scholarly journals 5-Azacytidine Is Effective for Targeting Leukemia-Initiating Cells in Juvenile Myelomonocytic Leukemia

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4342-4342
Author(s):  
Christopher Felix Krombholz ◽  
Lorena Gallego Villar ◽  
Pritam Kumar Panda ◽  
Sushree Sangita Sahoo ◽  
Marcin W. Wlodarski ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) is a myeloproliferative neoplasm of young children that originates from early hematopoietic stem/progenitor cells. We have previously developed an in vivo disease model using xenotransplantation of primary JMML cells into Rag2-/-γc-/- mice (Haematologica 2016;101:597). The model reproduces a characteristic JMML phenotype including myelomonocytic proliferation, hepatosplenomegaly, and lung infiltration. Case-specific driver mutations and DNA methylation patterns are unchanged after xenologous engraftment, indicating their origin in leukemia-initiating cells. We and others recently discovered a tight link between prognosis and differential DNA methylation in JMML (Nat Commun 2017;8:2126; Nat Commun 2017;8:2127; Blood 2018;131:1576). We also reported that the DNA methyltransferase inhibitor 5-azacytidine (azacitidine, 5AC) has unprecedented clinical activity in JMML and induces complete or partial remissions before allogeneic HSCT (Blood 2015;125:2311). Cytosine arabinoside (araC) is a structurally related nucleoside which is commonly used for cytoreduction in JMML but lacks the ability to induce remissions. Here we employed the xenotransplantation model to investigate the antileukemic activity and epigenetic effects of 5AC on JMML in comparison to araC. After eight weeks of leukemic expansion, 15 xenograft mice were treated with two cycles of 5AC (3 mg/kg/d x 5 days every two weeks). Control groups included mice treated with araC 20 mg/kg/d (N=15) or carrier solution (0.9% NaCl, N=20). The experimental animals maintained stable body weight, and no major toxicity on murine hematopoiesis was observed. 5AC and araC exhibited antileukemic activity and substantially reduced the human JMML cell content in bone marrow, spleen, liver, and lung. However, we noted that CD34+ stem/progenitor cells within the human leukemia population were depleted after treatment with 5AC but not after araC (5AC, 20.2% +/- 7.3%; araC, 35.6% +/- 6.1 %; carrier, 39.4% +/- 3.5%; p<0.01). To demonstrate that the selective reduction of CD34+ cells impaired the leukemia-initiating capacity of the xenograft, we treated a subsequent series of mice as above and retransplanted the bone marrow into secondary recipient mice. JMML cells obtained from 5AC-treated primary recipients sustained engraftment in only one of 9 secondary recipients at 30 weeks after retransplantation whereas JMML xenografts treated with araC or carrier engrafted in 8/13 or 4/8 secondary mice, respectively (p=0.03). We then examined the genome-wide DNA methylation in 5AC-treated xenografts (N=5) using Infinium 450K arrays. The JMML genomes exhibited global and profound DNA demethylation with near-complete loss of fully methylated CpG sites. A focused analysis of approximately 5,000 CpG sites with JMML-specific methylation illustrated that the profiles of 5AC-treated JMML cells were more similar to healthy human CD34+ cells than untreated JMML cells. As expected, no change in DNA methylation was observed in xenografts treated with araC. Next we studied the early effects of 5AC on the transcriptome and epigenome of JMML. Xenograft mice were treated as above, and JMML cells were harvested from bone marrow on days 0, 2, 4, and 6. RNA sequencing readily identified non-random changes in gene transcription that progressed over time from days 2 to 6 and were reproducible across replicate mice. Between days 0 and 6 we observed >2fold upregulation of 856 transcripts and downregulation of 958 transcripts (<0.01 false discovery rate, multitest-corrected). CpG-rich 5' regions (putative promoters) of corresponding genes were invariably demethylated. Gene Ontology enrichment analysis linked upregulated transcripts to myeloid differentiation whereas downregulated transcripts were involved in nucleosome assembly/organization and chromatin silencing. In summary, the xenograft experiments highlight the therapeutic potential of 5AC in JMML and thus encourage the further clinical development of epigenetic therapy with hypomethylating agents for this disease. Disclosures Niemeyer: Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2038-2038
Author(s):  
Hein Than ◽  
Naoto Nakamichi ◽  
Anthony D. Pomicter ◽  
John O'Shea ◽  
Orlando Antelope ◽  
...  

Abstract Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) are complex clonal hematopoietic stem cell malignancies with overlapping dysplastic and proliferative features. Genomic analyses have charted the somatic mutation spectrum of MDS/MPN and revealed a major role for epigenetic dysregulation in their pathogenesis. No disease-modifying therapies are currently available, as progress has been hampered by a lack of genetically faithful in vivo model systems suitable for the preclinical development of new strategies. Yoshimi et al (Blood. 2017;130:397-407) recently showed that patients' chronic myelomonocytic leukemia (CMML) and juvenile myelomonocytic leukemia (JMML) cells transplanted into NOD/SCID-IL2Rγ-/-mice expressing human IL3, GM-CSF and SCF transgenes (NSG-3GS mice) produced xenografts that had mutations characteristic of the input cells. Since we had demonstrated a superior level of chimerism achieved from transplants of normal human CD34+cord blood cells in SirpaNOD/Rag1-/-/IL2rγc-/-/W41/41mice with c-KIT deficiency (with an otherwise mixed NOD-C57Bl/6 background - SRG-W41 mice) compared to conventional NSG or NRG hosts (Miller et al. Exp Hematol. 2017;48:41-49), it was of interest to explore their use as hosts of samples from patients with MDS/MPN: CMML, atypical chronic myeloid leukemia (aCML) and secondary acute myeloid leukemia (sAML) progressed from CMML or aCML. Heparinized blood or bone marrow samples were obtained from patients treated at Huntsman Cancer Institute after informed consent. Diagnoses included CMML (n=5), aCML (n=2), and sAML (n=2). Unseparated cells were shipped by overnight courier to Vancouver and CD34+cells isolated on the same day were injected intravenously into sub-lethally irradiated female NRG mice or SRG-W41 mice, or in some cases the same sex and strains also carrying the human 3GS transgenes (NRG-3GS or SRG-W41-3GS mice) in accordance with British Columbia Cancer Agency institutional guidelines. Occasionally when mice were not immediately available, or large numbers of cells were available, cells were viably cryopreserved and transplanted later after thawing. Mice were observed for up to 36 weeks after xenotransplantation with .05 to 1.1x106 human CD34+cells. Engraftment of human CD45+cells in xenografts was evaluated by immunophenotyping, and a median of 90% human chimerism (range: 1% - 95%) was achieved at the time of bone marrow harvest from xenografts. Variant allele frequencies (VAF) were determined in genomic DNA extracted from both the patient samples (CD34+cells) and matching fluorescence-activated cells (FACS)-sorted human CD45+cells (hCD45+cells) purified from xenografts (1-5 xenografts per patient sample). DNA samples were subjected to PCR amplification with extension primers and analyzed using a MALDI-TOF mass spectrometer (MassArray, Agena Bioscience, San Diego, CA). Each mutation call was assigned by the software based on the molecular weight of the extended primer. Analysis of hCD45+cells from eight xenograft samples so far demonstrated a strong correlation of VAF between the patient samples and hCD45+cells from xenografts, in both SRG-W41-3GS (R2=0.94, p<0.01) and NRG-3GS (R2=0.97, p<0.01) models (Figure 1). This tight correlation of VAF was illustrated in hCD45+cells from xenografts transplanted with CMML, aCML or sAML cells. The majority of mutations detected were those in epigenetic regulator genes, such as ASXL1, EZH2 and TET2. No significant difference in VAF was observed between CD34+and CD34- compartments within the hCD45+cells. Additional samples, including specimens from patients with the related myeloproliferative neoplasm, chronic neutrophilic leukemia (CNL) are being analyzed and will be presented. These findings demonstrate the utility of SRG-W41-3GS as well as NRG-3GS as receptive hosts of primary human MDS/MPN cells with genetic evidence of their growth in these mice closely recapitulating the mutational profiles of the transplanted cells. These new strains may facilitate the development of functional screening and pre-clinical testing of novel therapeutic strategies for a range of human MDS/MPN and related myeloid disorders. Disclosures Deininger: Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Blueprint: Consultancy.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2818-2818
Author(s):  
Silvia Bresolin ◽  
Chiara Frasson ◽  
Marco Giordan ◽  
Giuseppe Gaipa ◽  
Cristina Bugarin ◽  
...  

Abstract Abstract 2818 Juvenile myelomonocytic leukemia (JMML) is a rare myeloproliferative disorder of early childhood characterized by excessive proliferation of granulocytic and myelomonocytic cells. So far, about 85–90% of patients harbor mutations in genes (RAS, PTPN11, CBL, NF1) involved in the RAS signaling pathway and 25% of cases show monosomy of chromosome 7. In 10–15% of patients with JMML no recurrent mutations have been identified. In JMML, evidence of mutated cells in the myeloid lineage is well known; however, to which extend cell populations of other hematopoietic lineages harbor the same mutation is not well understood. We calculated the mutated allele frequencies of PTPN11, KRAS and CBL mutations of 10 patients with JMML using amplicon ultra deep sequencing 454 Roche Technology. KRAS and PTPN11 mutated allele frequencies in the total bone marrow (BM) varied in a range from 37.40% to 51.58%, and no statistical difference was identified in the variants allele read counts of the latter two genes. The only patient analyzed with a CBL mutation showed a mutated allele frequencies of nearly 100% pointing to homozygosity of the mutation and suggesting loss of the non-mutated CBL allele through somatic uniparental disomy of chromosomal region 11q23. Assuming occurrence of PTPN11 and KRAS mutations in heterozygosity (presence once per diploid genome), a mutated allele frequency of approximately 50% would imply that all cells were mutated (100% of cells). Along this line, our analysis of mutation frequencies, largely below 50%, pointed to a mosaicism of mutated and non-mutated cells in the BM of JMML patients. To further analyze the spectrum of mutations in BM of JMML, we sorted different hematopoietic subpopulations from 6 patients carrying a PTPN11 mutation. We choose different combinations of surface markers: CD34 and CD38 for stem and progenitor cells, CD33, CD14, CD15 and CD16, CD11b for myeloid lineages at different stages of maturation, CD3 and CD19 for T and B cells, respectively. Analysis of sorted cells revealed high mutated allele frequencies for all maturation stages of the myeloid lineage with more than 90% heterozygous mutated cells. Presence of high mutated allele frequencies were also identified in hematopoietic stem and progenitors cells and in B-cell lineage. Interestingly, in the T-cell lineage, a low mutated allele frequency was detected ranging from 9.4% to 29,35%, this finding suggesting that about 20% to 60% of T cells in the BM harbor PTPN11 mutations. Furthermore, with the aim of detecting still unknown mutations, associated with JMML we performed whole exome sequencing of BM cells of 4 JMML patients lacking any of the currently known mutations. In conclusion all hematopoietic lineage cells carry PTPN11 mutations in different percentages, indicating a mosaic distribution of mutated and non-mutated cells in the whole BM of JMML patients. We studied the power function of a binomial test to understand the probability of detecting a real deviation of variant allele read count from the hypothetical 50%, as expected for 100% of mutated heterozygous alleles. Moreover we provided a sensitivity analysis of the possible p-values when the 50% hypothetical model is true. Also using this model, a scenario of mosaicism appeared in the various subpopulations of JMML BM. The presence of non-mutated apparently healthy cells in the BM of JMML patients may hold a promise for exploring therapies that could interfere only with the mutated population sparing healthy cells in the myeloid progenitor cells populations. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4890-4890 ◽  
Author(s):  
Y. Lucy Liu ◽  
Shelly Y. Lensing ◽  
Yan Yan ◽  
Cody Webster ◽  
Peter D. Emanuel

Abstract Juvenile myelomonocytic leukemia (JMML) is a mixed myelodysplastic /myeloproliferative disorder (MDS/MPD). It occurs in infancy and young children with a progressive course leading to death within one year after diagnosis. This disease is characterized by monocytosis, leukocytosis, elevated fetal hemoglobin, hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF), a low percentage of myeloblasts in the bone marrow, and absence of the Philadelphia chromosome or the BCR/ABL fusion gene. Mutations or other abnormalities in RAS, NF1, PTPN11, and CBL have been linked to be responsible for the pathogenesis of JMML in up to 85% of cases. Treatment is very difficult in JMML, and only allogeneic stem cell transplantation (SCT) can extend survival. However, the relapse rate from allogeneic SCT is inordinately high in JMML (28-55%), with 5-year disease-free survival rates of 25-40%. JMML occurs in an age-range when genes are actively being turned on or off in children in adaption to the oxygenized environment after birth. Epigenetics plays a key role in this developmental plasticity. We previously reported hypermethylation on the promoter of PTEN in 77% of JMML patients, and decitabine, a DNA-hypomethylating reagent, significantly inhibited colony formation (CFU-GM) in JMML cells in vitro. In addition, other groups found that aberrant DNA methylation on promoters of BMP4, CALCA, CDKN2B, and RARB is significantly associated with poor prognosis in JMML. Taking together, these data suggest that epigenetic mechanisms may contribute to the pathogenesis of JMML. MicroRNAs (miRNAs) have been reported to play an important role in myeloid differentiation and activation. miRNA function is highly dependent on the cell type. Recently, we reported that miR-183 is overexpressed in JMML. Other groups have reported aberrant expression of miR-29a in acute myeloid leukemia and other cancers. Both miR-183 and miR-29a are located on chromosome 7q32 in humans, which is frequently disrupted in JMML. We hypothesized that miR-29a may be deregulated in JMML, and contribute to the aberrant epigenetic regulation in JMML. In order to test our hypothesis, we collected peripheral blood or bone marrow from 41 JMML patients and 14 normal individuals. Total RNAs were extracted from mononuclear cells (MNCs) using Trizol. We first evaluated the expression levels of miR-29a by using relative-quantitative real-time RT-PCR (qRT-PCR). We found that the expression levels (RQ) of miR-29a in patients are significantly lower than that in normal individuals (median 0.45 vs. 1.11, p< 0.001). By analysis of the expression levels of miR-29a together with previous data on miR-183 in these JMML patients and normal controls, we found that the RQ of miR-29a is inversely correlated with RQ of miR-183 (Spearman’s r=-51, p<0.001). This suggests that the expression of miR-29a is segregated from that of miR-183 in MNCs, although they are located only 1.1 million base-pairs apart on chromosome 7q32. When further evaluating the mRNA expression levels of miR-29a targeting genes by using qRT-PCR, we found that RQ of DNA methyltransferases 3A and 3B (DNMT3a and DNMT3b) were significantly increased in JMML patients’ samples in comparison with normal controls (median 5.16 vs. 1.00, p=0.001 for DNMT3a; median 3.45 vs. 1.08, p=0.001). Strikingly, the RQ of miR-29a was inversely correlated with the RQs of both DNMTs (Spearman’s r=-0.62 for DNMT3a; Spearman’s r=-0.79 for DNMT3b, p<0.001), which suggests that miR-29a plays a significant role in the regulation of DNMT 3a and DNMT3b in MNCs. In conclusion, we found that overexpression of DNMT3a and DNMT3b, the two key molecules in DNA methylation of epigenetic modification in developmental plasticity, is related to downregulation of miR-29a in JMML. In addition to our previous report on overexpression of miR-183 in JMML, this finding provides new insights into the role of miRNAs in the aberrant epigenetic regulation in JMML, and it may apply to other pediatric malignancies. Further investigation is ongoing to uncover the mechanism of down-regulated miR-29a in JMML. If the underexpression of miR-29a in JMML is due to the hypermethylation on miR-29a promoter, as reported in prostate cancer cells, demethylating agents, such as decitabine, or soybean products, isoflavone, could be potential drugs to treat JMML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1655-1655
Author(s):  
Christopher Felix Krombholz ◽  
Angelina Meier ◽  
Konrad Aumann ◽  
Silvia Fluhr ◽  
Matthias Kollek ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative disorder of early childhood with often fatal outcome. Despite many attempts to develop alternative treatment options allogeneic hematopoietic stem cell transplantation (HSCT) remains the only curative modality. In the past our group has linked the prognosis of JMML to differential DNA methylation patterns (Olk-Batz, Blood 2011;117:4871-80 and Poetsch, Epigenetics 2014;9:1252-60), suggesting a key role of epigenetic modifications in JMML pathophysiology. To overcome the lack of suitable preclinical JMML research models we have developed an ex vivo JMML xenotransplantation system using neonatal Rag2-/- gamma-c-/- mice. Transplantation of 1x106 primary JMML cells resulted in stable xenologous engraftment and reproduced a characteristic JMML phenotype including myelomonocytic expansion; infiltration of spleen, liver and, notably, lung; splenomegaly; and reduced survival (median 26 weeks). Persistent human engraftment and leukemic organ infiltration was confirmed by both flow cytometry and immunohistology. Ras pathway mutations present in xenotransplanted patient samples were invariably confirmed in engrafted tissues. In addition, the model sustained serial transplantations and can therefore be used to amplify scarce patient material. We first tested if DNA methylation patterns in JMML cells were stable even after xenologous engraftment because such stability would be a prerequisite if the model were to be used for preclinical investigation of DNA methyltransferase inhibitors. JMML cells before xenotransplantation and those retrieved from the bone marrow of engrafted mice were profiled for global CpG methylation using Illumina 450K arrays. DNA methylation patterns in JMML were patient-specific and surprisingly robust in functional regions over several months of engraftment time (on average, 0.29% of 30877 promoters and 0.25 % of 30725 intragenic regions were called as "differentially methylated" between source and xenograft; 0.2 β-value change cutoff). These findings confirm the suitability of the xenograft model to investigate JMML epigenetics and, more importantly, indicate that patient-specific epigenetic profiles originate in leukemia-initiating stem cells, reinforcing a fundamental role of these alterations in JMML biology. Our group recently published a retrospective case series demonstrating unprecedented clinical efficacy of the DNA methyltransferase inhibitor 5-azacytidine (5AC) to induce partial or complete remissions in JMML before allogeneic HSCT (Cseh, Blood 2015;125:2311-3). To further investigate the drug on the preclinical level we administered 5AC to Rag2-/- gamma-c-/- mice xenografted with primary JMML cells. After a leukemia establishment phase the mice were divided into treatment or mock groups and treated with 5AC (3mg/kg body weight i.p., N=6) or saline (N=6) for 2 cycles (1 dose daily for 5 days; 9 days of recovery). This regimen was tolerated well by the animals. We found that 5AC reduced JMML infiltration in all organs analyzed, with most pronounced effects in spleen (human CD45+ fraction of all CD45+ cells, 0.24% +/- 0.04% vs 39.78% +/- 10.72%; p<0.01) and lung (0.41% +/-0.18% vs 42.88% +/-8.42%; p<0.01). The proportion of early progenitor cells (CD34+) within the human leukemia population in murine bone marrow was dramatically reduced after 5AC treatment (7.89% +/-0.74% vs 32.65% +/-3.76%; p<0.01) while the amount of granulocytes increased simultaneously (44.90% +/-1.74% vs 9.35% +/-1.95%; p<0.01). These findings suggest a loss of JMML cells induced by forced differentiation of more immature cells into mature myelomonocytic cells with reduced proliferation potential. Bisulfite pyrosequencing of the human BMP4 promoter CpG island, a locus frequently hypermethylated in JMML, showed significantly reduced DNA methylation in JMML cells retrieved from 5AC-treated mice (31.32% +/-2.66% vs 52.46% +/-1.39%; p<0.001). In summary we created an ex vivo JMML xenograft model in immunodeficient mice that reflects many important aspects of this disorder and proved its usefulness for preclinical research of DNA methyltransferase inhibition because of extraordinary stability of leukemic DNA methylation patterns. 5AC showed clear preclinical efficacy in this model, supporting its further development in clinical treatment strategies for JMML. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 828-828
Author(s):  
Christiane Batz ◽  
Inga Sandrock ◽  
Peter Nöllke ◽  
Brigitte Strahm ◽  
Henrik Hasle ◽  
...  

Abstract Abstract 828 Promoter DNA hypermethylation contributes to the malignant phenotype in cancer including myeloproliferative neoplasms and myeloid leukemia. We hypothesized that aberrant DNA methylation also occurs in juvenile myelomonocytic leukemia (JMML) and asked whether it is associated with clinical, hematologic or prognostic features of the disease. Denaturing liquid chromatography was used to analyze peripheral blood or bone marrow samples from 87 children with JMML and 17 healthy control subjects for changes in DNA methylation at 14 candidate gene loci (BMP4, CALCA, CDKN1C, CDKN2B, DAPK1, MGMT, MLH1, PAWR, RARB, RASA1, RASSF1, RECK, SOCS1, TP73). We identified 4 genes with promoter DNA hypermethylation in JMML: BMP4 (34% of cases), CALCA (30%), CDKN2B (28%) and RARB (23%); the other 10 loci were unmethylated. The pattern of hypermethylation of the 4 genes allowed the categorization of JMML cases into three groups: no methylation (40/87 patients), intermediate methylation (1 or 2 genes; 29/87 patients) or high methylation (3 or 4 genes; 18/87 patients). Lineage-specific cell sorting demonstrated that aberrant methylation was restricted to clonal cell populations and could be traced back to the CD34+ JMML progenitor cell compartment. This observation supports the concept that DNA methylation is associated with early pathogenetic events in JMML. A correlative analysis of methylation groups with clinical or hematologic features showed that high methylation was strongly associated with higher age and increased hemoglobin F level at diagnosis (both p<0.01). By contrast, there was no significant association with leukocyte count, absolute monocytes, platelet count, blast percentage in blood or bone marrow, spleen size, karyotype or mutational category (PTPN11, RAS, CBL, NF1). Importantly, the presence of hypermethylation at diagnosis predicted cases with poor outcome; the 5-year overall survival (OS) in the no / intermediate / high methylation groups was 0.63 [0.47–0.79] / 0.52 [0.30–0.74] / 0.24 [0.04–0.44] (p<0.01). Among patients receiving hematopoietic stem cell transplantation, hypermethylation characterized cases with significantly increased probability of relapse: the 5-year relapse incidence in the no / intermediate / high methylation groups was 0.22 [0.11–0.45] / 0.21 [0.09–0.50] / 0.69 [0.49–0.96] (p<0.01). The predictive power of high methylation was also reflected in a multivariate Cox model for OS that included age at diagnosis, sex, platelet count and mutational category as other variables; here methylation was the only significant prognostic factor. Furthermore, longitudinal analyses indicated that some cases had acquired a higher methylation phenotype at relapse. In summary, we report aberrant DNA methylation as the most important molecular predictor of outcome in JMML. We suggest that a high-methylation phenotype characterizes an aggressive biologic variant of this leukemia. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Vol 159 (42) ◽  
pp. 1710-1719
Author(s):  
Krisztián Kállay ◽  
Judit Csomor ◽  
Emma Ádám ◽  
Csaba Bödör ◽  
Csaba Kassa ◽  
...  

Abstract: Introduction: Acquired bone marrow failures are rare but fatal diseases in childhood. Since 2013, Hungary has been participating as a full member in the work of the European Working Group on uniform diagnostics and therapy in patients with acquired bone marrow failure syndromes. Hypocellular refractory cytopenia of childhood has been emphasized as a frequent entity, transplanted by reduced intensity conditioning with excellent outcomes. Aim: To analyse and compare the results of treatment before and after our joining. Method: A total of 55 patients have been treated in the 8 centres of the Hungarian Pediatric Oncology Network during 5 years between 2013 and 2017 (severe aplastic anemia: 9, myelodysplastic syndrome: 41, juvenile myelomonocytic leukemia: 5 patients). Allogeneic hematopoietic stem cell transplantation was performed in severe aplastic anemia in 7 cases, while antithymocyte globulin was administered in one case and one patient died before diagnosis. In patients with myelodysplastic syndromes, watch and wait strategy was applied in 4, while transplantation in 37 cases. Reduced intensity conditioning was used in 54 percent of these cases. Transplantation was the treatment of choice in all 5 patients with juvenile myelomonocytic leukemia. Results: In the whole patient cohort, the time from diagnosis to treatment was median 92 (3–393) days, while in severe aplastic anemia median 28 (3–327) days only. Grade II–IV acute graft versus host disease occurred in 22.6%, grade III–IV in 6.8% and chronic in 11.2%. All the patients treated with severe aplastic anemia are alive and in complete remission (100%). The overall estimated survival rate is 85.1% in myelodysplastic syndrome, while 75% in juvenile myelomonocytic leukemia. The median follow-up was 30.4 (1.1–62.5) months. There was a remarkable increase in overall survival comparing the data before (1992–2012) and after (2013) joining the international group, 70% vs. 100% (p = 0.133) in severe aplastic anemia and 31.3% vs. 85.1% (p = 0.000026) in myelodysplastic syndrome. Conclusion: Due to a change in the paradigm of the conditioning regimen in hypocellular refractory cytopenia of childhood, the overall survival rate has significantly increased. Orv Hetil. 2018; 159(42): 1710–1719.


Blood ◽  
2000 ◽  
Vol 95 (2) ◽  
pp. 510-518 ◽  
Author(s):  
June Helen Myklebust ◽  
Erlend B. Smeland ◽  
Dag Josefsen ◽  
Mouldy Sioud

Protein kinase C (PKC) is a family of serine/threonine protein kinases involved in many cellular responses. Although the analysis of PKC activity in many systems has provided crucial insights to its biologic function, the precise role of different isoforms on the differentiation of normal hematopoietic progenitor cells into the various lineages remains to be investigated. The authors have assessed the state of activation and protein expression of PKC isoforms after cytokine stimulation of CD34+ progenitor cells from human bone marrow. Freshly isolated CD34+ cells were found to express PKC-, PKC-β2, and PKC-ɛ, whereas PKC-δ, PKC-γ, and PKC-ζ were not detected. Treatment with erythropoietin (EPO) or with EPO and stem cell factor (SCF) induced a predominantly erythroid differentiation of CD34+ cells that was accompanied by the up-regulation of PKC- and PKC-β2 protein levels (11.8- and 2.5-fold, respectively) compared with cells cultured in medium. Stimulation with EPO also resulted in the nuclear translocation of PKC- and PKC-β2 isoforms. Notably, none of the PKC isoforms tested were detectable in CD34+ cells induced to myeloid differentiation by G-CSF and SCF stimulation. The PKC inhibitors staurosporine and calphostin C prevented EPO-induced erythroid differentiation. Down-regulation of the PKC-, PKC-β2, and PKC-ɛ expression by TPA pretreatment, or the down-regulation of PKC- with a specific ribozyme, also inhibited the EPO-induced erythroid differentiation of CD34+ cells. No effect was seen with PKC-β2–specific ribozymes. Taken together, these findings point to a novel role for the PKC- isoform in mediating EPO-induced erythroid differentiation of the CD34+ progenitor cells from human bone marrow.


Blood ◽  
1997 ◽  
Vol 90 (2) ◽  
pp. 658-668 ◽  
Author(s):  
Amy C. Ladd ◽  
Robert Pyatt ◽  
Andre Gothot ◽  
Susan Rice ◽  
Jon McMahel ◽  
...  

Abstract Bone marrow (BM) CD34+ cells residing in the G0 phase of cell cycle may be the most suited candidates for the examination of cell cycle activation and proliferation of primitive hematopoietic progenitor cells (HPCs). We designed a double simultaneous labeling technique using both DNA and RNA staining with Hoechst 33342 and Pyronin Y, respectively, to isolate CD34+ cells residing in G0(G0CD34+ ). Using long-term BM cultures and limiting dilution analysis, G0CD34+ cells were found to be enriched for primitive HPCs. In vitro proliferation of G0CD34+ cells in response to sequential cytokine stimulation was examined in a two-step assay. In the first step, cells received a primary stimulation consisting of either stem cell factor (SCF), Flt3-ligand (FL), interleukin-3 (IL-3), or IL-6 for 7 days. In the second step, cells from each group were washed and split into four or more groups, each of which was cultured again for another week with one of the four primary cytokines individually, or in combination. Tracking of progeny cells was accomplished by staining cells with PKH2 on day 0 and with PKH26 on day 7. Overall examination of proliferation patterns over 2 weeks showed that cells could progress into four phases of proliferation. Phase I contained cytokine nonresponsive cells that failed to proliferate. Phase II contained cells dividing up to three times within the first 7 days. Phases III and IV consisted of cells dividing up to five divisions and greater than six divisions, respectively, by the end of the 14-day period. Regardless of the cytokine used for primary stimulation, G0CD34+ cells moved only to phase II by day 7, whereas a substantial percentage of cells incubated with SCF or FL remained in phase I. Cells cultured in SCF or FL for the entire 14-day period did not progress beyond phase III but proliferated into phase IV (with &lt;20% of cells remaining in phases I and II) if IL-3, but not IL-6, was substituted for either cytokine on day 7. G0CD34+ cells incubated with IL-3 for 14 days proliferated the most and progressed into phase IV; however, when SCF was substituted on day 7, cells failed to proliferate into phase IV. Most intriguing was a group of cells, many of which were CD34+, detected in cultures initially stimulated with IL-3, which remained as a distinct population, mostly in G0 /G1 , unable to progress out of phase II regardless of the nature of the second stimulus received on day 7. A small percentage of these cells expressed cyclin E, suggesting that their proliferation arrest may have been mediated by a cyclin-related disruption in cell cycle. These results suggest that a programmed response to sequential cytokine stimulation may be part of a control mechanism required for maintenance of proliferation of primitive HPCs and that unscheduled stimulation of CD34+ cells residing in G0 may result in disruption of cell-cycle regulation.


Sign in / Sign up

Export Citation Format

Share Document