scholarly journals Molecular Targeted Therapy in Myelodysplastic Syndromes: New Options for Tailored Treatments

Cancers ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 784
Author(s):  
Simona Pagliuca ◽  
Carmelo Gurnari ◽  
Valeria Visconte

Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic disorders characterized by ineffective hematopoiesis, progressive cytopenias and increased risk of transformation to acute myeloid leukemia. The improved understanding of the underlying biology and genetics of MDS has led to better disease and risk classification, paving the way for novel therapeutic opportunities. Indeed, we now have a vast pipeline of targeted agents under pre-clinical and clinical development, potentially able to modify the natural history of the diverse disease spectrum of MDS. Here, we review the latest therapeutic approaches (investigational and approved agents) for MDS treatment. A deep insight will be given to molecularly targeted therapies by reviewing new agents for individualized precision medicine.

2019 ◽  
Vol 55 (1) ◽  
pp. 35-42
Author(s):  
Beata Celuch ◽  
Iwona Urbanowicz ◽  
Jadwiga Nowicka ◽  
Wiesława Nahaczewska ◽  
Iwona Bil-Lula

Myelodysplastic syndromes (MDS) are a heterogeneous group of hematopoietic neoplastic diseases characterized by inefficient hematopoiesis, resistant peripheral cytopenias and an increased risk of transformation to acute myeloid leukemia. They may exist as primary forms, which most often are accompanied by molecular and cytogenetic changes or secondary forms, among others after chemotherapy or other cancers. MDS diagnostics are multi-stage and time-consuming. Includes multidirectional examination of peripheral blood and bone marrow for cytomorphology, cytogenetics, molecular disorders, immunohistopatology and immunophenotyping. The evolution of molecular changes in the course of MDS makes the clinical picture and laboratory parameters change over time, which requires constant updating of medical knowledge and high competences from cytomorphologists and histopathologists. The development of hematooncological diagnostics resulted in updating the MDS classification in 2016. The aging population will undoubtedly increase the incidence of myelodysplastic syndromes, which will be one of the most demanding diagnostic and clinical problems for haematologists and laboratory diagnostics in the near future.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1675-1675
Author(s):  
Anna Brady ◽  
Sarah Gibson ◽  
Lisa Rybicki ◽  
Eric Hsi ◽  
Edward Copelan ◽  
...  

Abstract Abstract 1675 Background: Acute myeloid leukemia (AML) is characterized by rapid growth, resistance to therapy, and poor overall survival. Clinical and biologic prognostic markers can help define pathogenesis, guide treatment, and identify novel therapies. Phosphorylated signal transducer and activator of transcription 5 (pSTAT5) is one such potential marker. The transcription factor STAT5 regulates many aspects of cell growth, survival, and differentiation. Constitutive activation of STAT5 (by phosphorylation) has been identified in a number of malignancies, including AML. We investigated whether the level of pSTAT5 expression correlates with complete remission (CR) rates, progression-free survival (PFS), and overall survival (OS) in patients (pts) with newly diagnosed AML. Methods: From 1999 to 2005, all adult pts with newly diagnosed AML (WHO criteria) receiving induction chemotherapy and with an available diagnostic bone marrow biopsy performed at our institution were evaluated. B5-fixed bone marrow core biopsies were reviewed for areas with the highest concentration of blasts. A tissue microarray was constructed using 1 mm cores. The cores were arrayed in duplicate in the majority of samples. Immunohistochemistry was performed for pSTAT5 with anti-pSTAT5 a/b Y695/99 mouse monoclonal antibody (AX1; Advantex Bioreagents) using automated stainers and heat-induced epitope retrieval. In each case, five hundred blasts were counted. The percentage of cells staining positive for pSTAT5 expression was determined by a pathologist blinded to clinical results. Standard metaphase karyotypes were classified into cytogenetic (CG) risk groups by CALGB criteria. Cox proportional hazards analysis was used to identify univariate and multivariate prognostic factors for CR, PFS, and OS, including age at diagnosis, history of an antecedent hematologic disorder (AHD), WBC at diagnosis, pSTAT5 expression, and CG risk group. Results: Adequate tissue and clinical data were available in 112 pts. The median age was 57 years, and median WBC at diagnosis 12.0 K/ μL. Twenty six percent of pts had favorable CG, 41% intermediate risk, 27% high risk, and 6% other (unknown or could not be classified). Nineteen percent of pts had an AHD. pSTAT5 expression was absent in 58% of pts. The remainder of the pts had: 1–5% pSTAT5 (25% of pts), 10% pSTAT5 (11% of pts), 20% pSTAT5 (4% of pts), 30% pSTAT5 (1% of pts), and 50% pSTAT5 (2% of pts). Seventy percent of all pts achieved a CR following induction chemotherapy. Sixty-four percent of pts received post-remission chemotherapy, 3.6% an autologous transplant, and 13.6% an allogeneic transplant in first CR. Median PFS and OS were 9.6 months and 16.0 months, respectively. On univariate analysis, age, history of AHD, WBC at diagnosis, CG risk, and any pSTAT5 expression were prognostic factors for PFS and OS. In multivariable analyses controlling for the above prognostic factors, pSTAT5 expression > 0 was also significantly associated with an increased risk of death (HR 1.96, 95% CI 1.19–3.23, p=0.008), progression or death (HR 1.64, 95% CI 1.01–2.66, p=0.046), and relapse after achieving CR (HR 2.31, 95% CI 1.16–4.63, p=0.018). pSTAT5 expression was not a predictor of achievement of CR. Conclusions: pSTAT5 expression in newly diagnosed adults with AML is associated with a decreased PFS, decreased OS, and increased risk of relapse. Validation of its prognostic value requires additional study. Agents targeting this signaling pathway might improve the outcome of pts with AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2905-2905
Author(s):  
Marta Cabezon ◽  
Joan Bargay ◽  
Blanca Xicoy ◽  
Laura Palomo ◽  
Sílvia Marcé ◽  
...  

Abstract INTRODUCTION: Myelodysplastic syndromes (MDS) are a group of myeloid neoplasms originated in hematopoietic stem cells, characterized by citopenias, dysplasia in one or more cell lines, ineffective hematopoiesis and an increased risk of progression to acute myeloid leukemia (AML). Treatment of MDS depends on subtype and prognostic category. DNA methyltranferase inhibitors are approved for high risk MDS. Over the past decade, the application of new high-throughput technologies to the study of MDS has led to the identification of several recurrently mutated genes. These include genes producing proteins involved in RNA splicing, DNA methylation, chromatin modification, transcription, DNA repair control, cohesin function, RAS pathway, and DNA replication. There is a significant overlap between the genes mutated commonly in MDS with those found in AML. Mutation status is not widely used to select treatment in MDS. The aim of this study is to define the mutational status of MDS and secondary AML (sAML) patients at diagnosis that have been treated with azacitidine (AZA) to see if it could help to discriminate which patients will respond from those who will not. MATERIAL AND METHODS: A prospective study was performed on 36 patients with MDS and sAML treated with AZA. Genomic DNA was obtained from bone marrow at diagnosis. SeqCap EZ and KAPA Library Preparation Kit (Roche) reagents have been used to enrich DNA of 83 genes implicated in myeloid neoplasm. The customized panel has been analyzed in MiSeq Illumina platform with 150bp paired-end reads. Samples were preliminary analyzed using Illumina MiSeq Reporter and Variant Studio softwares. Data from response to treatment and survival have been collected from all patients. RESULTS:The mean depth of the targeted resequencing per base was 685-fold. After filtering all the variations obtained for quality, biological consequence and discard the known SNPs, we have obtained 162 variations, including 145 single nucleotide variants (SNV) and 17 insertions/deletions. All patients harbored at least 1 alteration with a mean of 4.5 variants per sample. The average of alterations detected in each cytological category can be observed in Table 1.Table 1.Average abnormalities detected by cytological category.Nº patientsAverage of alterations detected for patient (range)sAML104,8 (1-8)RAEB-274,9 (2-8)RAEB-1123,7 (1-6)RCDM54,4 (3-7)RCDM-RS16RARs11The most frequent altered genes have been TP53, TET2 and DNMT3A. The numbers of variations detected for each gene are represented in Table 2.Complete results, including correlation with treatment response will be presented in the meeting.Table 2.Number of variations in each gene.GeneNº of variations foundNº of diferent variationsNº of patients with variationsFrequency of variationsTP5322191952,8%TET214101027,8%DNMT3A88822,2%CREBBP75719,4%SRSF271719,4%ASXL165616,7%U2AF162616,7%EP30053513,9%STAG255513,9%CUX144411,1%ETV643411,1%MLL (KMT2A)43411,1%RUNX14438,3%BCOR3338,3%CDH133338,3%CTNNA13238,3%EZH23338,3%GCAT3338,3%MLL2 (KMT2D)3338,3%NF13338,3%PDGFRB3338,3%SH2B33338,3%TGM23238,3%UMODL13338,3%CEBPA2125,6%CSF3R2225,6%GATA22125,6%PHLPP12225,6%RAD212225,6%SF3B12125,6%SUZ122225,6%TIMM502125,6%Others*1112,8%*ABL1, BCORL1, CALR, CDH3, IDH2, KRAS, LUC7L2, NPM1, NRAS, PHF6, SF3A1, SFPQ, SMC3, TERT, WT1, ZRSR2. CONCLUSIONS: Targeted deep-sequencing technique is a good tool to study mutational profile in MDS and sAML. SNV are the most frequent type of alteration found in our cohort. The patients with sAML and RAEB-2 present more variations than patients with RAEB-1. The rest of groups are less representing to be evaluated. The most affected genes match with those described in the literature, with some exceptions that need to be studied in more detail. We expect to predict in advance which patients are going to respond when we study the correlation of mutational analysis with treatment response. Acknowledgments: Instituto de Salud Carlos III, Ministerio de Sanidad y Consumo, Spain (PI 11/02519); 2014 SGR225 (GRE) Generalitat de Catalunya; Fundació Josep Carreras, Obra Social "La Caixa" and Celgene Spain. Diana Domínguez for her technical assistance Disclosures Valcarcel: Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; GSK: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Honoraria, Speakers Bureau; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.


2019 ◽  
Vol 3 (7) ◽  
pp. 1084-1091 ◽  
Author(s):  
Adrian G. Minson ◽  
Katherine Cummins ◽  
Lucy Fox ◽  
Ben Costello ◽  
David Yeung ◽  
...  

Abstract Although second-generation tyrosine kinase inhibitors (TKIs) show superiority in achieving deep molecular responses in chronic myeloid leukemia in chronic phase (CML-CP) compared with imatinib, the differing adverse effect (AE) profiles need consideration when deciding the best drug for individual patients. Long-term data from randomized trials of nilotinib demonstrate an increased risk of vascular AEs (VAEs) compared with other TKIs, although the natural history of these events in response to dose modifications or cessation has not been fully characterized. We retrospectively reviewed the incidence of nilotinib-associated AEs in 220 patients with CML-CP at 17 Australian institutions. Overall, AEs of any grade were reported in 95 patients (43%) and prompted nilotinib cessation in 46 (21%). VAEs occurred in 26 patients (12%), with an incidence of 4.1 events per 100 patient-years. Multivariate analysis identified age (P = .022) and dyslipidemia (P = .007) as independent variables for their development. There was 1 fatal first VAE, whereas the remaining patients either continued nilotinib (14 patients) or stopped it immediately (11 patients). Recurrent VAEs were associated with ongoing therapy in 7 of 14 who continued (with 2 fatal VAEs) vs 1 of 11 who discontinued (P = .04). Nineteen of the 23 evaluable patients surviving a VAE ultimately stopped nilotinib, of whom 14 received an alternative TKI. Dose reduction or cessation because of VAEs did not adversely affect maintenance of major molecular response. These findings demonstrate that in contrast to other AEs, VAEs are ideally managed with nilotinib cessation because of the increased risk of additional events with its ongoing use.


2012 ◽  
Vol 30 (2) ◽  
pp. 179-183 ◽  
Author(s):  
Lynn R. Goldin ◽  
Sigurdur Y. Kristinsson ◽  
Xueying Sharon Liang ◽  
Åsa R. Derolf ◽  
Ola Landgren ◽  
...  

Purpose Apart from rare pedigrees with multiple cases of acute myeloid leukemia (AML), there is limited data on familial aggregation of AML and myelodysplastic syndromes (MDSs) in the population. Patients and Methods Swedish population-based registry data were used to evaluate risk of AML, MDS, and other malignancies among 24,573 first-degree relatives of 6,962 patients with AML and 1,388 patients with MDS compared with 106,224 first-degree relatives of matched controls. We used a marginal survival model to calculate familial aggregation. Results AML and/or MDS did not aggregate significantly in relatives of patients with AML. There was a modest risk ratio (RR, 1.3; 95% CI, 0.9 to 1.8) in myeloproliferative/myeloid malignancies combined. The risks for any hematologic or any solid tumor were modestly but significantly increased. Relatives of patients with MDS did not show an increased risk for any hematologic tumors. In contrast, we found a significantly increased risk (RR, 6.5; 95% CI, 1.1 to 38.0) of AML/MDS and of all myeloid malignancies combined (RR, 3.1; 95% CI, 1.0 to 9.8) among relatives of patients diagnosed at younger than age 21 years. Conclusion We did not find evidence for familial aggregation of the severe end of the spectrum of myeloid malignancies (AML and MDS). The risks of myeloproliferative neoplasms were modestly increased with trends toward significance, suggesting a possible role of inheritance. In contrast, although limited in sample size, relatives of young patients with AML were at increased risk of AML/MDS, suggesting that germline genes may play a stronger role in these patients. The increased risk of all hematologic malignancies and of solid tumors among relatives of patients with AML suggests that genes for malignancy in general and/or other environmental factors may be shared.


2011 ◽  
Vol 29 (21) ◽  
pp. 2897-2903 ◽  
Author(s):  
Sigurdur Y. Kristinsson ◽  
Magnus Björkholm ◽  
Malin Hultcrantz ◽  
Åsa R. Derolf ◽  
Ola Landgren ◽  
...  

Purpose Patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) often present with infections, but there are little data to assess whether a personal history of selected infections may act as pathogenic triggers. To additionally expand our knowledge on the role of immune stimulation in the causation of AML and MDS, we have conducted a large, population-based study to evaluate the risk of AML and MDS associated with a prior history of a broad range of infections or autoimmune diseases. Patients and Methods By using population-based central registries in Sweden, we included 9,219 patients with AML, 1,662 patients with MDS, and 42,878 matched controls. We used logistic regression to calculate odds ratios (ORs) and 95% CIs for the association of AML or MDS with infectious and/or autoimmune diseases. Results Overall, a history of any infectious disease was associated with a significantly increased risk of both AML (OR, 1.3; 95% CI, 1.2 to 1.4) and MDS (OR, 1.3; 95% CI, 1.1 to 1.5). These associations were significant even when we limited infections to those occurring 3 or more years before AML/MDS. A previous history of any autoimmune disease was associated with a 1.7-fold (95% CI, 1.5 to 1.9) increased risk for AML and 2.1-fold (95% CI, 1.7 to 2.6) increased risk for MDS. A large range of conditions were each significantly associated with AML and MDS. Conclusion Our novel findings indicate that chronic immune stimulation acts as a trigger for AML/MDS development. The underlying mechanisms may also be due to a common genetic predisposition or an effect of treatment for infections/autoimmune conditions.


Leukemia ◽  
2021 ◽  
Author(s):  
Uwe Platzbecker ◽  
Anne Sophie Kubasch ◽  
Collin Homer-Bouthiette ◽  
Thomas Prebet

AbstractMyelodysplastic syndromes (MDS) represent a heterogeneous group of myeloid neoplasms that are characterized by ineffective hematopoiesis, variable cytopenias, and a risk of progression to acute myeloid leukemia. Most patients with MDS are affected by anemia and anemia-related symptoms, which negatively impact their quality of life. While many patients with MDS have lower-risk disease and are managed by existing treatments, there currently is no clear standard of care for many patients. For patients with higher-risk disease, the treatment priority is changing the natural history of the disease by delaying disease progression to acute myeloid leukemia and improving overall survival. However, existing treatments for MDS are generally not curative and many patients experience relapse or resistance to first-line treatment. Thus, there remains an unmet need for new, more effective but tolerable strategies to manage MDS. Recent advances in molecular diagnostics have improved our understanding of the pathogenesis of MDS, and it is becoming clear that the diverse nature of genetic abnormalities that drive MDS demands a complex and personalized treatment approach. This review will discuss some of the challenges related to the current MDS treatment landscape, as well as new approaches currently in development.


2019 ◽  
Vol 17 (11.5) ◽  
pp. 1444-1447
Author(s):  
Rafael Bejar

Often unrecognized and underdiagnosed, myelodysplastic syndromes (MDS) are a rare group of cancers in which the bone marrow fails to produce sufficient healthy blood cells. Although patients with lower-risk MDS can live for >5 years, those with high-risk disease that evolves into acute myeloid leukemia is associated with significantly lower overall survival. At the NCCN 2019 Annual Congress: Hematologic Malignancies, Dr. Rafael Bejar summarized current standard treatment options for patients with MDS and discussed the importance of genetic testing to identify mutations that may impact treatment. Finally, Dr. Bejar described emerging personalized treatment strategies for the management of this disease.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 823-823
Author(s):  
Radhika Gangaraju ◽  
Yanjun Chen ◽  
Lindsey Hageman ◽  
Jessica Wu ◽  
Liton F. Francisco ◽  
...  

Abstract BACKGROUND: Patients undergoing BMT are uniquely vulnerable to venous thromboembolism (VTE) due to high-intensity therapeutic exposures, prolonged hospitalizations (and attendant immobility), a high burden of comorbidities, and the pro-inflammatory state induced by graft-versus-host disease (GvHD) in allogeneic BMT recipients. An assessment of the long-term risk of VTE in BMT survivors and subsequent cause-specific late mortality remains unstudied. We used the BMTSS to address this gap. METHODS: In patients with hematologic malignancy treated with BMT at 3 sites in US, had survived ≥2y after BMT, were alive and ≥18yo at BMTSS survey completion, we examined: i) incidence/risk factors for VTE; ii) subsequent mortality by VTE status. The survey covered sociodemographics, tobacco use, diagnosis by a healthcare provider of specific chronic health conditions, including VTE, relapse, or development of new cancer and medication use. The outcome of interest (VTE) was defined as self-report of VTE diagnosed by a health care provider, supplemented by history of anticoagulant use. BMTSS survey was administered over two time periods: i) Survey completed between 2000 and 2004: 1,022 patients transplanted between 1974 and 1998 and survived ≥2y (original BMTSS cohort; participation rate: 63%). This cohort was followed for a median of 15.1y (range: 0.04-17.9) after survey completion, to determine the association between VTE and subsequent mortality using National Death Index; ii) Survey completed between 2014 and 2017: 1,711 patients transplanted between 1974 and 2010 and survived ≥2y (expanded BMTSS cohort; participation rate: 68%) - this cohort was used to determine demographic/clinical variables associated with VTE risk. A cohort of 644 siblings also completed the survey and served as a comparison group. Human Subjects Committee at participating sites approved the protocol. RESULTS: Risk of VTE: Of the 1,711 participants, 891 (52.1%) had received an allogeneic BMT; 53.5% were males and 78.8% were non-Hispanic whites; median age at BMT was 48y. The cohort was followed for a median of 10.9y (3.8-40.6). BMT survivors were at a 2.8-fold higher risk of VTE as compared to siblings (95%CI:1.7-4.5, p<0.0001), after adjusting for sociodemographics. Conditional on surviving ≥2y after BMT, the cumulative incidence of VTE was 6.9±0.9% at 10y (Fig 1), with similar rates in autologous (6.1±1.1%) and allogeneic BMT recipients (7.5±1.5%%, p=0.5). Risk of VTE in allogeneic BMT recipients: History of chronic GvHD (HR=3.6, 95%CI:2.2-6.0, p<0.0001), primary diagnosis of acute myeloid leukemia/myelodysplasia (HR=1.9, 95%CI:1.0-3.7, p=0.05) or non-Hodgkin lymphoma (NHL) (HR=2.3, 95%CI:1.1-4.8, p=0.02) (ref: chronic myeloid leukemia), male sex (HR=1.6, 95%CI:1.0-2.6, p=0.05), and older age at BMT (HR=1.03/y, 95%CI:1.0-1.1, p=0.002), were associated with increased VTE risk. Risk of VTE in autologous BMT recipients: Diagnosis of a plasma cell disorder (HR=2.4, 95%CI: 1.3-4.2, p=0.004) (ref: NHL) and annual house hold income <$50,000 (HR=2.0, 95%CI 1.2-3.6, p=0.02) (ref: income >$50,000) were associated with increased VTE risk. Of note, relapse of primary disease or development of new cancer were not associated with increased VTE risk in either autologous or allogeneic BMT patients. Subsequent mortality: In this cohort of 1,022 BMT survivors, median age at BMT was 34.9y; 55% had received allogeneic BMT. The overall survival among patients with and without VTE was 50.4% vs. 72.3%, respectively at 15y from survey completion, p<0.0001 (Fig 2), yielding a higher risk of subsequent mortality in VTE patients when compared to those without VTE (HR=1.6, 95%CI:1.1-2.3, p=0.02) after adjusting for relevant clinical/demographic predictors, relapse of primary cancer and development of new cancer. The VTE-associated mortality risk was primarily due to non-relapse mortality (NRM: HR=1.7, 95%CI:1.1-2.5, p=0.02) and not due to relapse (HR=0.7, 95%CI, 0.2-1.8, p=0.5). Leading causes of NRM among those with VTE included infection (15.5%), cardiac (8.5%) and subsequent neoplasms (7%). CONCLUSION: BMT survivors are at a 2.8-fold increased risk of developing VTE when compared with a non-BMT sibling comparison group. Those with VTE are at an increased risk of subsequent NRM. Development of risk prediction models to identify BMT survivors at highest risk would facilitate targeted thromboprophylaxis. Disclosures Weisdorf: Equillium: Consultancy; SL Behring: Consultancy; Seattle Genetics: Consultancy; FATE: Consultancy; Pharmacyclics: Consultancy. Forman:Mustang Therapeutics: Other: Licensing Agreement, Patents & Royalties, Research Funding.


2011 ◽  
Vol 29 (17) ◽  
pp. 2410-2415 ◽  
Author(s):  
Magnus Björkholm ◽  
Åsa R. Derolf ◽  
Malin Hultcrantz ◽  
Sigurdur Y. Kristinsson ◽  
Charlotta Ekstrand ◽  
...  

Purpose Patients with myeloproliferative neoplasms (MPNs), including polycythemia vera, essential thrombocythemia, and primary myelofibrosis, have a propensity to develop acute myeloid leukemia (AML) and myelodysplastic syndromes (MDSs). Using population-based data from Sweden, we assessed the role of MPN treatment and subsequent AML/MDS risk with special focus on the leukemogenic potential of hydroxyurea (HU). Methods On the basis of a nationwide MPN cohort (N = 11,039), we conducted a nested case-control study, including 162 patients (153 and nine with subsequent AML and MDS diagnosis, respectively) and 242 matched controls. We obtained clinical and MPN treatment data for all patients. Using logistic regression, we calculated odds ratios (ORs) as measures of AML/MDS risk. Results Forty-one (25%) of 162 patients with MPNs with AML/MDS development were never exposed to alkylating agents, radioactive phosphorous (P32), or HU. Compared with patients with who were not exposed to HU, the ORs for 1 to 499 g, 500 to 999 g, more than 1,000 g of HU were 1.5 (95% CI, 0.6 to 2.4), 1.4 (95% CI, 0.6 to 3.4), and 1.3 (95% CI, 0.5 to 3.3), respectively, for AML/MDS development (not significant). Patients with MPNs who received P32 greater than 1,000 MBq and alkylators greater than 1 g had a 4.6-fold (95% CI, 2.1 to 9.8; P = .002) and 3.4-fold (95% CI, 1.1 to 10.6; P = .015) increased risk of AML/MDS, respectively. Patients receiving two or more cytoreductive treatments had a 2.9-fold (95% CI, 1.4 to 5.9) increased risk of transformation. Conclusion The risk of AML/MDS development after MPN diagnosis was significantly associated with high exposures of P32 and alkylators but not with HU treatment. Twenty-five percent of patients with MPNs who developed AML/MDS were not exposed to cytotoxic therapy, supporting a major role for nontreatment-related factors.


Sign in / Sign up

Export Citation Format

Share Document