ALBERT

All Library Books, journals and Electronic Records Telegrafenberg

feed icon rss

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • 2010-2014  (15)
  • 1
  • 2
    Publication Date: 2012-11-16
    Description: Abstract 2862 Chronic and juvenile myelomonocytic leukemias (CMML and JMML) are overlap myelodysplastic/myeloproliferative neoplasia (MDS/MPN) syndromes that respond poorly to conventional treatment regimens. Both diseases are characterized by aberrant N-Ras, K-Ras, Cbl, and SHP-2 proteins, which are not attractive drug target candidates. Focus has shifted to downstream effector pathways, which include Raf/MEK/ERK, phosphoinositide-3-OH kinase (PI3K)/Akt, and Ral-GDS/Ral-A cascades. However, it is unclear which pathways, if any, should be targeted. In part to address this question, we previously developed a mouse model of CMML and JMML by expressing a conditional “knock-in” KrasG12D oncogene in bone marrow. Our earlier studies showed that inhibition of MEK yields a significant reduction in disease burden in this model, including reduced leukocytosis, improved anemia and enhanced survival. Here, we interrogate the role of the PI3K/Akt pathway in KrasG12D driven MPN and further explore which specific pathways are responsible for leukemogenesis due to hyperactive Ras. We administered GDC-0941, a selective PI3K inhibitor, to Mx1-Cre, KrasG12D mutant mice Mice with well established MPN and wild-type (WT) littermates were randomly chosen to receive daily oral administration of GDC-0941 or a control vehicle. Treated mice exhibited dramatic corrections of leukocytosis and anemia as well as decrease in splenomegaly. Flow cytometry of bone marrow and peripheral populations also imply that GDC-0941 treatment corrects the aberrant proliferation, amends differentiation of bone marrow progenitors, and revives ineffective erythropoiesis found in KrasG12D mice. Treatment also resulted in markedly improved survival of KrasG12D mice; virtually all KrasG12Dmice in the treatment arm outlived their control counterparts. Our data suggest PI3K inhibition may play a role in suppressing hematologic dysfunction in JMML and CMML patients. Potential crosstalk between PI3K and MEK signaling further suggest that combinatorial activities of PI3K and MEK inhibition should be investigated. Disclosures: Friedman: Genentech, Inc.: Employment. Sampath:Genentech, Inc.: Employment.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 3
    Publication Date: 2011-04-21
    Description: Src homology 2 domain-containing phosphatase 2 (Shp2), encoded by Ptpn11, is a member of the nonreceptor protein-tyrosine phosphatase family, and functions in cell survival, proliferation, migration, and differentiation in many tissues. Here we report that loss of Ptpn11 in murine hematopoietic cells leads to bone marrow aplasia and lethality. Mutant mice show rapid loss of hematopoietic stem cells (HSCs) and immature progenitors of all hematopoietic lineages in a gene dosage-dependent and cell-autonomous manner. Ptpn11-deficient HSCs and progenitors undergo apoptosis concomitant with increased Noxa expression. Mutant HSCs/progenitors also show defective Erk and Akt activation in response to stem cell factor and diminished thrombopoietin-evoked Erk activation. Activated Kras alleviates the Ptpn11 requirement for colony formation by progenitors and cytokine/growth factor responsiveness of HSCs, indicating that Ras is functionally downstream of Shp2 in these cells. Thus, Shp2 plays a critical role in controlling the survival and maintenance of HSCs and immature progenitors in vivo.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 4
    Publication Date: 2012-10-25
    Description: Ras proteins are critical nodes in cellular signaling that integrate inputs from activated cell surface receptors and other stimuli to modulate cell fate through a complex network of effector pathways. Oncogenic RAS mutations are found in ∼ 25% of human cancers and are highly prevalent in hematopoietic malignancies. Because of their structural and biochemical properties, oncogenic Ras proteins are exceedingly difficult targets for rational drug discovery, and no mechanism-based therapies exist for cancers with RAS mutations. This article reviews the properties of normal and oncogenic Ras proteins, the prevalence and likely pathogenic role of NRAS, KRAS, and NF1 mutations in hematopoietic malignancies, relevant animal models of these cancers, and implications for drug discovery. Because hematologic malignancies are experimentally tractable, they are especially valuable platforms for addressing the fundamental question of how to reverse the adverse biochemical output of oncogenic Ras in cancer.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 5
    Publication Date: 2011-11-18
    Description: Abstract 797FN2 Neurofibromatosis type 1 (NF1) is a dominant genetic disorder that is caused by germ line mutations in the NF1 tumor suppressor gene. Children with NF1 have a 200- to 500-fold increase in the risk of developing malignant myeloid disorders, particularly JMML, an aggressive myeloproliferative neoplasm (MPN) characterized by over-production of myeloid lineage cells that show extensive tissue infiltration, anemia, and thrombocytopenia. The only curative therapy for JMML is hematopoietic stem cell transplantation, and 50% of children relapse after HSCT and die of recurrent JMML or of transformation to acute myeloid leukemia (AML). Somatic inactivation of Nf1 in hematopoietic cells causes a progressive MPN in Mx1-Cre, Nf1flox/flox mice that closely models JMML. We administered the MEK inhibitor PD0325901 (901) to Mx1-Cre, Nf1flox/flox mice with MPN to investigate the therapeutic effects of inhibiting Raf/MEK/ERK signaling in vivo and to interrogate the underlying mechanism of any observed response. Mx1-Cre, Nf1flox/flox mice (n = 35) and WT littermates (n = 38) were randomized to receive 901 (5 mg/kg/day) or control vehicle at 6 months of age. Treatment was continued for 10 weeks or until the mice became moribund. Mx1-Cre, Nf1flox/flox mice that received 901 demonstrated a rapid reduction in leukocyte counts and enhanced erythropoiesis. In addition, spleen size was markedly reduced with correction of aberrant proliferation and differentiation of progenitor cells. Flow cytometric analysis showed that 901 treatment restored a normal pattern of erythroid differentiation and greatly reduced splenic hematopoiesis. Remarkably, genetic studies revealed persistence of Nf1-deficient hematopoietic cells despite dramatic improvement in hematologic abnormalities. Based on these data, we conclude that treatment with 901 rebalances the proliferation and differentiation of Nf1 mutant cells in vivo rather than eliminating them. These preclinical data and a recent study in Kras mutant mice with MPN (Lyubynska N, et al. Science Trans Med 2011; 3: 76ra27) support clinical trials of MEK inhibitors in JMML and chronic myelomonocytic leukemia and suggest differentiation of malignant cell as a potential mechanism of response to this class of targeted agents. Disclosures: No relevant conflicts of interest to declare.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 6
    Publication Date: 2011-02-10
    Description: NRAS is frequently mutated in hematologic malignancies. We generated Mx1-Cre, Lox-STOP-Lox (LSL)-NrasG12D mice to comprehensively analyze the phenotypic, cellular, and biochemical consequences of endogenous oncogenic Nras expression in hematopoietic cells. Here we show that Mx1-Cre, LSL-NrasG12D mice develop an indolent myeloproliferative disorder but ultimately die of a diverse spectrum of hematologic cancers. Expressing mutant Nras in hematopoietic tissues alters the distribution of hematopoietic stem and progenitor cell populations, and Nras mutant progenitors show distinct responses to cytokine growth factors. Injecting Mx1-Cre, LSL-NrasG12D mice with the MOL4070LTR retrovirus causes acute myeloid leukemia that faithfully recapitulates many aspects of human NRAS-associated leukemias, including cooperation with deregulated Evi1 expression. The disease phenotype in Mx1-Cre, LSL-NrasG12D mice is attenuated compared with Mx1-Cre, LSL-KrasG12D mice, which die of aggressive myeloproliferative disorder by 4 months of age. We found that endogenous KrasG12D expression results in markedly elevated Ras protein expression and Ras-GTP levels in Mac1+ cells, whereas Mx1-Cre, LSL-NrasG12D mice show much lower Ras protein and Ras-GTP levels. Together, these studies establish a robust and tractable system for interrogating the differential properties of oncogenic Ras proteins in primary cells, for identifying candidate cooperating genes, and for testing novel therapeutic strategies.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 7
    Publication Date: 2012-11-16
    Description: Abstract 4916 Background: Aberrant signal transduction plays a central role in the pathogenesis of MDS/MPN, as indicated by the high prevalence of mutations that activate Ras signaling. Yet despite the central role of Ras signaling in the pathogenesis of JMML, at this time there are no signal transduction inhibitors with established efficacy in JMML. A screen of inhibitors has the potential to reveal potential therapeutic strategies and inform efforts to treat other neoplasms driven by hyperactive Ras signaling, both in the hematopoietic system and elsewhere. Aim: To investigate novel therapeutic options for JMML by utilizing a novel, reproducible system for rapid screening in primary cells. Innovations include using flow cytometry to isolate a highly clonogenic, disease-relevant “PreGM” population of primary bone marrow cells that recapitulate the abnormal growth pattern characteristic of JMML and unsorted bone marrow, the use of a genetically engineered mouse model, and the development of automated microscopy protocols. Method: Unfractionated bone marrow cells harvested from Mx1-Cre, KrasD12 and wildtype mice were utilized in the screens. PreGM cells, identified as Lineage lo/- Sca1- c-kit+ CD34+ CD16/32- CD105- CD150-, were purified from harvested bone marrow using flow cytometry. The purified PreGM cells were sorted into 96 well plates containing various inhibitors at set concentrations ranging from 1X (5 μg/ml, approx. 10 μM for most compounds) to 10−7X (5×10−7 μg/ml). The freshly sorted PreGM cells were exposed to inhibitors for 3 days under standard culture conditions (at 37°C, 98% humidity and 5% CO2) in 80% IMDM, 20% FBS and saturating dose of 10ng/ml of GM-CSF. At the end of that period, cell growth was quantified using the IN Cell Analyzer 2000 (GE). A total of 94 different inhibitors were screened using this method. The screen included a negative control (DMSO) and cytotoxic positive controls (Cytarabine, Adriamycin and Gemcitabine). Compound families included cyotoxic agents, tyrosine kinase inhibitors, PI3K family inhibitors, mitotic kinase inhibitors, epigenetic modifiers, hedgehog signaling inhibitors, and others. The majority of compounds were either FDA approved drugs or agents used in recent clinical trials. Candidates were screened for preferential activity against Mx1-Cre, KrasD12 cells. Results: Primary bone marrow cells were harvested from a total of 28 mice, 18 wild type (WT) and 10 Mx1-Cre, KrasD12. PreGM growth was quantified and dose response curves constructed for WT and mutant cells. WT and mutant IC50s for each compound were calculated using the ‘drc’ package from the R Project for Statistical Computing. Out of 94 candidates tested in this screen, none were found to demonstrate preferential inhibitory activity against Mx1-Cre, KrasD12 cells. Neither were any of the drugs found to be comparatively toxic to WT cells or to have significantly higher IC50s in mutant PreGM cells in comparison to WT cells. Some compounds of interest included Vorinostat, an epigenetic inhibitor, which was found to have robust inhibitory activity against both mutant and WT cells. It has comparable IC50s in mutant and WT cells with a calculated IC50 of 0. 0480X (std. error: 0. 135) in Mx1-Cre, KrasD12 cells and 0. 0244X (std. error: 0. 0293) in WT cells. Conclusion: None of the 94 compounds used in the screen were found to preferentially inhibit mutant or WT cell growth, indicating that Kras mutant cells have similar drug sensitivities to normal cells over a broad range of mechanistic approaches. These findings suggest that it may be difficult to find “synthetic lethal” opportunities for drugs that are selectively toxic to primary cells driven by hyperactive Ras signaling. Disclosures: No relevant conflicts of interest to declare.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 8
    Publication Date: 2012-09-27
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 9
    Publication Date: 2014-12-06
    Description: Juvenile Myelomonocytic Leukemia (JMML) is an aggressive myeloproliferative neoplasm of childhood with a 5-year event free survival of 52% after hematopoietic stem cell transplantation (HSCT). A hallmark of JMML is aberrant Ras pathway activation due to mutations in NF1, NRAS, KRAS, PTPN11 and CBL. However, robust predictors of response are lacking, as individual mutations are not reliably associated with outcome, and relapse remains the most common reason for treatment failure. Recently, massively parallel sequencing has identified recurrent mutations in the SKI domain of SETBP1 in a variety of myeloid disorders, including JMML (Piazza et al Nat Genet 2012, Makishima et al Nat Genet 2013, Sakaguchi et al Nat Genet, 2013). These mutations had a lower allelic frequency compared to Ras pathway mutations, but were associated with poor prognosis. These and other data suggested that SETBP1 mutations contribute to disease progression rather than initiation. We identified several patients with JMML who had clonal SETBP1 mutations detected at relapse. Analysis of mononuclear cell extracted DNA from serial samples of two patients who relapsed revealed an increase in the SETBP1 mutant allele frequency over time (Figure 1). Similarly, analysis of colonies plated in methylcellulose from serial time points indicated that the percentage of individual myeloid progenitor colonies that were heterozygous or homozygous for the SETBP1 mutation increased with each sequential sample despite intensive treatment. Based on these data, we tested the hypothesis that rare SETBP1 mutant clones exist at diagnosis in many patients who relapse, and that these rare cells undergo positive selection during treatment. Using a droplet digital PCR (ddPCR) technology with a detection threshold as low as 0.001% of mutant DNA, we identified SETBP1 mutations in 16/53 (30%) of diagnostic JMML specimens from children treated on Children's Oncology Group trial AAML0122. Of these mutations, 12 were subclonal and 4 were clonal. Event free survival (EFS) at 4 years in patients with SETBP1 mutations was 19% ± 10% compared to 51% ± 8% in those with wild type SETBP1 (p=0.006). While samples of patients who relapsed on the AAML0122 trial were not available for analysis, one patient recently undergoing treatment who had a subclonal SETBP1 mutation (0.45% allelic fraction) detected at diagnosis by ddPCR, demonstrated an overt SETBP1 mutation at relapse. Finally, we isolated and analyzed hematopoietic stem (HSC), multipotent progenitor (MPP), common myeloid progenitor (CMP), and granulocyte-monocyte progenitor (GMP) populations from a relapsed sample with a SETBP1 mutation. Sanger sequencing demonstrated that all four progenitor compartments were affected by the mutation. Analysis of additional samples is underway. We conclude that the presence of a subclonal mutation in SETBP1 is a novel biomarker of adverse outcome in JMML. Understanding the mechanisms underpinning SETBP1-mediated resistance and relapse, and further identifying therapeutic vulnerabilities of HSCs expressing these mutant proteins will be critical to improve outcomes for patients with JMML and other myeloid malignancies. Furthermore, the presence of a subclonal SETBP1 mutation at diagnosis might identify JMML patients who will benefit from more intensive conditioning prior to HSCT or from novel therapeutic strategies. Figure 1 Figure 1. Disclosures Troup: Bio-Rad Laboratories: Employment.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 10
    Publication Date: 2013-11-15
    Description: Somatic mutations that activate Ras signaling are common in hematologic malignancies. They are particularly associated with juvenile and chronic myelomonocytic leukemias, which are classified as ‘overlap’ myelodysplastic syndromes / myeloproliferative neoplasms (MDS/MPN). These diseases are characterized by myelomonocytic proliferation and dysplasia leading to anemia and thrombocytopenia. We have previously shown that conditional expression of KrasG12D in mice models MDS/MPN, producing a fatal disease with increased granulocyte/macrophage (G/M) progenitors and a reduction in the megakaryocyte/erythroid (Meg/E) lineage. Despite intensive study of Ras signaling, the mechanisms by which oncogenic Ras regulates cell fate decisions in hematopoiesis remain unclear; better understanding of these processes may allow development of novel therapies for MDS/MPN. We found that mRNA and protein levels of dual specificity phosphatase-6 (Dusp6), a negative regulator of MAPK signaling, is elevated in the bone marrow of KrasG12D mice. This suggested that a negative feedback loop generated by Dusp6 might regulate the proliferation and differentiation of hematopoietic progenitors in MDS/MPN. To test this hypothesis, we conditionally expressed KrasG12D from its endogenous locus in the bone marrow of wild type and Dusp6 knockout mice. Interestingly, many characteristics of MDS/MPN were more severe in Dusp6-/-; KrasG12D mice. For example, we observed significantly lower hemoglobin levels in 3-4 week old Dusp6-/-; KrasG12D mice (8.35g/dL, n=4) compared to Dusp6+/+; KrasG12D mice (11.7g/dL, n=10). Moreover, the peripheral blood of Dusp6+/+; KrasG12D mice contained approximately 13% of reticulocytes (n=12), whereas Dusp6-/-; KrasG12D mice had nearly 36% reticulocytes (n=4) in the peripheral blood. Furthermore, we observed acanthocytes, poikilocytes and other structural abnormalities in the red blood cells from Dusp6-/-; KrasG12D mice and not in mice wild type for Dusp6. Thus, Dusp6 loss accelerates the onset of anemia caused by KrasG12D, which suggests an exacerbation of the erythroid differentiation defect. We did not observe any significant differences in white blood count or spleen size attributable to Dusp6 genotype. Differentiation of hematopoietic progenitors was assessed in the bone marrow by flow cytometry. The frequency of pre-Meg/E progenitors (Lin-Sca1-Kit+CD34+FcgR-CD150+CD105-) from Dusp6-/-; KrasG12D mice was reduced by approximately 50% when compared to Dusp6+/+; KrasG12D mice. Consistent with this result, Dusp6-/-; KrasG12D mice possessed a significantly greater percentage of immature red blood cells (proerythroblasts; CD71hi Ter119+) and fewer mature red blood cells (orthochromatic erythroblasts; CD71+ Ter119+) than bone marrow cells from Dusp6+/+; KrasG12D mice. By contrast, Dusp6 expression does not have an effect on the frequency of pre-G/M cells (Lin-Sca1-Kit+CD34+FcgR-CD150-CD105-), which remains unchanged between Dusp6+/+; KrasG12D and Dusp6-/-; KrasG12D mice. Together, these findings demonstrate that negative feedback through Dusp6 functions to maintain erythropoiesis the context of oncogenic Ras signaling. Futher, these data strengthen the association between excessive MAPK activity and dyserythropoiesis, and further support development of inhibitors of MEK and/or ERK to treat MDS/MPN. Disclosures: No relevant conflicts of interest to declare.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...