Abstract

Acute myeloid leukemia (AML) is a molecularly and clinically heterogeneous disease. PDE3A is a well-characterized cyclic nucleotide phosphodiesterase (PDE) that hydrolyzes cAMP, cGMP, and cUMP. Recent studies have demonstrated that PDE3A was upregulated in several solid tumors of patients including breast cancer, ovarian carcinoma, melanoma, which correlated with poor prognosis. PDE3A activates inflammatory pathways that mediate cancer cell stemness by suppressing the cAMP/PKA pathway. Targeting PDE3A with chemicals suppressed cancer cells by inducing cell apoptosis and reducing metastasis. PDE3A inhibitor Anagrelide (ANA) is an antithrombotic and platelet reducing agent used to treat the thrombocythemia associated with myeloproliferative diseases. This study aims to explore the relationship between PDE3A expression and AML prognosis, and to investigate the feasibility and mechanism of PDE3A as a therapeutic target for AML. To explore the relationship between PDE3A expression and the survival time of patients, a total of 141 pediatric AML patients were recruited in this retrospective study from October 2013 to September 2021 in the Children's Hospital of Soochow University. The follow-up period was until March 2022. The transcriptomic expression level of PDE3A in newly diagnosed bone marrow samples was obtained according to the results of RNA sequencing. Patients were divided into two groups based on median PDE3A expression: PDE3Ahigh group (n= 70) and PDE3Alow group(n=71). Results showed AML patients in PDE3Ahigh group had significantly lower 5-year overall survival (OS) (71.4% vs 92.9%, P = 0.0010), event free survival (52.1% vs 84.6%, P = 0.0150) and relapse free survival (73.2% vs 91.4%, P = 0.0325) compared to patients in PDE3Alow group. Next, to evaluate PDE3A expression level in AML cell lines, we used RT-PCR and Western Blot to compare the difference of PDE3A expression in HEL, Kasumi-1, MV4-11, NB4, K562, HL60 and MEG-01. Among these cell lines, HEL has significantly higher PDE3A expression compared to others at both transcriptional and protein levels. We found that ANA specifically inhibited HEL growth in a concentration dependent manner after 72 hours of ANA treatment. ANA reached half of the inhibitory concentration on HEL at 0.9μM. Interestingly, ANA had no inhibitory effects on other tested AML cell lines which expressed low level of PDE3A. Thus, we choose HEL as PDE3Ahigh AML cell model and treatment with 0.9μM ANA for 72 hours for further experiments. By flow cytometry, we found ANA interrupted cell cycle by G1 arrest in HEL, but did not induce cell apoptosis. In consistent with these results, the expression of cell cycle-related proteins including CDK2, CDK4, CDK6, CDK9 and C-MYC were significantly down-regulated in a time-dependent manner after ANA treatment in HEL compared to control group. To explore the effect of ANA on PDE3A high expression AML in vivo, we established the preclinical model of AML by injecting 2 million luciferase overexpressed HEL cells into NSG mice via tail vein. Fourteen days later, the mice were treated with vehicle or 5mg/kg ANA treatment by gavage daily for 15 days as the therapeutic model. More interestingly, in vivo imaging assay indicated that ANA treatment reduced leukemia burden compared with the vehicle group. The anti-leukemia effects of ANA significantly prolonged survival time compared to vehicle (median survival time, 32.5 vs 24.5 days, P = 0.0091). In conclusion, our clinical data suggested that high expression of PDE3A in newly diagnosed pediatric AML patients was associated with poor clinical survival. Besides, our in vitro and in vivo data indicated that ANA targeting PDE3A suppressed progression of AML by interrupting cell cycle, providing a promising approach to eradicate AML with high level of PDE3A expression.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call