Part 1: Molecular analysis of phenotypic heterogeneity in JAK2V617Fpositive myeloproliferative neoplasms reveals a potential target for therapy
Abstract
Background: Classical BCR-ABL-negative myeloproliferative neoplasms (MPNs) are a group of clonal hematopoietic disorders, including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis. JAK2V617F is the most frequent mutation in MPNs. It is an important but not the only determinant of MPN phenotype. Recent studies have revealed several factors contributing to phenotypic heterogeneity of MPN, including driver mutation, additional mutation, mutational order, tumor loads and hematopoietic stem cell (HSC) heterogeneity. However, it is still not fully elucidated how a single JAK2V617F mutation, and even the same tumor load, lead to different disease phenotypes.
Objective: The aim of the study is to unveil factors involved in phenotypic heterogeneity and to identify novel therapeutic targets for MPN.
Methods: Next-generation sequencing (NGS) was performed on JAK2V617F+ET and PV patient samples to find molecular markers revealing phenotypic heterogeneity. RNA sequencing was performed on HSCs and megakaryocyte-erythroid progenitors (MEPs) of healthy donors and MPN patients to find signaling pathways differentially regulated during hematopoietic differentiation of different disease phenotypes. The effects of targeting Wnt/β-catenin signaling on inhibiting megakaryocyte (MK) and platelet production were confirmed by in vitro MK differentiation. The phenotype of MK was identified using the following methods: observation of cell morphology under microscope, analysis of the proportion and mean fluorescence intensity of CD41- and CD61-positive cells by immunofluorescence,analysis of the size and number of MK colonies by colony forming units-MK assays, analysis of the proportion of CD41- and CD42b-positive cells and MK ploidy by flow cytometry analysis. JAK2V617F+MPN mice were injected with XAV939, and then underwent blood routine test to validate the effect of thrombocytosis remission of Wnt/β-catenin inhibitor in vivo. Mice bone marrow hematopoietic stem progenitor cells (HSPCs) were analyzed to study the mechanism of action of the drug in reducing platelet levels.
Results: Using NGS technology, two concurrent mutations that may affect phenotype were identified, including mutations in SH2B3, which is primarily prevalent in PV, and SF3B1, which is more commonly mutated in ET. Higher JAK2V617F allele burden was associated with PV phenotype. A receiver operating characteristic curve identified JAK2V617F allele burden of 34.8% as the optimal cut-off level to distinguish PV and ET. Transcriptomic analysis of HSCs and MEPs from ET, PV and healthy donors showed that inflammatory signaling pathways were elevated in both HSCs and MEPs of ET, unlike in PV HSCs and MEPs. Notably, Wnt/β-catenin signaling was uniquely upregulated during ET hematopoietic differentiation from HSC to MEP, and inhibiting Wnt/β-catenin signaling blocked the formation of MK and platelets in vitro. CD34+cells derived from ET patients were more sensitive to Wnt/β-catenin inhibitor compared with that from healthy donors, and MK differentiation from ET cells was more inhibited at the same drug concentration. Consistently, Wnt/β-catenin inhibitor administration decreased platelet counts in JAK2V617F+MPN mice by blocking MEPs and megakaryocyte progenitors and by inhibiting MK maturation. While in wild-type mice, Wnt/β-catenin inhibitor did not significantly reduce platelet counts.
Conclusion: Our findings provide new insights into the mechanisms underlying phenotypic
differentiation of JAK2V617F+ PV and ET at the level of hematopoietic differentiation. Wnt/β-catenin signaling is specifically upregulated during ET hematopoietic differentiation. Inhibiting Wnt/β-catenin signaling can inhibit MK differentiation in vitro and reduce platelet counts in vivo, indicating that Wnt/β-catenin signaling may serve as a potential therapeutic target of MPN.
Part 2: Development and validation of a multiple factor-based prognostic score system of thrombosis in polycythemia vera
Abstract
Background: Polycythemia vera (PV) is a subtype of classic BCR-ABL-negative myeloproliferative neoplasm (MPN), characterized by abnormally elevated mature red blood cells and high incidence of thrombosis. Thrombosis and thrombotic complications are important causes of death for patients with PV. The conventional stratification of thrombosis divides patients into low-risk and high-risk groups according to age and previous thrombosis.However, this two-tiered thrombosis stratification may ignore some potential risk factors.
Objectives: This study aimed to develop and validate a multiple factor-based prediction model of thrombosis for the 2016 World Health Organization (WHO)-defined PV.
Methods: Patients from our institution were included as training cohort, and patients from another center were included as external validation cohort. Clinical and next-generation sequencing (NGS) data from two cohorts of patients with PV were analyzed. Multivariable Cox regression analysis was conducted for identification of thrombotic risk factors and model development. The discrimination of the model was evaluated by the area under receiver operating characteristic curve (AUC) and the C index. The calibration of the model was evaluated by the calibration curve and the goodness-of-fit test. Thrombosis-free survival of patients in different risk groups was displayed using Kaplan-Meier survival curves and compared by log-rank test.
Results: The study involved 372 patients in the training cohort and another 195 patients in the external validation cohort. In the training cohort, thrombotic events were recorded in 136 patients (36.6%), including 122 patients (32.8%) with only arterial thrombosis, 7 patients (1.9%) with only venous thrombosis, and 7 patients (1.9%) with both arterial and venous thrombosis. There were 68 patients (18.3%) with thrombosis after diagnosis, corresponding to an incidence rate of 3.6/100 patient-years during follow-up. Multivariable analysis indicated that age≥60 years (hazard ratio [HR] 2.56, 95% confidence interval [CI] 1.51–4.35, p<0.001), cardiovascular risk factors (HR 4.22, 95%CI 2.00–8.92, p<0.001), at least one high-risk mutation for thrombosis (mutations in DNMT3A, ASXL1, or BCOR/BCORL1) (HR 4.35, 95%CI 2.62–7.21, p<0.001) and previous thrombosis (HR 5.93, 95%CI 3.29–10.68, p<0.001) were independent risk factors of thrombosis. Those risk factors remained significant contributors to thrombosis after accounting for the competing risk of death. According to the weight of β coefficient, those factors above were assigned 1 point, 1.5 points, 1.5 points and 2 points respectively. A multiple factor-based prognostic score system of thrombosis (MFPSPV) was then developed, classifying patients into low-risk (≤1 point; n=96, 25.8%), intermediate-risk (1.5–2.5 points; n=146, 39.2%) and high-risk (≥3 points; n=130, 35.0%)groups. Patients in the three groups had significantly different thrombosis-free survival rates (p<0.001). The MFPS-PV outperformed the conventional model in discrimination power (Cstatistic: 0.87 [95%CI 0.83–0.91] vs 0.80 [95%CI 0.74–0.86]; AUC: 0.90 [95%CI 0.86–0.95] vs. 0.82 [95%CI 0.76–0.89]). The MFPS-PV was well-calibrated and remained consistent during external validation.
Conclusions: The MFPS-PV, integrating clinical characteristics and NGS data for the first time, successfully incorporated molecular genetic indicators into thrombosis prediction, and showed excellent accuracy and utility for the 2016 WHO-defined PV.