A Highly Selective SF3B1-Targeted Splicing Inhibitor Reduces Human CD34+ Cell Survival and Self-Renewal In Acute Myeloid Leukemia

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1653-1653
Author(s):  
Larissa Balaian ◽  
Leslie A Crews ◽  
Marianna Zipeto ◽  
Kulidjian Anna ◽  
Edward D. Ball ◽  
...  

Abstract Introduction Myelodysplastic syndromes (MDS) result from ineffective hematopoietic stem cell (HSC) maintenance in an aged bone marrow microenvironment and have a proclivity for evolution to acute myeloid leukemia (AML). Progression to therapy resistant AML is driven by leukemia stem cells (LSC) harboring enhanced survival, dormancy and self-renewal capacity in supportive niches. Seminal next-generation DNA sequencing Results suggest that MDS evolution is controlled by mutations in splicing related genes and epigenetic modifiers of gene expression. However little is known about the cell type and context specific functional effects of these mutations on LSC transcriptional alterations that have been shown to promote MDS/AML progression and resistance to therapies such as 5-azacytidine (Vidaza). Therefore, we investigated the effect of splicing inhibitors on LSC survival and self-renewal 1) during progression of MSD to AML and 2) before and after clinical Vidaza treatment in a bone-marrow stromal co-cultures that recapitulates key aspects of the human LSC niche. Methods Mouse bone marrow cell lines, transfected to producehuman SCF,IL3 and G-CSF, were used as a stromal monolayers. Then human CD34+ cellswere selected from MDS (n=1) and AML primary samples (n=6). As normal controls, CD34+cells from cord blood (CB, n=3) or aged bone marrow (n=3) were utilized for the co-culture experiments. Survival and self-renewal of the CD34+ cells were investigated by colony forming and replating assays. Two SF3B1-targeted splicing inhibitors: FD 895 and a FD-analog were added at the initiation of co-culture at concentrations ranging from 0.1 to10 uM. Results After 2 weeksof stromal co-culture, none of the compounds demonstrated inhibition of the cell viability. Meanwhile, the splicing inhibitors demonstratedno reduction in survival in cord blood, and minor cytotoxicity toward aged bone marrow, MDS and AML samples showed a dose- and time-dependent significant (up to 80%) inhibition of colony formation. To analyzethe effect of splicing inhibitors on LSC self-renewal, replatingassayswere performed. While compounds at high doses mediated only a slight decrease in colony formation in normal CB and a-BM samples, MDS and AML samples exhibited a dose dependent inhibition of 38.2+/-8.1% of LSC survival (p<0.001) for FD895 and considerably lower 13.8+/-3.6 % of LSC survival for FD analog (p<0.001). Analysis of pre- and post progression samples from the same patient revealed the capacity of splicing inhibitors to diminish LSC survival. In CMML, FD895 induced significantly less cytotoxicity (35% compared with 75%) after progression to AML. Notably, in aged-BM both compounds reduced only CFU-GM survival, but not HSC self-renewal. In sequential primary samples from AML patients collected before and after clinical treatment with Vidaza, naïve samples exhibited similar sensitivity to FD-895 treatment in stromal co-culture models, and LSC survival and self-renewal capacity was reduced following incubation with FD-895. In contrast, following clinical treatment in patients that were responsive to Vidaza, these samples acquired resistance to splicing inhibition. However, patients that were non-responders to Vidaza treatment retained sensitivity to FD-895 treatment. Conclusions These data indicate that RNA editing and splicing activities represent novel regulators of LSC self-renewal and survival in LSC supportive niches. These properties can be inhibited using novel splicing inhibitors with minimal toxicity toward normal progenitors. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2820-2820
Author(s):  
Hironori Harada ◽  
Daichi Inoue ◽  
Noriko Doki ◽  
Ye Ding ◽  
Yuka Harada ◽  
...  

Abstract Abstract 2820 RUNX1/AML1 mutations have been frequently detected in patients with myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML). RUNX1 mutations are rarely detected in lower-risk MDS, whereas approximately 20% patients with higher-risk MDS (H-MDS) have the mutations. The mutations were distributed throughout the RUNX1 protein, and replacement of D171 amino acid in runt homology domain was the most frequent target of mutations in the RUNX1 gene. The D171N mutant showed a loss of normal RUNX1 trans-activation potential and dominant-negative suppression. In mouse transplantation systems D171N-transduced mice exhibited AML with multilineage dysplasia in collaboration with Evi1 overexpression. However, EVI1 overexpression was very rare in patients. Instead, most of H-MDS patients with RUNX1 mutations displayed a high expression of BMI1. RUNX1 D171N mutant showed an increased self-renewal capacity, differentiation block, dysplasia in all 3 lineages, slightly increased immature cells and no proliferation ability using enforced expression in human CD34+ cells, and the D171N-transduced cells showed low expression level of BMI1. Both D171N and BMI1 transduced cells displayed long-term proliferation ability. When BMI1 transduced later into D171N cells, the cells expanded with a retained CD34+ cell fraction, suggesting that BMI1 have a potential to boost the D171N cells to H-MDS. To confirm the collaboration of BMI1 overexpression with D171N mutant in vivo, we performed mouse BMT using BM cells transduced with both D171N and BMI1. Ly-5.1 murine BM mononuclear cells infected with retrovirus harboring D171N/BMI1 or control vectors were transplanted into sublethally irradiated syngeneic Ly-5.2 mice. Mice that received transplants of BMI1-transduced cells remained healthy over the observation period (n=12/12), as well as those that were transplanted with empty vectors-transduced cells (n=4/4). Most of the mice that received transplants of D171N -transduced cells developed MDS/AML mainly 6–8 months after transplantation (n=6/11, P<0.0001), as observed in the previous report. Of note, mice that received transplants of BM cells expressing D171N/BMI1 developed MDS/AML with significantly shorter latencies (mainly 3–5 months) compared with the D171N group (n=12/12, P=0.001). Morbid mice with D171N or D171N/BMI1 exhibited similar phenotypes, characterized by leukocytosis, anemia, and marked splenomegaly, while the mice with BMI1 or empty vectors, sacrificed 8 months after BMT, showed none of these phenotypes. In the leukemic mice with D171N or D171N/BMI1, BM and spleen were occupied by immature myeloid cells including myeloid blasts. More myeloblasts in BM were observed in D171N/BMI1 mice than in D171N ones. The leukemic cells displayed similar morphological abnormalities and surface markers: leukemic cells were CD11blow to high, Gr-1low, B220low and c-kitlow to high. The normal structure of the spleen was completely destroyed with massive blast and immature myeloid cell infiltration, and these cells also invaded into the hepatic portal areas in the liver. Meanwhile, BMI1-transduced BM cells did not become dominant in vivo and myeloid cells showed normal differentiation. Collectively, BMI1 overexpression has a strong potential to induce MDS/AML in concert with D171N in a mouse BMT model, although BMI1 overexpression by itself does not result in maturation block or leukemogenesis. BMI1 is well-known to be essential for self-renewal of HSCs, in part via repression of genes involved in senescence, and self-renewal of HSCs is enhanced by BMI1 expression. To address the mechanisms by which BMI1 would contribute to MDS/AML development, we analyzed gene expressions involved in BMI1 downstream signaling pathways. It is suggested that BMI1 overexpression may act as one of the partner abnormalities collaborating with master gene mutations for MDS-genesis. RUNX1 D171N mutant showed no proliferation ability using enforced expression in human CD34+ cells, and D171N-transduced mice exhibited MDS/AML in collaboration with Evi1 overexpression. In addition, co-transduction of D171N and BMI1 into BM cells resulted in faster induction of MDS/AML in BMT mice. Taken together, the RUNX1 mutant may require collaborating genes such as EVI1 and BMI1 to develop MDS/AML. We confirmed that BMI1 have a potential to boost the D171N cells to MDS/AML in vivo. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3931-3931
Author(s):  
Shaneice Renee Mitchell ◽  
Nicole Grieselhuber ◽  
Yerdanos Asemelash ◽  
Matthew Cannon ◽  
Shelley Orwick ◽  
...  

Abstract Acute Myeloid Leukemia (AML) is a heterogeneous disease that is characterized by an accumulation of neoplastic myeloid precursor cells in the bone marrow. Recently, multiple agents targeting AML associated mutations in FLT3, IDH1 and IDH2 have been developed. However, a majority of AML patients lack these mutations. Therefore, development of a novel therapeutic approach broadly relevant to AML would be attractive. The clonal capacity of AML cells is maintained by leukemic initiating cells (LICs), which possess self-renewal capabilities and are resistant against cytotoxic combination chemotherapy. Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step in the generation of NAD+, which is an important enzymatic cofactor and serves as a metabolite required for cellular respiration. LICs show a higher energy turnover rate than normal cells and are heavily reliant on oxidative phosphorylation. This suggests that energy generation processes, such as NAD+ biosynthesis, are critically required in myeloid malignancies. Thus, targeting the regeneration of NAD+ offers an attractive alternative therapeutic strategy in AML. Unlike many targeted therapies that are limited to one genetic subtype of AML, targeting regeneration of NAD+ via NAMPT inhibition could be relevant to a much broader patient population based upon metabolic differences between tumor and normal cells. We show that inhibition of NAMPT using the agent KPT-9274 induces loss of glycolytic and mitochondrial activity, more specifically a depleted total spare reserve capacity (p<0.001), in MV4-11 and THP-1 cell lines. KPT-9274 treatment decreased colony formation in AML patient samples across multiple genotypes (p<0.001) with a minimal decrease in colony formation by normal CD34+ hematopoietic cells. In addition, KPT-9274 treatment of AML cells decreased re-plating capacity, suggesting decreased self-renewal capacity (p=0.031). In contrast to pre-clinical activity observed in AML, there was minimal depletion of absolute counts observed in cell numbers of B, T and NK-lymphocyte treated ex-vivo as measured by flow cytometry, suggesting potent selectivity of KPT-9274. Furthermore, in a PDX mouse model of AML, KPT-9274 treatment significantly reduced disease burden (p<0.001) and prevented leukemic infiltration in spleen, bone marrow, and liver. In addition, KPT-9274 treatment induced differentiation, which is evident by a decrease in blast percentage and an introduction of segmented and band neutrophils in bone marrow differentials. Mice treated with KPT-9274 showed a reduction of bone marrow CD34+/CD38- cells (p<0.001), which are considered the stem cell population within the bulk leukemic cells. Extending our findings, consecutive re-plating of KPT-9274-treated PDX leukemic cells showed a marked decrease in colony formation, indicative of a loss of self-renewal capacity. KPT-9274 in combination with the anthracycline doxorubicin further decreased the infiltration of AML cells in comparison to either treatment alone. Overall, our data show that the NAMPT inhibitor KPT-9274 targets the leukemia initiating cell population, a potential source of resistance to cytotoxic chemotherapy, which is not adequately eradicated by current therapies in most patients. Disclosures Mims: Abbvie Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Agios Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy. Baloglu:Karyopharm Therpeutics: Employment. Senapedis:Karyopharm Therapeutics: Employment.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 5162-5162
Author(s):  
Ying Pang ◽  
Ying Feng ◽  
Xue Ye ◽  
Hanyun Ren

Abstract Objective To explore the feasibility, long-term hematopoiesis and complication of transplantation with two units of HLA-mismatched unrelated umbilical cord blood in treatment of adult acute myeloid leukemia. Methods A 32 years old, 50kg male with acute myeloid leukemia in complete remission received transplantation with two units of HLA-mismatched unrelated umbilical cord blood.The conditioning regimen were modified(BU/CY)+ATG. The prophylaxis regimen for graft versus-host disease(GVHD) consisted of cyclosporine(CSA) and mycophenolate mofetil(MMF). The umbilical cord blood obtained from two different donors, both with mismatched HLA B/DRB locus from the recipient and mismatched HLA- B locus between the two donors. The umbilical cord blood was preserved in liquid nitrogen, recovered in a 40o C water bath immediately, infused via a catheter from the femoral artery to the arc of aorta, The doses of nucleated cells infusion were 4.4×107/kg (from donor 1) and 2.8×107/kg (from donor 2). Results An absolute neutrophil count of more than 0.5×109/L at day 26,and a platelet count of more than 20×109/L at day 42. Septicemia with MRSA and pseudomomanas occurred at day 9 and day 14 because of agranulocytosis. The infection was controlled by Vancomycin, Tienam and HD-dose Gama immunoglobulin. Neither acute nor chronic GVHD developed in a follow-up period of 90 days. DNA-STR and HLA distinct analysis assay revealed a complete implant of cells from only one donor(donor2). Conclusions 1.No donor with matched HLA bone marrow stem cell was available for the adult patient at the time of his relapse. HLA mismatched umbilical cord blood was obtained from two donors. Although cell counts for transplantation are much lower than the requirement of routine bone marrow transplantation, the speed of blood cell regeneration in the recipient is compatible with routine bone marrow transplantation. 2.Furthermore, Although DNA-STR and HLA analysis indicate complete implant of cells from only one donor, the result indicates transplantation with umbilical cord blood cells obtained from two different donors is promising in the situation where the cell number obtained from one donor is not enough.3.HLA- B/DRB locus was mismatched in the two donors. GVHD did not develop even after tapered off immunosuppresents 3 months post transplantation. No cross rejection observed by clinical presentation and blood cell analysis. The results indicate the incidence of GVHD is low after umbilical cord blood transplantation.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2010-2010
Author(s):  
Kazuhiro Masuoka ◽  
Shigesaburo Miyakoshi ◽  
Kazuya Ishiwata ◽  
Masanori Tsuji ◽  
Shinsuke Takagi ◽  
...  

Abstract &lt;Objectives&gt; Promising results of cord blood transplants from unrelated donors have been reported in adults. To compare of outcomes of bone marrow transplants (BMT, n = 51), and umbilical cord blood transplants (UCBT, n = 110) from unrelated donors in adult patients with acute myeloid leukemia (AML) / myelodysplastic syndrome (MDS), we analyzed retrospectively the results of 161 adult patients with AML and MDS in our hospital. &lt;Patients and Methods&gt; We reviewed medical records of 161 patients with AML/MDS who had received a hematopoietic stem cell transplant from an unrelated donor between August 2000 and April 2007 at Toranomon Hospital, Tokyo, Japan. &lt;Results&gt; Patient’s median age was 55 years (17–71). Diagnoses include de novo AML (n =85), MDS overt AML (n=48), refractory anemia (RA) (n=13), and refractory anemia with excess of blasts (RAEB) (n=15). Disease status consisted of standard (CR1 of AML and RA, n=30) and advanced (other status, n=131). Recipients of UCBT had more advanced disease than recipients of BMT at the time of transplantation (89 percent vs. 65 percent, P&lt;0.001). The median number of nucleated cells that were infused was 0.26×108 per kilogram of the recipient’s body weight for cord blood and 2.5×108 per kilogram for bone marrow (P&lt;0.001). The major difference were higher number in the UCBT group of HLA mismatches (defined by serology for class 1 and molecular typing for DRB1).The donor was HLA mismatched in 96% of UCBT recipients, and in 41% of BMT recipients (P&lt;0.001). Other significant differences were observed in preparative regimens, and graft-versus-host disease (GVHD) prophylaxis. Nonadjusted estimates of 2-year OS and DFS rates were 53% and 48% in the BMT group, and 33% and 25% in the UCBT group (P&lt;0.001). However, 2-year OS and DFS rates in the standard group were not significantly different in the two groups (63% and 63% in the BMT group, and 75% and 58% in the UCBT group; p=0.98 and 0.32). Compared with BMT recipients, UCBT recipients had delayed hematopoietic recovery (Hazard ratio [HR]= 0.52; 95% confidence interval [95CI]: 0.36–0.75; p&lt;0.001), increased 100 day TRM (HR=3.07; 95CI 1.45–6.51; p&lt;0.01) and decreased grade II–IV acute graft-versus-host disease (aGVHD) (HR=0.58; 95CI 0.35–0.96; p=0.03). Two-year relapse rate was not significantly different in the two groups. &lt;Conclusion&gt; We conclude that UCBT from an unrelated donor is a therapeutic option for adult AML/MDS patients who lack an HLA-identical donors. Higher mortality, especially from non-relapse causes, is the biggest problem to be solved to increase the feasibility of this approach.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1349-1349
Author(s):  
Carolien Woolthuis ◽  
Lina Han ◽  
Djoke van Gosliga ◽  
Philip Kluin ◽  
Edo Vellenga ◽  
...  

Abstract Mutations in the nucleophosmin (NPM) gene are found in about 30% of cases of acute myeloid leukemia (AML) and lead to a dislocation of the nucleophosmin protein from the nucleus to the cytoplasm (NPMc+ AML). NPMc+ AML shows distinctive biological and clinical features, including a unique gene expression profile, a distinct microRNA signature, low percentage of CD34+ cells, increased incidence of Flt3-ITD (about 40% of cases), good response to induction chemotherapy and (in the absence of Flt3-ITD) a favourable prognosis. Despite significant progress in the characterization of the NPMc+ AML subgroup, questions remain about the leukemia-initiating cell. Distinct features of NPMc+ AML, including multilineage involvement and overexpression of HOX-genes, may point to an early progenitor as the leukemia-initiating cell, but the characteristic low percentage of CD34+ cells may point to a more differentiated leukemic stem cell in NPMc+ AML. To gain more insight in the leukemia-initiating cell in AML with mutated NPM, NPMc+ AML cells were sorted based on the expression of CD34 (n=8, the percentage of CD34+ in the total AML fraction varied between 0.06 and 37%). Western blotting, using an antibody that specifically recognizes the nucleophosmin mutant protein revealed that the NPM mutant protein is expressed in both CD34+ and CD34− cells, proving that the CD34+ NPMc+ AML cells belong to the leukemic clone. This was verified by sequencing the NPM gene in CD34+ and CD34− AML cells. Importantly, culture of sorted CD34+ and CD34− NPMc+ AML cells on a stromal layer revealed that the CD34+ but not the CD34− cells of NPMc+ AML were capable of expanding and initiating long-term growth. In the first 5 weeks of culture an at least 16 fold (range 16–208) expansion of CD34+ AML cells was seen in 5 out of 6 NPMc+ AML cases. This expansion was associated with the formation of cobblestone areas (CAs) under the stromal layer within 3 weeks after plating. The NPMc+ AML cells which expanded in culture were able to expand further after replating in 4 out of 5 investigated cases (fold expansion range 1.6–2.5), indicative of the self renewal capacity of these CD34+ NPMc+ AML cells. Gene expression analysis of CD34+ and CD34− NPMc+ AML cells of 4 cases analyzed thus far revealed the presence of the characteristic HOX-overexpression profile in both CD34+ and CD34− NPMc+ AML cells. In summary, this study shows that the NPM mutation is not only present in CD34−, but also in CD34+ cells of NPMc+ AML and that the properties of long-term expansion and self renewal belong exclusively to the CD34+ subfraction of NPMc+ AML.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1320-1320
Author(s):  
James Ropa ◽  
Nirmalya SAHA ◽  
Andrew G. Muntean

Abstract Epigenetic regulators play an important role in normal and malignant hematopoiesis. Epigenetic deregulation of the HOXA gene cluster drives transformation of about 50% of acute myeloid leukemia (AML), including those harboring MLL rearrangements and NPM mutations, as well as others. Expression of Hoxa9 and its co-factor Meis1 is sufficient to transform bone marrow into a lethal AML in mouse models. We previously demonstrated that the pro-leukemic genes Hoxa9 and Meis1 are critically regulated by the histone H3 Lysine 9 (H3K9) methyltransferase SETDB1. Recent studies show that SETDB1 is required for normal hematopoiesis and MLL-AF9 mediated leukemia (Koide, et al. Blood 2016). Our lab recently demonstrated that SETDB1 negatively regulates the expression of HoxA9 and Meis1 through deposition of promoter H3K9 methylation in MLL-AF9 AML cells (Ropa et al. Oncotarget 2018). Consistent with these data, HOXA9 and MEIS1 expression negatively correlates with SETDB1 expression in AML patient samples. Therefore, we investigated the biological impact of SETDB1 on AML. We first noted that expression of SETDB1 in AML patient samples is significantly lower compared to normal hematopoietic cells. Further, higher SETDB1 expression correlated with a significantly better overall survival (p=0.003) and lower expected hazard (HR=0.9/100RSEM; p=0.009) in AML patients compared with lower SETDB1 expression. These data are consistent with SETDB1 negatively regulating pro-leukemic genes and suggests that SETDB1 expression may be correlated with AML patient prognosis. We tested this directly by expressing high levels of SETDB1 in AML cells. Ex vivo assays show that retroviral overexpression of SETDB1 in MLL-AF9 AML cells leads to cell differentiation, decreased leukemia colony formation, and decreased cell proliferation. Consistent with the AML patient data, overexpression of SETDB1 significantly delays MLL-AF9 mediated leukemogenesis in vivo (p=0.01). Further, we observed a strong selective pressure against exogenous SETDB1 expression in moribund mice. Transcriptome analyses demonstrate that SETDB1 globally represses Hox and pluripotency gene programs. Strikingly, we found that SETDB1 represses many of the same genes that exhibit reduced promoter H3K9me3 in AML patient samples relative to CD34+ cells. These data point to a role for SETDB1 in negatively regulating pro-leukemic target genes and suppressing AML. We also explored how chemical and genetic inhibition of H3K9 methylation and Setdb1 affects AML initiation and maintenance. We first confirmed the previously reported requirement for Setdb1 in AML cell lines by genetically deleting both alleles of Setdb1 in MLL-AF9 cells, which resulted in a complete arrest of proliferation (Koide, et al. Blood 2016). Combined with our data presented above, these results suggest a narrow window of SETDB1 expression is maintained in AML cells. To achieve reduced (but not complete loss of) activity, we investigated how small molecule inhibition of H3K9 methylation (UNC0638) or shRNA mediated knock down of Setdb1 affects AML initiation. We observed increased ex vivo colony formation of normal ckit+ bone marrow cells upon shRNA mediated knockdown of Setdb1 or upon UNC0638 treatment. We hypothesized that this expansion of colony forming unit potential of hematopoietic cells may translate to increased transformation potential by leukemic oncogenes. Indeed, cells pretreated with UNC0638 followed by retroviral transduction with MLL-AF9 exhibit significantly higher capacity for leukemic colony formation than vehicle treated cells. These data are consistent with H3K9 methylation repressing genes required for AML transformation. Our data identified a narrow window of expression of SETDB1 in AML patient samples. SETDB1 expression is reduced in AML patients relative to normal cells and chemical inhibition of H3K9 methylation expands the pool of cells amenable to MLL-AF9 mediated transformation ex vivo. While inhibition of SETDB1 and other H3K9 methyltransferases has been suggested as a possible therapeutic strategy, our data suggests this may also prime bone marrow cells for transformation by inhibiting epigenetic processes that repress pro-leukemic target genes. Further investigation of the roles of SETDB1 and H3K9 methylation levels is necessary to determine the value of these epigenetic modifiers as therapeutic targets in AML and is currently ongoing. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3426-3426
Author(s):  
M. Gorre ◽  
I. Jilani ◽  
R. Chang ◽  
H. Chan ◽  
R. Urcia ◽  
...  

Abstract Heat shock proteins (HSPs) are molecular chaperones involved in binding and regulating the levels of several client cellular proteins. HSPs are induced by stress and play a role in the modulation of apoptosis and proliferation. Expression of HSPs by acute myeloid leukemia (AML) cells has been reported to correlate with more aggressive disease. Here we used a flow cytometry approach to quantify the expression of HSP90 and HSP70 in specific cell populations in bone marrow. Quantification using QuantiBRITE and PE (phycoerythrin)-labeled antibodies with a 1:1 ratio allowed us to specifically measure the antibody binding capacity in 100 CD34+ cells (molecules/100 CD34+ or CD3+ cells). Using this approach, we compared bone marrow samples from patients with myelodysplastic syndrome (MDS) and patients with AML. This approach can also be used to monitor patients treated with therapeutic agents that target HSPs, such as 17-allylamino-17-demethoxygeldamycin (17AAG). The percentage of CD34+ cells expressing HSP90 (P=0.008) and HSP70 (P&lt;0.001) was significantly lower in MDS (n=20) than in AML (n=33) patients. Similarly, the percentage of CD3+ cells expressing HSP90 and HSP70 was significantly higher in MDS than in AML patients (P&lt;0.01). This suggests that the environment in the bone marrow (cytokines, chemokines, other factors) may affect the levels of HSPs in neoplastic and normal cells in a similar fashion. In patients with MDS, higher levels of HSP90 were associated with shorter survival (P=0.03). However, after achieving remission (CR), MDS patients who expressed high levels of HSP90 had significantly longer remission duration (CRD) (P=0.03). These findings not only confirm that the environment and blasts in patients with MDS are different from those in patients with AML, but also suggest that therapy targeting HSPs may be a rational approach in patients with MDS. Figure Figure


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 151-151
Author(s):  
Ramiro Garzon ◽  
Stefano Volinia ◽  
Chang G. Liu ◽  
Flavia Pichiorri ◽  
Tiziana Palumbo ◽  
...  

Abstract MicroRNAs are small non-coding RNAs of 19–25 nucleotides in length that are negative regulators of gene expression. Findings over the last few years indicate that microRNAs are involved in fundamental cellular process, including development and hematopoietic differentiation. Acute myeloid leukemia (AML) is a heterogeneous disorder that is characterized by proliferation of immature cells. Although there are well defined molecular subtypes of AML, the pathogenesis in the majority of cases is largely unknown. Focusing on known genes will not likely suffice to uncover the nature of the AML. The integration of a whole genome approach including non-coding RNA gene products may lead to an improve understanding of the biology of AML. Methods: To determine whether microRNAs are associated with known cytogenetic abnormalities and biological features in AML, we evaluated the microRNA expression profiles of 176 samples of adult AML with intermediate and poor risk cytogenetics and 10 CD34+ cells from healthy donors using a microarrays platform. After normalization, data were analyzed using significance analysis of microarrays and prediction analysis of microarrays software. An independent set of 28 patients with AML was used to validate the signatures using quantitative real time PCR. Treatment response was evaluated in 29 newly AML diagnosed patients 4 to 6 weeks after induction chemotherapy with idarubicin and cytarabine by bone marrow examination. Complete remission was defined as less than 5% blasts in the bone marrow. Otherwise it was categorized as resistant disease. Results: We found several microRNAs differentially expressed between CD34+ cells and all the AML samples. A subset of these microRNAs reflects the differentiation stage of the leukemias and correlate with the French-American-British classification of AML. Likewise, microRNAs are closely associated with the prevalent cytogenetic abnormalities. A common signature including the over expressed miR-20; miR-17, miR-25 and miR-191 are associated with short overall survival, while miR-29b is found down-regulated in patients with resistant disease. Furthermore, we proved experimentally that miR-29b regulates negatively MCL-1, a critical apoptosis regulator, which has been found up-regulated and associated with relapse and chemotherapy resistance in leukemia. Conclusions: MicroRNAs expression in AML is closely associated with differentiation stage, morphology and cytogenetics. A subset of MicroRNAs is correlated with survival and treatment response.


Blood ◽  
2009 ◽  
Vol 114 (6) ◽  
pp. 1150-1157 ◽  
Author(s):  
Steven W. Lane ◽  
David T. Scadden ◽  
D. Gary Gilliland

Abstract The genetic events that contribute to the pathogenesis of acute myeloid leukemia are among the best characterized of all human malignancies. However, with notable exceptions such as acute promyelocytic leukemia, significant improvements in outcome based on these insights have not been forthcoming. Acute myeloid leukemia is a paradigm of cancer stem (or leukemia initiating) cells with hierarchy analogous to that seen in hematopoiesis. Normal hematopoiesis requires complex bidirectional interactions between the bone marrow microenvironment (or niche) and hematopoietic stem cells (HSCs). These interactions are critical for the maintenance of normal HSC quiescence and perturbations can influence HSC self-renewal. Leukemia stem cells (LSCs), which also possess limitless self-renewal, may hijack these homeostatic mechanisms, take refuge within the sanctuary of the niche during chemotherapy, and consequently contribute to eventual disease relapse. We will discuss the emerging evidence supporting the importance of the bone marrow microenvironment in LSC survival and consider the physiologic interactions of HSCs and the niche that inform our understanding of microenvironment support of LSCs. Finally, we will discuss approaches for the rational development of therapies that target the microenvironment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2351-2351
Author(s):  
Michael Heuser ◽  
Gernot Beutel ◽  
Jürgen Krauter ◽  
Nils von Neuhoff ◽  
Brigitte Schlegelberger ◽  
...  

Abstract Cytogenetic aberrations are important prognostic factors in acute myeloid leukemia (AML). However, approximately half of adult AML patients lack cytogenetic abnormalities and identification of predictive molecular markers might improve therapy. Fusion of meningioma-1 (MN1) to TEL (ETV6) has been found in AML and MDS with t(12;22)(p13;q11). However, expression levels of MN1 have not been reported previously in AML. We evaluated MN1 expression as a prognostic marker in 142 AML patients aged 18–60 years with normal cytogenetics, who were uniformely treated according to the AML-SHG 1/99 trial. Patients received intensive, cytarabine-based induction and consolidation treatment including allogeneic progenitor cell transplantation if an HLA-compatible sibling was available, or in case of relapse. Specimens were obtained at diagnosis, and routine cytogenetic, FLT3-mutation, and MLL-PTD analyses were performed. MN1 expression was quantified by real-time RT-PCR on a LightCycler using QuantiTect SYBR Green. AML samples were dichotomized at the median value resulting in two groups: a low MN1 group and a high MN1 group. Baseline characteristics and outcome parameters were compared between these two groups. In addition, CD34+ cells were immunomagnetically enriched from mobilized blood of a healthy donor using MACS CD34 isolation kit. Cells were cultured in IMDM medium with various cytokines including either G-CSF, M-CSF or EPO. At various time points, cells were harvested and analyzed for MN1 expression. There were no significant differences between low MN1 and high MN1 expressing patients with respect to age, gender, ECOG performance status, diagnosis of de novo or secondary AML, FAB morphology, white blood cell count, percentage of blasts in blood or bone marrow, FLT3 mutations, or MLL-PTD. Low MN1 expressing patients significantly more often achieved a good response to the first course of induction treatment defined as blasts in bone marrow below 5%, no blasts in peripheral blood, and no extramedullary manifestation at day 15 compared to high MN1 expressing patients (87.3% vs. 71.8%, p=.02). There was no significant difference for remission status between the two groups. High MN1 expression predicted significantly shorter event-free survival (19% vs. 45.8% at 3 years, log-rank p=.0009), shorter relapse-free survival (23% vs. 52.8% at 3 years, log-rank p=.001), and shorter overall survival (38.2% vs. 58.8% at 3-years, log-rank p=.03). The high MN1 group relapsed significantly more often compared to the low MN1 group (56.7% vs. 35%, p=.02), and thus received an allogeneic transplant significantly more often (50.7% vs. 33.8%, p=.04). In multivariate analysis including known risk factors only MN1 expression, age (above the median compared to below the median age), and ECOG performance status (0 or 1 compared to 2) remained significant (hazard ratio: 2 (p=.01), 2.1 (p=.005) and 2.8 (p=.005), respectively). MN1 expression in CD34+ cells was 37-fold higher compared to the CD34− cell fraction. However, by in vitro differentiation of CD34+ cells using various cytokines including either G-CSF, M-CSF or EPO, MN1 expression dropped to levels found in the CD34− fraction within 7 days of culture. In conclusion, high MN1 expression predicts adverse prognosis and may define an important risk factor in AML with normal cytogenetics. Its upregulation in hematopoietic progenitor cells hints at a functional role of MN1 in blocking differentiation.


Sign in / Sign up

Export Citation Format

Share Document