) with best available therapy (BAT) for the treatment of polycythemia Vera (PV). Blood. 130 (Suppl 1):S3222017.

25 

Verstovsek S, Gotlib J, Mesa RA, Vannucchi AM, Kiladjian JJ, Cervantes F, Harrison CN, Paquette R, Sun W, Naim A, et al: Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J Hematol Oncol. 10:1562017. View Article : Google Scholar : PubMed/NCBI

26 

Harrison CN, Vannucchi AM, Kiladjian JJ, Al-Ali HK, Gisslinger H, Knoops L, Cervantes F, Jones MM, Sun K, McQuitty M, et al: Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs. best available therapy for myelofibrosis. Leukemia. 31:7752016. View Article : Google Scholar

27 

Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT, Talpaz M, et al: Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J Hematol Oncol. 10:552017. View Article : Google Scholar : PubMed/NCBI

28 

Harrison C, Kiladjian JJ, Al-Ali HK, Gisslinger H, Waltzman R, Stalbovskaya V, McQuitty M, Hunter DS, Levy R, Knoops L, et al: JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 366:787–798. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Avis I, Hong SH, Martínez A, Moody T, Choi YH, Trepel J, Das R, Jett M and Mulshine JL: Five-lipoxygenase inhibitors can mediate apoptosis in human breast cancer cell lines through complex eicosanoid interactions. FASEB J. 15:2007–2009. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Melstrom LG, Bentrem DJ, Salabat MR, Kennedy TJ, Ding XZ, Strouch M, Rao SM, Witt RC, Ternent CA, Talamonti MS, et al: Overexpression of 5-lipoxygenase in colon polyps and cancer and the effect of 5-LOX inhibitors in vitro and in a murine model. Clin Cancer Res. 14:6525–6530. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Hennig R, Ding XZ, Tong WG, Schneider MB, Standop J, Friess H, Büchler MW, Pour PM and Adrian TE: 5-Lipoxygenase and leukotriene B(4) receptor are expressed in human pancreatic cancers but not in pancreatic ducts in normal tissue. Am J Pathol. 161:421–428. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Hoque A, Lippman SM, Wu TT, Xu Y, Liang ZD, Swisher S, Zhang H, Cao L, Ajani JA and Xu XC: Increased 5-lipoxygenase expression and induction of apoptosis by its inhibitors in esophageal cancer: A potential target for prevention. Carcinogenesis. 26:785–791. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Matsuyama M, Yoshimura R, Mitsuhashi M, Hase T, Tsuchida K, Takemoto Y, Kawahito Y, Sano H and Nakatani T: Expression of lipoxygenase in human prostate cancer and growth reduction by its inhibitors. Int J Oncol. 24:821–827. 2004.PubMed/NCBI

34 

Hyde CA and Missailidis S: Inhibition of arachidonic acid metabolism and its implication on cell proliferation and tumour-angiogenesis. Int Immunopharmacol. 9:701–715. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Ding XZ, Iversen P, Cluck MW, Knezetic JA and Adrian TE: Lipoxygenase inhibitors abolish proliferation of human pancreatic cancer cells. Biochem Biophys Res Commun. 261:218–223. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Wen Z, Liu H, Li M, Li B, Gao W, Shao Q, Fan B, Zhao F, Wang Q, Xie Q, et al: Increased metabolites of 5-lipoxygenase from hypoxic ovarian cancer cells promote tumor-associated macrophage infiltration. Oncogene. 34:1241–1252. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Hassan S and Carraway RE: Involvement of arachidonic acid metabolism and EGF receptor in neurotensin-induced prostate cancer PC3 cell growth. Regul Pept. 133:105–114. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Karlage KL, Mogalian E, Jensen A and Myrdal PB: Inhalation of an ethanol-based zileuton formulation provides a reduction of pulmonary adenomas in the A/J mouse model. AAPS PharmSciTech. 11:168–173. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Romano M, Catalano A, Nutini M, D'Urbano E, Crescenzi C, Claria J, Libner R, Davi G and Procopio A: 5-Lipoxygenase regulates malignant mesothelial cell survival: Involvement of vascular endothelial growth factor. FASEB J. 15:2326–2336. 2001. View Article : Google Scholar : PubMed/NCBI

40 

Ye YN, Wu WK, Shin VY, Bruce IC, Wong BC and Cho CH: Dual inhibition of 5-LOX and COX-2 suppresses colon cancer formation promoted by cigarette smoke. Carcinogenesis. 26:827–834. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, et al: Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 139:891–906. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, Le QT and Giaccia AJ: Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 15:35–44. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Kirschmann DA, Seftor EA, Fong SF, Nieva DR, Sullivan CM, Edwards EM, Sommer P, Csiszar K and Hendrix MJ: A molecular role for lysyl oxidase in breast cancer invasion. Cancer Res. 62:4478–4483. 2002.PubMed/NCBI

44 

Graham SM, Vass JK, Holyoake TL and Graham GJ: Transcriptional analysis of quiescent and proliferating CD34+ human hemopoietic cells from normal and chronic myeloid leukemia sources. Stem Cells. 25:3111–3120. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Roos J, Oancea C, Heinssmann M, Khan D, Held H, Kahnt AS, Capelo R, La Buscato E, Proschak E, Puccetti E, et al: 5-Lipoxygenase is a candidate target for therapeutic management of stem cell-like cells in acute myeloid leukemia. Cancer Res. 74:5244–5255. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Chen Y, Hu Y, Zhang H, Peng C and Li S: Loss of the Alox5 gene impairs leukemia stem cells and prevents chronic myeloid leukemia. Nat Genet. 41:783–792. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Ruiz-Herguido C, Guiu J, D'Altri T, Inglés-Esteve J, Dzierzak E, Espinosa L and Bigas A: Hematopoietic stem cell development requires transient Wnt/β-catenin activity. J Exp Med. 209:1457–1468. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Hu Y, Chen Y, Douglas L and Li S: Beta-catenin is essential for survival of leukemic stem cells insensitive to kinase inhibition in mice with BCR-ABL-induced chronic myeloid leukemia. Leukemia. 23:109–116. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, Lagoo A and Reya T: Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 12:528–541. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Heidel FH, Bullinger L, Feng Z, Wang Z, Neff TA, Stein L, Kalaitzidis D, Lane SW and Armstrong SA: Genetic and pharmacologic inhibition of beta-catenin targets imatinib-resistant leukemia stem cells in CML. Cell Stem Cell. 10:412–424. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Lucas CM, Harris RJ, Giannoudis A, Mcdonald E and Clark RE: Low leukotriene B4 receptor 1 leads to ALOX5 downregulation at diagnosis of chronic myeloid leukemia. Haematologica. 99:1710–1715. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Dolinska M, Piccini A, Wong WM, Gelali E, Johansson AS, Klang J, Xiao P, Yektaei-Karin E, Strömberg UO, Mustjoki S, et al: Leukotriene signaling via ALOX5 and cysteinyl leukotriene receptor 1 is dispensable for in vitro growth of CD34+ CD38 stem and progenitor cells in chronic myeloid leukemia. Biochem Biophys Res Commun. 490:378–384. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Y, Zhao H, Luo J, Liao Y, Tan K and Hu G: A drug targeting 5‑lipoxygenase enhances the activity of a JAK2 inhibitor in CD34<sup>+</sup> bone marrow cells from patients with JAK2V617F‑positive polycythemia vera <em>in vitro</em>. Oncol Lett 21: 351, 2021
APA
Chen, Y., Zhao, H., Luo, J., Liao, Y., Tan, K., & Hu, G. (2021). A drug targeting 5‑lipoxygenase enhances the activity of a JAK2 inhibitor in CD34<sup>+</sup> bone marrow cells from patients with JAK2V617F‑positive polycythemia vera <em>in vitro</em>. Oncology Letters, 21, 351. https://doi.org/10.3892/ol.2021.12612
MLA
Chen, Y., Zhao, H., Luo, J., Liao, Y., Tan, K., Hu, G."A drug targeting 5‑lipoxygenase enhances the activity of a JAK2 inhibitor in CD34<sup>+</sup> bone marrow cells from patients with JAK2V617F‑positive polycythemia vera <em>in vitro</em>". Oncology Letters 21.5 (2021): 351.
Chicago
Chen, Y., Zhao, H., Luo, J., Liao, Y., Tan, K., Hu, G."A drug targeting 5‑lipoxygenase enhances the activity of a JAK2 inhibitor in CD34<sup>+</sup> bone marrow cells from patients with JAK2V617F‑positive polycythemia vera <em>in vitro</em>". Oncology Letters 21, no. 5 (2021): 351. https://doi.org/10.3892/ol.2021.12612