The impact of mutational burden, spliceosome and epigenetic regulator mutations on transfusion dependency in dysplastic neoplasms
-
Bernhard Strasser
, Sebastian Mustafa
Abstract
Objectives
Myelodysplastic neoplasms and dysplastic chronic myelomonocytic leukemia are characterized by cytopenia. Therefore, transfusion dependency is high in these dysplastic neoplasms. We investigated the impact of molecular genetics on the transfusion dependency in dysplastic neoplasms.
Methods
We investigated the impact of the myeloid mutation burden on transfusion dependency in myelodysplastic neoplasms and dysplastic chronic myelomonocytic leukemia. In addition, the effect of different functional genetic groups, such as spliceosomes and epigenetic regulator gene mutations, on transfusion dependency was assessed in these patients. Confounding transfusion triggers were ruled out by the patient selection criteria and regression analyses.
Results
A greater number of mutations lead to a higher transfusion dependency for red blood cells and platelet concentrates. A higher transfusion dependency was associated with a higher transformation to acute myeloid leukemia. Spliceosome mutations were associated with a higher transfusion dependency of red blood cell concentrates than epigenetic regulator mutations.
Conclusions
Molecular genetics has the potential to improve the precision of patient blood management in dysplastic neoplasms.
Introduction
In the updated 5th edition of the World Health Organization (WHO) Classification of Hematolymphoid Tumors, genetic-based diagnosis has become more incorporated and refined. Defining genetic abnormalities is integral to this classification in significant conditions, such as myelodysplastic neoplasm (MDS) and chronic myelomonocytic leukemia (CMML). In recent years, massive parallel sequencing or next-generation sequencing (NGS) evolved from a sophisticated scientific technique to a widely available method for routine practice. This advancement has improved diagnostic precision and enabled more targeted therapeutic options [1], 2]. Patients with MDS or CMML often require the supportive therapy, including red blood cell concentrates (RBC) or platelet concentrates (PC) along with disease-specific treatments [3].
In MDS, the leading cytopenia is macrocytic anemia, which can be regarded as the principal symptom of MDS and is a major contributor to impairments in patients’ health-related quality of life [4], 5]. The estimated prevalence of thrombocytopenia in MDS ranges from 40 to 65 %. At platelet counts <50 × 109/L, hemorrhagic events can occur in half of MDS patients [6]. Tang et al. reported severe thrombocytopenia (defined as platelets <20 × 109/L) in 26.5 % of patients, which was associated with life-threatening complications, including gastrointestinal and intracranial bleeding events, resulting in poor overall survival outcomes [7]. Rozema et al. investigated the transfusion frequency in MDS patients and found that almost 50 % had a high transfusion burden, which was defined as ≥8 units/16 weeks. The study identified older age, a high-risk MDS status, and MDS with multilineage dysplasia or MDS with blast excess as risk factors for higher transfusion requirements. However, none of the above studies investigated the association between genetic lesions and transfusion dependency [8].
CMML is divided into myelodysplastic (MD-CMML) and myeloproliferative (MP-CMML) types depending on the absolute leukocyte count (cut off: MD-CMML <13 × 109/L). MD-CMML and MDS share similar phenotypes with predominant cell lineage myelodysplasia and consequent cytopenia. However, MD-CMML exhibits an absolute monocytosis of ≥0.5 × 109/L. The criteria of absolute monocytosis were refined in the updated WHO classification and lowered to a new cut-off value [9]. In CMML, as with MDS, anemia and thrombocytopenia are highly prevalent, which are also standard criteria for eligible prognostic scoring systems for the disease [10], 11].
Considering the mutational landscape of MDS and CMML, evidence regarding their typical mutations has grown in recent years. In CMML, TET2 mutations are the most frequent. TET2 mutations have also been identified as early events in CMML, affecting the hematopoietic stem cell level. Second-order mutations that accumulate in progenitor cells are mutations encoding the spliceosome apparatus, such as SRSF2, SF3B1, U2AF1, and ZRSR2, or mutations in genes that are relevant to epigenetic control of transcriptional processes involving histone modification (EZH2, ASXL1) and DNA methylation (TET2, DNMT3A, IDH1, and IDH2). Mutations of cell signaling pathways, such as JAK2, KRAS, NRAS, CBL, PTPN11, and FLT3, are typically associated with a myeloproliferative CMML phenotype. In CMML, particularly in MD-CMML, spliceosome mutations (SM) and epigenetic regulatory mutations (ERM) have been iteratively confirmed as the most prevalent mutations [10], [11], [12].
The recurrent mutations of MDS are similar to those in MD-CMML. The clonal architecture of MDS at diagnosis is characterized by the presence of 2–4 causative driver mutations. In MDS, as in CMML, the main mutations belong to the groups of spliceosome mutations and epigenetic regulator mutations. Previous studies have indicated mutations in the functional groups of SM and ERM are early events in disease initiation, while mutations in miscellaneous gene categories are responsible for disease progression. Co-mutations in other cellular pathways are limited to less than 10 % in MDS and predominately involve transcription factors, cohesin, cellular signaling pathways, DNA repair, or tumor suppressor genes [13], [14], [15], [16].
Objectives
In transfusion medicine, the establishment of patient blood management as a multimodal approach to better standardize patient supply with blood components has been a groundbreaking improvement in transfusion management [17]. However, more than 15 years after its establishment, transfusion indications should not only be restricted to traditional criteria but should also include precision medicine, incorporating molecular genetics into therapeutic decision-making. In MDS and MD-CMML the knowledge about genotype-to-phenotype correlations and possible therapeutic options is increasing, particularly regarding recurrent mutations in ERM and SM genes. To date, no study has elucidated the influence of the most pertinent mutations on the transfusion frequency in MDS and MD-CMML. Therefore, we investigated the impact of molecular genetics on the transfusion dependency in dysplastic neoplasms.
Materials and methods
A retrospective database analysis was performed to assess the impact of myeloid mutations on transfusion requirements in patients with MDS/MD-CMML. A homogenous study cohort was ensured by including only dysplastic hematological neoplasms with comparable hematological phenotypes, such as cytopenia and myelodysplasia.
The genes included in the ERM group were TET2 (NM_017628.4), DNMT3A (NM_022552.5), ASXL1 (NM_015338.6, Exon 10, 12, 13), EZH2 (NM_004456.5), IDH1 (NM_005896.4, Exon 4), and IDH2 (NM_002168.4, Exon 4) [11], 14], 18]. The SM group genes were SRSF2 (NM_003016.4, Exon 1), U2AF1 (NM_006758.3, Exon 2, 6), SF3B1 (NM_012433.4, Exon 10–16), and ZRSR2 (NM_005089.4) [11], 15], 16]. Additionally, the association between higher transfusion requirements and an increase transformation rate of MDS/MD-CMML into acute myeloid leukemia (AML) was analyzed. AML was defined as the presence of >20 % myeloblasts in peripheral blood and/or bone marrow. Genetic exceptions that allow AML classification below this myeloblast threshold, as per WHO criteria, were not present in our cohort.
Molecular genetic analysis (SM and ERM or NON-SM/ERM mutations) was carried out using the NGS Sophia Genetics® myeloid solution panel, including 30 genes and gene sections of ABL1, ASXL1, BRAF, CALR, CBL, CEBPA, CSF3R, DNMT3A, ETV6, EZH2, FLT3, HRAS, IDH1, IDH2, JAK2, KIT, KRAS, MPL, NPM1, NRAS, PTPN11, RUNX1, SETBP1, SF3B1, SRSF2, TET2, TP53, U2AF1, WT1, and ZRSR2. NGS was conducted on the Illumina MiSeq platform. DNA was first fragmented and ligated to platform-specific adapters during library preparation. Adapter-ligated fragments were then enriched for the target regions using a hybridization-based capture system to ensure comprehensive coverage of the regions of interest. The prepared libraries were loaded onto the MiSeq flow cell, where cluster generation was achieved through bridge amplification. Sequencing was performed using Illumina’s sequencing-by-synthesis chemistry, enabling high-fidelity base calling. Primary sequence data processing, including base calling and demultiplexing, was carried out with the MiSeq control software. The resultant sequence reads were aligned to the hg19 human reference genome using the Data-Driven Medicine (DDM) software from Sophia Genetics, which also facilitated variant calling and initial classification. Variants were annotated and interpreted using tertiary analysis tools, including ClinVar, Alamut Visual, and VarSome. Variants were classified according to the guidelines of the American College of Medical Genetics and Genomics (ACMG). Variants classified as benign or likely benign were excluded.
Cytogenetics in this study encompassed all structural and numerical chromosomal abnormalities, which were detected through conventional karyotyping and/or fluorescence in situ hybridization (FISH) analyses.
Basic patient data and the administered RBC and PC were assessed by analyzing electronic patient records. The initial diagnoses of MDS/MD-CMML were established according to the 2017 WHO diagnostic criteria. During the retrospective database analysis, the updated 2022 WHO diagnostic criteria were carefully considered to ensure consistency with the revised definitions and classifications (updated 2022 WHO diagnostic criteria).
The electronic medical records of the hospital, Klinikum Wels-Grieskirchen, were analyzed from 2018 (the time of the establishment of Next Generation Sequencing of myeloid mutations in our laboratory institute) until 2023. Patients with MDS or MD-CMML in whom at least one somatic mutation was detected were included in the study. Exclusion criteria were defined to reflect potential confounding transfusion triggers. Thus, patients with concomitant neoplastic disease, chronic renal impairment (higher than CKD stage 3b), and hemolytic or hemorrhagic anemia were excluded a priori. It was unnecessary to add iron, folate, or vitamin B12 deficiencies or alcohol abuse to our exclusion criteria as these conditions are routinely ruled out in clinical practice by our standard hematological diagnostic workup as the most common differential diagnoses of dysplastic neoplasms in Middle Europe.
Statistical analysis
Patients’ baseline characteristics are described by the medians for continuous variables and frequencies and percentages for categorical variables. Further statistical analyses were conducted to compare the effect of the number of mutations on transfusion requirements (≥3 vs. <3), the transfusion differences between the two functional genetic groups (SM vs. ERM) and to analyze the association between transfusion requirements and the AML transformation. A two-sided unpaired t test was applied when a normal distribution was determined. The Wilcoxon rank sum test was applied when no normal distribution was evident. Normal distribution was checked graphically (frequency, Quantile-Quantile Plots) and tested using the Shapiro–Wilk test. The equality of variances was assessed using the Levene test. Frequent transfusion triggers were already considered in the exclusion criteria to control for possible confounding of the collected data. Additionally, a linear regression model and a generalized regression model (negative binomial regression) with the main target variable of red blood cell concentrate transfusion requirement were used for the regression analysis, which included the co-mutations (NON-SM/ERM) and the cytogenetic aberration as model variables (potential confounders, transfusion triggers). The statistical analyses were performed using the R 4.3.2 statistics program.
Results
The primary aim was to investigate whether the number of myeloid mutations influences transfusion dependency and if an increasing number of myeloid mutations increases the need for transfusion. Secondly, the different transfusion requirements between mutations in the main functional genetic groups, such as SM and ERM, were scrutinized. The mutational status of patients was assessed at the time of diagnosis. Transfusion requirements were recorded starting from the time of diagnosis and tracked over the subsequent observation period. A total of 173 patients with MD-CMML or MDS were diagnosed at Klinikum Wels-Grieskirchen during the observation period, and after applying the aforementioned restrictive inclusion and exclusion criteria, 119 were included in the study (Table 1).
Summary of hematological and molecular genetic parameters at diagnosis, including transfusion requirements.
Total Mut. | ERM | SM | NON-ERM/SM | Cytogenetics | Hemoglobin, g/dL | Platelets, 103/L | RBC/month | PC/month | Observational period | ||
---|---|---|---|---|---|---|---|---|---|---|---|
MDS/MD-CMML | |||||||||||
|
|||||||||||
Total patients: 119 | Mean | 2.76 | 1.69 | 0.57 | 0.50 | 0.45 | 10.68 | 161.21 | 9.45 | 1.50 | 15.09 |
Mean age: 67.76 years | Std | 1.60 | 1.12 | 0.58 | 0.86 | 0.87 | 1.96 | 138.91 | 13.70 | 4.57 | 14.85 |
M 72, F 47 | Median | 2.00 | 2.00 | 1.00 | 0.00 | 0.00 | 10.80 | 124.00 | 4.00 | 0.00 | 10.00 |
|
|||||||||||
MD-CMML | |||||||||||
|
|||||||||||
Total patients: 30 | Mean | 2.80 | 1.90 | 0.43 | 0.47 | 0.57 | 11.66 | 146.63 | 9.03 | 1.03 | 15.50 |
Mean age: 70.1 years | Std | 1.35 | 0.88 | 0.50 | 0.78 | 1.07 | 1.82 | 144.16 | 15.29 | 2.62 | 12.21 |
M 15, F 14 | Median | 2.00 | 2.00 | 0.00 | 0.00 | 0.00 | 12.10 | 121.00 | 0.00 | 0.00 | 15.00 |
|
|||||||||||
MDS | |||||||||||
|
|||||||||||
Total patients: 89 | Mean | 2.75 | 1.62 | 0.62 | 0.52 | 0.42 | 10.35 | 166.12 | 9.58 | 1.66 | 14.96 |
Mean age: 67.0 years | Std | 1.69 | 1.18 | 0.59 | 0.89 | 0.79 | 1.90 | 137.58 | 13.21 | 5.07 | 15.70 |
M 56, F 33 | Median | 2.00 | 1.00 | 1.00 | 0.00 | 0.00 | 10.20 | 124.00 | 6.00 | 0.00 | 9.00 |
-
M, male; F, female; std, standard deviation; Total Mut., total mutations; ERM, epigenetic regulator mutations; SM, spliceosome mutations; NON-ERM/SM, non-epigenetic/spliceosome mutations; RBC/month, red blood cell transfusions per month; PC/month, platelet cell transfusions per month.
The study cohort exhibited demographic and clinical characteristics typical for patients with MD-CMML/MDS, with a mean age of 67.76 years and a slight male predominance (M:F ratio 72:47). The cohort consisted of 30 CMML (25.2 %) and 89 MDS (74.8 %) patients. Anemia was more frequently observed than thrombocytopenia in the cohort, with a higher proportion of MDS cases compared to MD-CMML cases. This is reflected by a median hemoglobin level of 10.8 g/dL, indicative of anemia, and a platelet count of 124 × 103/μL, which highlights the presence of thrombocytopenia in a subset of patients.
On a molecular level, the most frequently observed mutations were in epigenetic regulators (ERM; median: 2 mutations per patient), followed by spliceosome mutations (SM; median: 1 mutation) and non-ERM/SM mutations (NON-ERM/SM; median: 0 mutations). RBC requirements were notable, with a mean of 9.45 RBC units per month (median: 4.0 RBC units). PC transfusions were infrequent, with a mean of 1.50 PC units per month (median: 0).
First, the impact of the number of mutations on transfusion dependency in MDS/MD-CMML was investigated. Patients with ≥3 myeloid mutations (55 patients) were compared to those with <3 mutations (64 patients). In the group with ≥3 myeloid mutations, the mean hemoglobin value at diagnosis was 11.32 g/dL compared to 9.95 g/dL in patients with <3 mutations. Anemia was more prevalent in patients with a higher number of mutations (93 %) compared to those with fewer mutations (72 %). Transfusion dependency was higher in the cohort with ≥3 mutations, with a mean of 2.60 RBC transfusions per month compared to 0.54 per month in the group with fewer mutations (p<0.001, r=0.6). In addition, a lower mean platelet count (147,000/µL) and a higher rate of thrombocytopenia (83 %) were observed in patients with ≥3 mutations compared to those with fewer mutations (mean platelet count of 173,000/µL and thrombocytopenia rate of 59 %). The PC transfusion dependency was higher in patients with more mutations (mean 0.34 per month) compared to those with fewer mutations (0.01 per month) (Figure 1).

The number of mutations is consistently shown on the x-axis (A–D). Panel (A) illustrates the difference in RBC levels between individuals with high and low mutation counts. Panels (B–D) depict how transfusion requirements for PC and RBC increase with the number of mutations.
Subsequently, the impact of important confounding factors (RBC transfusion triggers), such as cytogenetic aberrations and NON-SM/ERM mutations, was assessed. A negative binomial regression analysis showed no significant effect of NON-SM/ERM (p=0.424) and cytogenetic aberrations (p=0.873) on RBC transfusion dependency in the study cohort. These results were confirmed by a simple linear regression which considered the observational period: NON-SM/ERM (p=0.6348) and cytogenetics (p=0.390) did not significantly alter the analysis of transfusion dependency of RBC per month in MDS/MD-CMML. Therefore, cytogenetic aberrations and NON-SM/ERM were not significant confounders in our study of the impact of SM and ERM and the total number of mutations on RBC transfusion dependency in MDS/MD-CMML. A negative binomial model, assumed due to strong overdispersion, showed a highly significant correlation between RBC transfusion rate and the total number of mutations (β1=0.53), resulting in an odds ratio of =1.69. Thus, the number of RBC transfusions per month increased by 69 % per mutation.
After analyzing the effect of mutation burden on RBC and PC transfusion dependency, we were interested in whether there were differences between functional mutational groups. The impact of SM and ERM mutations on transfusion requirements in MDS/MD-CMML was analyzed. Cases with at least one ERM or SM, but not both, were included. Significantly higher RBC transfusion requirements per month were observed In the SM-dominant sub-cohort (p<0.001), however the analysis of the PC transfusion dependency did not show a significant difference between the SM and ERM groups (p=0.16).
Finally, analysis revealed a higher transfusion dependency in the cohort of MDS/MD-CMML patients who progressed to AML (15 patients) than those who did not progress (p<0.0001).
Discussion
MDS/MD-CMML are characterized by cytopenia, including anemia and thrombocytopenia. Consequently, the requirements for RBC and PC transfusions are typically high for these patients [19], 20]. Morphological screening of those entities can now be supported by digital pathology systems [21], [22], [23]. Large cohort studies have demonstrated that incorporating molecular genetic data can increase diagnostic and prognostic precision when assessing critical primary endpoints, such as overall survival, progression-free survival, and leukemia-free survival [24], 25]. Given that molecular genetics improve the precision of patient care in MDS and CMML and that transfusion dependency is a frequent requirement of these patients, it was of interest to determine whether molecular genetics can impact transfusion dependency. Therefore, we analyzed the association between mutational burden and transfusion burden in MDS/MD-CMML patients.
Our study investigated whether a high myeloid mutational burden (defined as three or more mutations) is associated with a higher transfusion requirement compared to a low myeloid mutational burden (defined as 1–2 mutations). It was found that patients with a higher number of mutations had a higher prevalence of anemia and thrombocytopenia and required more RBC and PC transfusions. Potential transfusion triggers were excluded via the patient recruitment strategy and statistical analysis using linear regression and negative binomial regression analysis. These new clinical findings expand the spectrum of the genetic-to-phenotype correlation in MDS/MD-CMML. Previous studies have reported adverse patient outcomes in MDS and CMML patients with a higher number of myeloid mutations [24], 25]. The cumulative effect of these mutations results in a higher severity of disease, which is compatible with our findings and indicates higher PC and RBC requirements and a higher AML transformation rate with and increasing mutational burden.
Additionally, the effect of functional molecular genetic groups, beyond the number of mutations, was investigated. Specifically, we compared the ubiquitous molecular genetic groups of SM (including SRSF2, SF3B1, U2AF1, and ZRSR2) and ERM (including TET2, DNMT3A, ASXL1, EZH2, IDH1, and IDH2) in MDS/MD-CMML [12], 26], 27]. To exclude the quantity effect of mutations on transfusion dependency, patients with exclusively one SM or one ERM were compared. In this scenario, it was found that patients with SM had a higher RBC requirement but not a higher PC requirement. Previous in vitro studies have increased our understanding of SM and ERM and their contribution to myelodysplastic changes in hematopoiesis and their role in leukemogenesis. SM occurs in the splice machinery apparatus, where mature mRNA is received after removing intronic sequences from pre-mRNA and ligating the remaining exons. Different compositions and utilization of exons result in different protein isoforms with varying cell functions. One leading concept is that SM alters the removal of introns from the protein-coding transcripts [28], 29]. In two hotspot SM mutations affecting the U2AF1 gene, it has been reported that these mutations result in intron retention with sequence-dependent mis-splicing of genes involved in hematopoiesis [30]. U2AF1 is highly expressed in erythropoietic cells, and U2AF1 knockdown studies have demonstrated reduced erythropoietic cell growth, increased apoptosis, and delayed erythropoietic cell differentiation [31]. Many studies support the concept that in SRSF2 and U2AF1 mutations, alternative exon usage is primarily responsible for deregulated splicing processes, whereas SF3B1 mutations result in aberrant 3`splice site usage in three genes involved in heme biosynthesis and iron metabolism in erythropoietic cells. This dysregulated splicing pathway might be responsible for ring sideroblast formation in erythropoietic cells [32], 33].
The mechanisms of ERM in altering hematopoiesis differ from those of SM. Epigenetic processes predominately modify gene expression (primarily by methylation, posttranslational histone modification, and RNA-based mechanisms) and do not directly alter the DNA nucleotide sequence. Epigenetics in neoplastic processes often report on promotor hypermethylation with consecutive tumor suppressor gene silencing or hypomethylation with consecutive genomic instability. In dysplastic neoplasms, disrupted methylation patterns have been shown to primarily affect the granulopoiesis with insufficient maturation and dysgranulopoietic changes, such as hypogranulation and chromatin condensation [34], [35], [36]. Clinical studies have supported the basic research findings via a higher anemia tendency of SM mutations than in ERM [27], 37], 38]. In addition, ERM are considered earlier mutational events than SM in dysplastic neoplasms. Thus, ERM-dominant dysplastic neoplasms correspond to a lower severity of disease with lower tendency toward cytopenia. Based on this data, this study expanded the clinical characterization of SM vs. ERM and highlighted the clinical significance of the SM-related anemia dimension via increased RBC requirements.
Limitation of the study
This study did not capture information on potentially mutagenic treatments, which constitutes a limitation, as these could represent confounding factors affecting transfusion requirements. The retrospective design inherently restricts the systematic evaluation of such influences. To address this limitation, future prospective studies with detailed documentation of concomitant medications are recommended. These studies would provide a more robust framework to analyze the potential impact of mutagenic treatments on transfusion dependency.
Conclusions
A high number of myeloid mutations are related to a high need for RBC and PC transfusions. A high transfusion dependency is associated with a higher rate of AML transformation. Beyond the quantitative effect of myeloid mutations on transfusion dependency, SM is associated with higher RBC requirements (but not PC requirements) compared to ERM. Incorporating molecular genetics into patient care can improve the precision of transfusion requirements in dysplastic neoplasms, such as MDS/MD-CMML. A precise a priori assessment including molecular genetics at diagnosis, can improve the estimation of transfusion requirements, which is beneficial for patient follow-up. An added value is conceivable because, based on molecular genetics, some patients may need more transfusions and closer monitoring. Therefore, incorporating molecular genetics for estimating transfusion dependency is expedient for clinical practice. This is the first study to specifically analyze the differences in transfusion dependency caused by various functional gene groups such as spliceosome mutations and epigenetic regulator mutations. Further studies with larger sample sizes are needed to confirm the data from this exploratory study.
Acknowledgments
We would like to thank Gertraud Wallner for her continuous and comprehensive support.
-
Research ethics: Research involving human subjects complied with all relevant national regulations, institutional policies and is in accordance with the tenets of the Helsinki Declaration (as revised in 2013), and has been approved by the authors’ Institutional Review Board of Johannes Kepler University/Upper Austria.
-
Informed consent: Not applicable.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors state no conflict of interest.
-
Research funding: None declared.
-
Data availability: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
References
1. Ogbue, O, Unlu, S, Ibodeng, GO, Singh, A, Durmaz, A, Visconte, V, et al.. Single-cell next-generation sequencing to monitor hematopoietic stem-cell transplantation: current applications and future perspectives. Cancers 2023;15. https://doi.org/10.3390/cancers15092477.Search in Google Scholar PubMed PubMed Central
2. Seto, A, Downs, G, King, O, Salehi-Rad, S, Baptista, A, Chin, K, et al.. Genomic characterization of partial tandem duplication involving the KMT2A gene in adult acute myeloid leukemia. Cancers 2024;16. https://doi.org/10.3390/cancers16091693.Search in Google Scholar PubMed PubMed Central
3. Germing, U, Oliva, EN, Hiwase, D, Almeida, A. Treatment of anemia in transfusion-dependent and non-transfusion-dependent lower-risk MDS: current and emerging strategies. Hemasphere 2019;3:e314. https://doi.org/10.1097/hs9.0000000000000314.Search in Google Scholar PubMed PubMed Central
4. Stauder, R, Valent, P, Theurl, I. Anemia at older age: etiologies, clinical implications, and management. Blood 2018;131:505–14. https://doi.org/10.1182/blood-2017-07-746446.Search in Google Scholar PubMed
5. Wouters, H, Conrads-Frank, A, Koinig, KA, Smith, A, Yu, G, de Witte, T, et al.. The anemia-independent impact of myelodysplastic syndromes on health-related quality of life. Ann Hematol 2021;100:2921–32. https://doi.org/10.1007/s00277-021-04654-1.Search in Google Scholar PubMed PubMed Central
6. Kantarjian, H, Giles, F, List, A, Lyons, R, Sekeres, MA, Pierce, S, et al.. The incidence and impact of thrombocytopenia in myelodysplastic syndromes. Cancer 2007;109:1705–14. https://doi.org/10.1002/cncr.22602.Search in Google Scholar PubMed
7. Tang, Y, Zhang, X, Han, S, Chu, T, Qi, J, Wang, H, et al.. Prognostic significance of platelet recovery in myelodysplastic syndromes with severe thrombocytopenia. Clin Appl Thromb Hemost 2018;24:217S–222S. https://doi.org/10.1177/1076029618802363.Search in Google Scholar PubMed PubMed Central
8. Rozema, J, van Roon, EN, Kibbelaar, RE, Veeger, N, Slim, CL, de Wit, H, et al.. Patterns of transfusion burden in an unselected population of patients with myelodysplastic syndromes: a population-based study. Transfus 2021;61:2877–84. https://doi.org/10.1111/trf.16631.Search in Google Scholar PubMed PubMed Central
9. Khoury, JD, Solary, E, Abla, O, Akkari, Y, Alaggio, R, Apperley, JF, et al.. The 5th edition of the World Health Organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms. Leuk 2022;36:1703–19. https://doi.org/10.1038/s41375-022-01613-1.Search in Google Scholar PubMed PubMed Central
10. Patnaik, MM. How I diagnose and treat chronic myelomonocytic leukemia. Haematol 2022;107:1503–17. https://doi.org/10.3324/haematol.2021.279500.Search in Google Scholar PubMed PubMed Central
11. Patnaik, MM, Tefferi, A. Chronic myelomonocytic leukemia: 2022 update on diagnosis, risk stratification, and management. Am J Hematol 2022;97:352–72. https://doi.org/10.1002/ajh.26455.Search in Google Scholar PubMed
12. Palomo, L, Meggendorfer, M, Hutter, S, Twardziok, S, Adema, V, Fuhrmann, I, et al.. Molecular landscape and clonal architecture of adult myelodysplastic/myeloproliferative neoplasms. Blood 2020;136:1851–62. https://doi.org/10.1182/blood.2019004229.Search in Google Scholar PubMed PubMed Central
13. Chiereghin, C, Travaglino, E, Zampini, M, Saba, E, Saitta, C, Riva, E, et al.. The genetics of myelodysplastic syndromes: clinical relevance. Genes 2021;12. https://doi.org/10.3390/genes12081144.Search in Google Scholar PubMed PubMed Central
14. Heuser, M, Yun, H, Thol, F. Epigenetics in myelodysplastic syndromes. Semin Cancer Biol 2018;51:170–9. https://doi.org/10.1016/j.semcancer.2017.07.009.Search in Google Scholar PubMed PubMed Central
15. Rahman, MA, Lin, KT, Bradley, RK, Abdel-Wahab, O, Krainer, AR. Recurrent SRSF2 mutations in MDS affect both splicing and NMD. Genes Dev 2020;34:413–27. https://doi.org/10.1101/gad.332270.119.Search in Google Scholar PubMed PubMed Central
16. Jenkins, JL, Kielkopf, CL. Splicing factor mutations in myelodysplasias: insights from spliceosome structures. Trends Genet 2017;33:336–48. https://doi.org/10.1016/j.tig.2017.03.001.Search in Google Scholar PubMed PubMed Central
17. Franchini, M, Marano, G, Veropalumbo, E, Masiello, F, Pati, I, Candura, F, et al.. Patient Blood Management: a revolutionary approach to transfusion medicine. Blood Transfus 2019;17:191–5. https://doi.org/10.2450/2019.0109-19.Search in Google Scholar PubMed PubMed Central
18. Venney, D, Mohd-Sarip, A, Mills, KI. The impact of epigenetic modifications in myeloid malignancies. Int J Mol Sci 2021;22. https://doi.org/10.3390/ijms22095013.Search in Google Scholar PubMed PubMed Central
19. Jouzier, C, Cherait, A, Cony-Makhoul, P, Hamel, JF, Veloso, M, Thepot, S, et al.. Red blood cell transfusion burden in myelodysplastic syndromes (MDS) with ring sideroblasts (RS): A retrospective multicenter study by the Groupe Francophone des Myelodysplasies (GFM). Transfus 2022;62:961–73. https://doi.org/10.1111/trf.16884.Search in Google Scholar PubMed
20. Song, X, Qi, J, Li, X, Zhou, M, He, J, Chu, T, et al.. Exploration of risk factors of platelet transfusion refractoriness and its impact on the prognosis of hematopoietic stem cell transplantation: a retrospective study of patients with hematological diseases. Platelets 2023;34:2229905. https://doi.org/10.1080/09537104.2023.2229905.Search in Google Scholar PubMed
21. Mayes, C, Gwilliam, T, Mahe, ER. Improving turn-around times in low-throughput distributed hematology laboratory settings with the CellaVision® DC-1 instrument. J Lab Med 2025;49:71–5, https://doi.org/10.1515/labmed-2023-0073.Search in Google Scholar
22. Ingalls, K, O’Reilly, TA, Twohig, B, Conrad, DM. Development of a peripheral blood morphology proficiency assessment program using the CellaVision® Proficiency Software. J Lab Med 2025;49:89–93, https://doi.org/10.1515/labmed-2024-0011.Search in Google Scholar
23. Kim, H, Lee, G-H, Yoon, S, Hur, M, Kim, HN, Park, M, et al.. Performance of digital morphology analyzer Medica EasyCell assistant. Clin Chem Lab Med 2023;61:1858–66. https://doi.org/10.1515/cclm-2023-0100.Search in Google Scholar PubMed
24. Sauta, E, Robin, M, Bersanelli, M, Travaglino, E, Meggendorfer, M, Zhao, LP, et al.. Real-world validation of molecular international prognostic scoring system for myelodysplastic syndromes. J Clin Oncol 2023;41:2827–42. https://doi.org/10.1200/jco.22.01784.Search in Google Scholar
25. Bernard, E, Tuechler, H, Greenberg, PL, Hasserjian, RP, Arango Ossa, JE, Nannya, Y, et al.. Molecular international prognostic scoring system for myelodysplastic syndromes. NEJM Evid 2022;1. EVIDoa2200008. https://doi.org/10.1056/evidoa2200008.Search in Google Scholar PubMed
26. Yoshida, K, Sanada, M, Shiraishi, Y, Nowak, D, Nagata, Y, Yamamoto, R, et al.. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 2011;478:64–9. https://doi.org/10.1038/nature10496.Search in Google Scholar PubMed
27. Malcovati, L, Stevenson, K, Papaemmanuil, E, Neuberg, D, Bejar, R, Boultwood, J, et al.. SF3B1-mutant MDS as a distinct disease subtype: a proposal from the International Working Group for the Prognosis of MDS. Blood 2020;136:157–70. https://doi.org/10.1182/blood.2020004850.Search in Google Scholar PubMed PubMed Central
28. Graubert, TA, Shen, D, Ding, L, Okeyo-Owuor, T, Lunn, CL, Shao, J, et al.. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat Genet 2011;44:53–7. https://doi.org/10.1038/ng.1031.Search in Google Scholar PubMed PubMed Central
29. Visconte, V, Makishima, H, Maciejewski, JP, Tiu, RV. Emerging roles of the spliceosomal machinery in myelodysplastic syndromes and other hematological disorders. Leuk 2012;26:2447–54. https://doi.org/10.1038/leu.2012.130.Search in Google Scholar PubMed PubMed Central
30. Biancon, G, Joshi, P, Zimmer, JT, Hunck, T, Gao, Y, Lessard, MD, et al.. Precision analysis of mutant U2AF1 activity reveals deployment of stress granules in myeloid malignancies. Mol Cell 2022;82:1107–22.e7. https://doi.org/10.1016/j.molcel.2022.02.025.Search in Google Scholar PubMed PubMed Central
31. Zhang, J, Zhao, H, Wu, K, Peng, Y, Han, X, Zhang, H, et al.. Knockdown of spliceosome U2AF1 significantly inhibits the development of human erythroid cells. J Cell Mol Med 2019;23:5076–86. https://doi.org/10.1111/jcmm.14370.Search in Google Scholar PubMed PubMed Central
32. Shiozawa, Y, Malcovati, L, Galli, A, Sato-Otsubo, A, Kataoka, K, Sato, Y, et al.. Aberrant splicing and defective mRNA production induced by somatic spliceosome mutations in myelodysplasia. Nat Commun 2018;9:3649. https://doi.org/10.1038/s41467-018-06063-x.Search in Google Scholar PubMed PubMed Central
33. Masaki, S, Ikeda, S, Hata, A, Shiozawa, Y, Kon, A, Ogawa, S, et al.. Myelodysplastic syndrome-associated SRSF2 mutations cause splicing changes by altering binding motif sequences. Front Genet 2019;10:338. https://doi.org/10.3389/fgene.2019.00338.Search in Google Scholar PubMed PubMed Central
34. Yang, L, Rau, R, Goodell, MA. DNMT3A in haematological malignancies. Nat Rev Cancer 2015;15:152–65. https://doi.org/10.1038/nrc3895.Search in Google Scholar PubMed PubMed Central
35. Yamazaki, J, Jelinek, J, Lu, Y, Cesaroni, M, Madzo, J, Neumann, F, et al.. TET2 mutations affect non-CpG island DNA methylation at enhancers and transcription factor-binding sites in chronic myelomonocytic leukemia. Cancer Res 2015;75:2833–43. https://doi.org/10.1158/0008-5472.can-14-0739.Search in Google Scholar PubMed PubMed Central
36. Huerga Encabo, H, Aramburu, IV, Garcia-Albornoz, M, Piganeau, M, Wood, H, Song, A, et al.. Loss of TET2 in human hematopoietic stem cells alters the development and function of neutrophils. Cell Stem Cell 2023;30:781–99.e9. https://doi.org/10.1016/j.stem.2023.05.004.Search in Google Scholar PubMed
37. Wang, H, Guo, Y, Dong, Z, Li, T, Xie, X, Wan, D, et al.. Differential U2AF1 mutation sites, burden and co-mutation genes can predict prognosis in patients with myelodysplastic syndrome. Sci Rep 2020;10:18622. https://doi.org/10.1038/s41598-020-74744-z.Search in Google Scholar PubMed PubMed Central
38. Yip, BH, Steeples, V, Repapi, E, Armstrong, RN, Llorian, M, Roy, S, et al.. The U2AF1S34F mutation induces lineage-specific splicing alterations in myelodysplastic syndromes. J Clin Invest 2017;127:2206–21. https://doi.org/10.1172/jci96202.Search in Google Scholar PubMed PubMed Central
© 2025 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Editorial
- Can a digital smear review be helpful in the routine haematology laboratory?
- Original Articles
- The impact of mutational burden, spliceosome and epigenetic regulator mutations on transfusion dependency in dysplastic neoplasms
- Improving turn-around times in low-throughput distributed hematology laboratory settings with the CellaVision® DC-1 instrument
- Hematology instruments don’t speak the same language: a comparison study between flagging messages of sysmex XN-1000 and alinity H
- Platelet clump assessment using the Cellavision peripherical blood application – do we need manual microscopy?
- Short Communication
- Development of a peripheral blood morphology proficiency assessment program using the CellaVision® Proficiency Software
- Images From the Medical Laboratory
- Giant granules in white blood cells
Articles in the same Issue
- Frontmatter
- Editorial
- Can a digital smear review be helpful in the routine haematology laboratory?
- Original Articles
- The impact of mutational burden, spliceosome and epigenetic regulator mutations on transfusion dependency in dysplastic neoplasms
- Improving turn-around times in low-throughput distributed hematology laboratory settings with the CellaVision® DC-1 instrument
- Hematology instruments don’t speak the same language: a comparison study between flagging messages of sysmex XN-1000 and alinity H
- Platelet clump assessment using the Cellavision peripherical blood application – do we need manual microscopy?
- Short Communication
- Development of a peripheral blood morphology proficiency assessment program using the CellaVision® Proficiency Software
- Images From the Medical Laboratory
- Giant granules in white blood cells