GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 962-962
    Abstract: OBJECTIVES: DNA vaccines can be effective in the acquisition of humoral and cellmediated immune responses. Our studies on an acute promyelocytic leukemia (APL) mouse model show that DNA vaccination combined with all-trans retinoic acid (ATRA) results in a survival advantage with a significant increase in the Th1 cytokine IFNg. ATRA alone can act as an adjuvant to induce immune responses as measured by an increase in anti-RARa antibody production, which correlated with improved survival in mice. Similar increases in antibody production have been observed in our patients after maintenance therapy. The aim of this study is to use immunomonitoring and functional assays to evaluate the presence of activated T-cells and to demonstrate APL-specific killing and to determine if the protective effect of DNA vaccination is CD4+ and CD8+ mediated. METHODS: Using an APL transplant model in FVB/N mice, CD107a, expressed on the surface of lytic granules of activated T-cells, was measured by flow cytometry. A flow based CFSE assay was used to measure APL specific cell killing by cytotoxic T-cells (CTLs). As FVB/N mice have H2q haplotyes, blocking anti-H2q antibodies were used to determine if the cytotoxic activity was MHC restricted. Immunophenotyping by measuring CD4+ and CD8+ absolute counts were conducted. Mice injected with APL cells were depleted of CD4+ or CD8+ cells with anti-CD4 or anti-CD8+ antibody treatment initiated the day after DNA vaccination (early) and continued every 5 days and assayed for efficacy of the DNA+ATRA combined therapy or CD4+ or CD8+ cells were depleted in long term survivors ( 〉 100 days, late). RESULTS: Th1 cytokines TNFa and IFNg were increased indicative of DNA effects and specific activated CD3/CD8 T cells were detected and observed to release cytotoxic granules in the presence of APL cells in long term survivors. A dose dependent decrease in CFSE positive cells was observed assaying effectors from spleens of ATRA alone, ATRA+DNA treated mice and CD107a+ sorted cells from the latter using APL cells as targets. This effect was MHC restricted as anti-H2q antibodies reduced the specific cytotoxic activity. CD4+ absolute numbers measured on day 38 significantly correlated with survival (p=0.005). Although not significant a similar trend was observed for CD8+ counts. The CD4+ or CD8+ depleted mice treated with DNA + ATRA died earlier compared with the undepleted animals. When DNA + ATRA treated long term survivors were depleted of CD4+ or CD8+ cells, the CD4+ depleted mice relapsed and died in 3 months whereas the CD8+ depleted mice survived for a further 3 months when the experiment was terminated. These data are consistent with an increase in anti-RARa antibody production previously measured in other protocols. Interestingly the late depletions show that CD4+ cells are required for the maintenance of the remissions and show that memory T-cells are required. CONCLUSION: Therefore we have been able to detect protective cellular and humoral responses in mice with the combined treatment of DNA+ATRA, which correlates with outcome.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 235-235
    Abstract: The combination of ATRA and ATO is very effective in treating APL, both in mouse models (Lallemand JEM 1999) and in patients (Lo Coco NEJM 2013). Having previously shown that the combination of ATRA and a specific DNA vaccine (pCDNA3PML-RARAFrC) rescues APL mice from relapse and enhances specific immune responses (Padua Nat Med 2003; Robin Blood 2006; Furugaki Blood 2010; Pokorna Mol Cell Probes 2013), we applied this approach using a novel non-specific DNA, pVAX14, as add-on therapy to ATRA +/- ATO. Methods APL transplanted mice (APL cells injected in syngeneic 6-week old mice, Brown PNAS 1997) were treated with various combinations of ATRA +/- ATO and DNA (pVAX1 (control DNA), pCDNA3PML-RARAFrC (specific DNA vaccine) or pVAX14. pVAX14 is a novel construct containing GC-rich sequences and coding for unique peptides, 3 of which we have shown to be immunogenic. Mice were monitored for survival, MRD (by RQ-PCR of PML-RARA) on days 19, 37, 57 and 67 in peripheral blood (PB), and as a measure of LICs by secondary transplantation of bone marrow (BM) or spleen cells from long term (more than 120 days) survivors (LTS) into syngeneic lethally irradiated mice. Immune responses were assessed on CD4 Memory T-cells (Tmem), IFNg production (ELISPOT) and anti-RARa (ELISA) antibody production. Functional studies including challenge experiments of LTS with leukemic cells assessed the protective effects of immunotherapy. Results 1) Survival . In a first set of experiments, ATRA+pVAX14 significantly increased survival compared to placebo, ATRA alone or ATRA+pVAX1 (control DNA) (p 〈 0.001, p 〈 0.03, p 〈 0.05 respectively (Fig. 1); survival was similar with ATRA+pVAX14 and ATRA+pCDNA3PML-RARAFrC, with about 40% LTS. In a second set of experiments where ATO was introduced, ATO+pVAX14 was not superior to ATO alone, but ATRA+ATO+pVAX14 combination was significantly better than ATRA+ATO (p 〈 0.05), providing 85% LTS (Fig. 2). MRD for PML-RARA in ATRA+ATO+pVAX14 mice was significantly lower than in ATRA mice (p 〈 0.001) but the difference with ATRA+ATO mice was not significant up to day 67. Moreover, when we injected BM or spleen cells from both ATRA+ATO and ATRA+ATO+pVAX14 LTS none of the recipients develop APL (follow-up period of 140 days), indicating the clearance of LICs. 2) Immune response monitoring showed that effector Tmem were significantly higher in ATRA+ATO+pVAX14 〉 ATRA+ATO 〉 ATRA (p 〈 0.05 and p 〈 0.001 respectively). LTS of ATRA+ATO+pVAX14 had increased IFNg producing lymph node cells when stimulated with APL cells or PML-RAR peptide compared to ATRA+ATO (p 〈 0.02). Serum anti-RARa antibodies were significantly increased in all combinations compared to controls with ATRA+ATO+pVAX14 〉 ATRA+pVAX14 ≈ ATRA+ATO+pVAX1 (control DNA) ≈ ATRA+ATO 〉 ATRA+pVAX1 (Control DNA) 〉 ATRA (p 〈 0.05, p 〈 0.03, p 〈 0.0006 respectively). When LTS mice of ATRA+ATO+pVAX14 and ATRA+ATO groups were challenged with APL spleen cells, survival was significantly higher in both groups (median survival 105 vs 80 days, p=NS) than in controls (syngeneic mice injected with APL cells) (p 〈 0.001 and p 〈 0.01 respectively) Conclusion The combination of the non-specific vaccine (pVAX14) with ATRA+ATO further improved survival over ATRA+ATO alone and elicited specific humoral and T-cell mediated responses targeting LICs. This vaccine is also effective in a mouse model of high risk MDS (Submitted to ASH 2103). SP, LG and CLP contributed equally to this work. Disclosures: Patel: CEFIPRA: Research Funding. Fenaux:Celgene: Honoraria, Research Funding. Chomienne:Vivavacs: Equity Ownership, I have patents pending through INSERM/Paris-Diderot related to technology employed in this present study. , I have patents pending through INSERM/Paris-Diderot related to technology employed in this present study. Patents & Royalties, Membership on an entity’s Board of Directors or advisory committees. Padua:Vivavacs: Equity Ownership, I have patents pending through INSERM/Paris-Diderot related to technology employed in this present study. I, I have patents pending through INSERM/Paris-Diderot related to technology employed in this present study. I Patents & Royalties, Membership on an entity’s Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3308-3308
    Abstract: Abstract 3308 Background: Animal models enable us to understand disease progression and provide us with reagents to test various therapeutic strategies. We have previously developed a mouse model of myelodysplasia/acute myelogenous leukemia (MDS/AML) progression using mutant NRASD12 and overexpression of human hBCL-2 (Omidvar et al Cancer Res 67:11657-67, 2007). Expanded leukemic stem cells (LSC) were identified as Lin-/Sca1+/KIT+ (LSK) populations, with increased myeloid colony growth and were transplantable. Increased hBCL-2 and RAS-GTP complex were observed in both MDS/AML diseases. The MDS-like disease had increased apoptosis, whilst the AML-like mice had liver apoptosis patterns similar to wild type. The single NRASD12 line also had increased apoptosis. In this present study using a BCL-2 homology domain 3 (BH3) mimetic ABT-737 (Abbott), we have evaluated the effects of targeting BCL-2 in our preclinical models. Methods & Results: Treatment with the inhibitor shows a reduction of LSK cells, reduced progenitor numbers in colony assays and clearance of the liver infiltrations in both MDS and AML models. Gene expression profiling of the MDS mice shows regulation of 399 genes upon treatment including 58 genes expressed by the single mutant RAS mice and not expressed in the untreated AML mice. 78 genes were shared between single NRASD12 and diseased mice and not the treated mice. These studies potentially identify the contribution of NRASD12 genes to disease progression. By confocal microscopy we observed that in the MDS mice the majority of the RAS and BCL-2 co-localized to the plasma membrane, where active pro-apoptotic RAS is normally located, whereas in the AML disease RAS and BCL-2 co-localized in the mitochondria, where BCL-2 is normally found (Omidvar et al 2007). After treatment with the inhibitor the AML co-localization of RAS and BCL-2 shifted to the plasma membrane where single NRASD12 is normally localized. Furthermore, increased RAS-GTP levels was detected in both Sca1+ and Mac1+ enriched spleen cells and interestingly an increase in BCL-2 expression was observed in peripheral blood and in spleen cells after treatment; this increase in BCL-2 was associated with a decrease in the phosphorylation of serine 70 and an increase in phosphorylation of threonine 56 of BCL-2. ABT-737 treatment led to increased phosphorylated ERK resembling RAS and reduced MEK and AKT phosphorylation, changes detected by western blots and the nanoimmunoassay (NIA, NanoPro, Cell Biosciences) that might account for the increased apoptosis, measured by TUNEL and In vivo imaging by single-photon emission computed tomography (SPECT) using Tc-99m-labelled AnnexinV (SPECT). In contrast, although treated MDS mice had increased apoptosis they did not have an increase in overall expression of BCL-2 or in RAS-GTP levels. Treatment of both MDS and AML models with this inhibitor significantly extended lifespan from diagnosis with mean survival of 28 days untreated vs 80 days treated (p=0.0003) and mean survival from birth of 39 untreated vs 85 days treated (p 〈 0.0001) respectively Conclusions: Genomics, proteomics and imaging have been employed in the MDS/AML models to characterize disease progression and follow response to treatment to the BH3 mimetic ABT-737 in order to gain molecular insights in the evaluation of the efficacy. ABT-737 appears to target LSCs, induce apoptosis, regulating RAS and BCL-2 signalling pathways, which translated into significantly increased survival. Disclosures: Padua: Vivavacs SAS: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Auboeuf:GenoSplice technology: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. de la Grange:GenoSplice technology: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties. Fenaux:Celgene: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Janssen Cilag: Honoraria, Research Funding; ROCHE: Honoraria, Research Funding; AMGEN: Honoraria, Research Funding; GSK: Honoraria, Research Funding; Merck: Honoraria, Research Funding; Cephalon: Honoraria, Research Funding. Tu:Cell Biosciences Inc;: Employment. Yang:Cell Biosciences Inc;: Employment. Weissman:Amgen, Systemix, Stem cells Inc, Cellerant: Consultancy, Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Felsher:Cell Bioscience:. Chomienne:Vivavacs SAS: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 16 ( 2013-10-17), p. 2864-2876
    Abstract: BCL-2 homology domain 3 mimetic inhibitor ABT-737 targets leukemia initiating cells and progenitors. Dephosphorylates RAS signaling proteins and regulates proliferation and differentiation genes detected by gene expression profiling.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 200-200
    Abstract: Genomic instability is the driving force of disease progression to frank leukemia. Evidence suggests that aberrant repair of double strand breaks (DSB) by non homologous end-joining (NHEJ), a major repair pathway in mammalian cells, can lead to chromosomal instability and cancer. We previously reported significantly increased error-prone NHEJ in preleukemic syndromes, and a variety of myeloid malignancies, and demonstrated that these cells harbor constitutive DNA damage. We postulated that increased NHEJ misrepair may be a response to the increased DNA damage. Here, we have studied a mouse model for myeloproliferative/myelodysplastic syndrome (MPD/MDS) to determine whether the frequency of DNA damage and aberrant NHEJ repair may be an indicator for genomic instability as the disease progresses. Transgenic mice bearing mutant NRAS and BCL-2 driven by the MRP8 promoter, which directs expression of the transgene to committed myeloid progenitors and neutrophils, have a relatively mild phenotype with an increase of immature neutrophils. The BCL2 mice have an increase in marrow blasts, but have normal blood counts. Transgenic mice harboring both mutant NRAS and BCL2 genes results in a disease phenotype morphologically resembling human late MDS (FAB subtypes refractory anemia with excess blasts (RAEB), RAEB in transformation (RAEBt) or chronic myelomonocytic leukaemia (CMML)) with increased marrow blasts. We show that the bone marrow and spleen from the NRAS and BCL2 mice demonstrate an increase in the frequency of NHEJ misrepair activity, compared with normal (FVBN) mice (NRAS: 7.6% vs 3.7%, BCL2: 6.5% vs 3.7%, n=3). Strikingly, the NRAS +BCL2 double transgenic mice show a large and significant increase in NHEJ misrepair activity (19.02%, n=3, p 〈 0.001), compared with controls and single transgenics. Using an immunofluorescence-based assay for DNA damage, dependent on BrdU incorporation, we find that the magnitude of DNA damage mirrors NHEJ activity. Chromatin fibers from both NRAS and BCL2 mice demonstrate an increase in the frequency of DNA damage, compared to normal mice (NRAS: 35% vs 8%, mean [n=3]), (BCL2 22% vs 8%, [n=3] ). However, this damage increases even further in RAS +BCL2 mice (62% vs NRAS/BCL2 28%, [FVBN] 8%, n=3, p 〈 0.001). This DNA damage co-localizes with the variant histone γH2AX, a key protein in the repair of DSB. DNA damage and γH2AX also co-localize with the NHEJ protein Ku86 emphasizing that DNA damage is linked to repair by NHEJ in situ. Given that activated RAS produces increased reactive oxygen species (ROS), an established source for DSB, we considered whether ROS accounted for some of this DNA damage. We find that cells from transgenic mice show an increase (up to 2-fold) in ROS, compared with controls. The same is true for FDCP1 murine cells transduced with NRAS and BCL2, and treatment with the antioxidant, N-acetyl cysteine results in an up to 50% decrease in ROS, DNA damage and concomitant NHEJ misrepair activity. Our data suggest that increased DNA damage and error-prone repair may be a platform for the creation of increased genomic instability with disease progression in MPD/MDS in mice. Decreased DNA damage and error-prone repair with antioxidant treatment suggests a mechanism for the amelioration of the activities that drive disease progression.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 5497-5497
    Abstract: Introduction: Targeted drugs are needed for HR-MDS/AML, particularly in elderly patients and Venetoclax, approved for some CLL, gives promising results in elderly AML. Assays to predict response to treatment may enable us to deliver personalized treatment. We sought to determine the most informative assay to predict response; viability assays can directly measure the effects of reagents on growth. Progenitor assays can potentially determine if the reagents can target diseased primitive cells. PET scanning can be used to follow response to treatment. Methods: Peripheral blood (PB) or bone marrow (BM) from 7 MDS/AML patients were incubated in a) no treatment, b) ABT-199 (1 µM) (Abbvie), c) GDC-0973 (1 µM) (Genentech) or d) ABT-199+GDC-0973 (1 µM of each) and assessed for viability using the MTT assay (n=2); cell death followed using the Incucyte® Zoom System (Essen Bioscience) (n=2) or methocult progenitor assays (Stem Cell Technologies) (n=4). Having shown that RAS:BCL-2 co-localization correlated with prognosis in MDS/AML patients (Leuk Res 37:312-9, 2013), immunofluorescence was undertaken. A micro PET device dedicated to mice was used to measure BM blast proliferation. After injection of 18F-FLT(a thymidine analogue) in mice untreated (n=7) or ABT-199 (75mg/kg)+GDC-0973(10mg/kg) treated (n=5) normal FVB/N, HR-MDS mice treated with vehicle (n=4), 2-month old HR-MDS before (n=5) and 3-month old before (n=4) and after ABT-199 (75mg/kg)+GDC-0973(10mg/kg) treatment (n=8), PET imaging was performed (Inveon Siemens Medical Systems), analyzed for signal and quantified. Results: Patient details and results are summarized on Table 1. Using the MTT assay 2 PB patient samples were found to be sensitive to ABT-199 treatment (Figure 1A, AS, p=0.00042 and YA, 0.00002) and more sensitive to the combination compared to untreated (AS, p=0.00007 and YA, 0.000003). With the incucyte the BM of one patient (AE) was found to be resistant to both ABT-199 and GDC-0973, but sensitive to the combination (Figure 1B). PB and BM from patient JA were assayed for apoptosis with the incucyte and were found to be sensitive to ABT-199 with increased apoptosis, resistant to GDC-0973 with decreased apoptosis and sensitive to the combination. Four bone marrow samples were tested in the 4 conditions using the progenitor assay (Figure 1C). Three patients were sensitive to GDC-0973, inhibiting any colony formation and the fourth had reduced colony numbers. In this assay patient JA appeared to be sensitive to GDC-0973 treatment whereas the incucyte assay scored this sample to be resistant to apoptosis; thus the cytotoxic effects of GDC-0973 may not be via apoptopsis. As the progenitor assay is likely to score the primitive disease population, this assay may prove more informative than the others without prior selection. One patient (DH) was clearly resistant to ABT-199, whereas the other three (JA, CB and FL) had reduced colony growth. All patients were sensitive to the combination treatment and inhibited colony growth. The RAS:BCL-2 co-localization in the PB revealed no complex in either the Mito or PM upon treatment with ABT-199 alone and some localization in the Mito with GDC-0973. With both ABT-199 and GDC-0973, there were hardly any cells confirming the cytotoxic effects of the combination. As we have previously shown that PM co-localization of the complex is associated with drug resistance (Blood 130:2613, 2017Suppl), we used the combination on our HR-MDS mouse model, where the complex co-localizes in the PM and followed the mice by PET scanning (Figure 1D). Weak signal was visualized in the femurs of untreated and ABT-199+GDC-0973 treated FVB/N mice (FBR 1.17+/-0.34 and 1.02+/-0.08 respectively). Mild PET signal was seen in the femurs of 2 month-old HR-MDS mice, (FBR 1.79+/-0.98). Intense PET signal was seen in the femurs and proximal humerus of HR-MDS mice treated with vehicle (3 month-old, FBR=2.35+/-1.32). Low PET signals were seen in the femurs of 5/8 HR-MDS mice treated with ABT-199+GDC-0973 (FBR=1.93+/-0.84). FBRs of the 3 groups of HR-MDS mice were significantly higher than those of FBV/N groups. Conclusion: Combined Venetoclax (ABT-199) and GDC-0973 targets MDS/AML progenitors and can potentially overcome drug resistance with the disruption of the RAS:BCL-2 complex. Bone marrow disease progression in HR-MDS mice can be monitored with 18F-FLT-PET imaging; PET data shows that the combination slows down disease progression. Disclosures Padua: Abbvie: Research Funding; Genentech: Research Funding. Giraudier:Novartis: Research Funding. Konopleva:Stemline Therapeutics: Research Funding. Andreeff:Oncoceutics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; United Therapeutics: Patents & Royalties: GD2 inhibition in breast cancer ; Reata: Equity Ownership; Celgene: Consultancy; Jazz Pharma: Consultancy; Oncolyze: Equity Ownership; Amgen: Consultancy, Research Funding; Eutropics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Aptose: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Consultancy, Patents & Royalties: MDM2 inhibitor activity patent, Research Funding; SentiBio: Equity Ownership; Astra Zeneca: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 115, No. 3 ( 2010-01-21), p. 653-656
    Abstract: DNA vaccination and all-trans retinoic acid (ATRA) result in a survival advantage in a mouse model of acute promyelocytic leukemia (APL). Depletion of CD4+ or CD8+ cells abolished this effect. CD4+ depletions of long-term survivors resulted in relapse and death within 3 months, thus demonstrating the need of both CD4+ and CD8+ subsets for the generation of DNA-driven antileukemic immune responses and underscoring a crucial role of CD4+ cells in the maintenance of durable remissions. Degranulation and cytotoxic carboxyfluorescein diacetate succinimidyl ester–based assays showed major histocompatibility complex–restricted APL-specific T cell–mediated immune responses. Sorted APL-specific CD8+CD107a+ T cells showed an increase of antileukemic activity. Effectors from ATRA + DNA–treated mice were shown to secrete interferon-γ when stimulated with either APL cells or peptides from the promyelocytic leukemia-RARα vaccine-derived sequences as detected by ELISpot assays. Our results demonstrate that DNA vaccination with ATRA confers the effective boosting of interferon-γ–producing and cytotoxic T cells in the leukemic mice.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 118, No. 7 ( 2011-08-18), p. 1784-1796
    Abstract: The mechanisms regulating the emergence of BM prothymocytes remain poorly characterized. Genome-wide transcriptome analyses looking for genes expressed in human prothymocytes led to the identification of AF1q/MLLT11 as a candidate gene conceivably involved in this process. Analysis of AF1q protein subcellular localization and intracellular trafficking showed that despite pronounced karyophily, it was subjected to constitutive nuclear export followed by ubiquitin-mediated degradation in the centrosomal area. Using in vitro assays based on either forced expression or shRNA-mediated silencing of AF1q, we provide evidence that the protein promotes T- over B-cell differentiation in multipotent hematopoietic progenitors. At the molecular level, AF1q confers to multipotent progenitors an increased susceptibility to Delta-like/Notch-mediated signaling. Consistent with these findings, enforced AF1q expression in humanized mice fosters the emergence of BM CD34+CD7+ prothymocytes, enhances subsequent thymus colonization, and accelerates intrathymic T-cell development. In contrast, AF1q silencing provokes a global shift of BM lymphopoiesis toward the B-cell lineage, hinders prothymocyte development, inhibits thymus colonization, and leads to intrathymic accumulation of B cells. Our results indicate that AF1q cooperates with the Notch signaling pathway to foster the emergence of BM prothymocytes and drive subsequent intrathymic specification toward the T-cell lineage.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1510-1510
    Abstract: Abstract 1510 Background: Relapses are now relatively rare in APL and occur mainly in the bone marrow (BM) within 3 years of complete response achievement, but later BM relapses can also occur (Kelaidi, Leukemia 2006), while a few cases of extramedullary (EM) relapse are observed, particularly in the central nervous system (CNS) (De Botton, Leukemia 2006). The transplantable transgenic mouse model of APL is a well characterized preclinical model which mimics human APL in its biological characteristics. We have previously reported the response of these mice to ATRA and a combination of ATRA with a DNA vaccine (Padua, Nat Med 2003, Fuguraki, Blood 2010). We analyzed in those treated mice the presence of PML-RARα transcripts in the peripheral blood (PB), BM and various organs and tissues. Methods: The reproducible APL development was obtained, as previously described (Padua Nat Med 2003) by intravenous injection of 104 spleen cells from APL transgenic mice expressing the human PML-RARα cDNA (bcr1) into syngeneic recipients. ATRA and the DNA vaccine (a plasmid containing PML-RARα cDNA (bcr1) sequence fused to the tetanus toxin fragment C) were administered as described (Padua Nat Med 2003). In this model, ATRA alone gives transient remissions, while about 30 % of APL mice treated with ATRA combined with a DNA vaccine achieve long-term remissions. A standardized RT-qPCR MRD monitoring protocol was applied to assess PML-RARα-positive cell clearance in PB and BM. In this assay, the number of PML-RARα transcripts was normalized (normalized copy number or NCN) to 106 copies of 18S rRNA transcripts, allowing us to detect up to 1 PML-RARα-positive cell among 104 negative cells. Taking advantage of the presence of PML-RARα cDNA transgene in the transplanted leukemia cells, we next used genomic DNA as template for a qPCR assay, allowing us to use 10 times more template and increased sensitivity (1 in 105) in order to examine the presence of PML-RARα-positive cells in various organs and tissues of long-term survivors (LTS, ie with survival 〉 120 days). Results: APL mice treated with ATRA alone (n=55), ATRA combined with PML-RARαFrC DNA (n=94) or untreated (n=65) were analyzed. Untreated APL mice always remained positive in the PB and BM for PML-RARα transcripts, and in organs and tissues positive for PML-RARα cDNA. On day 60, in the surviving ATRA-treated mice (n=21), 15 (71%) had PB PML-RARα normalized copy number (NCN) 〉 100, 6 (29 %) an NCN between 10–100 and none an NCN 〈 10, while in ATRA+DNA-treated mice (n=35), 11 (31%) had NCN 〉 100, 9 (26%) NCN between 10–100 and 15 (43%) NCN 〈 10 (p 〈 0.01). ATRA+DNA-treated mice achieving NCN 〈 10 (43%) constituted the group with the best survival (p 〈 0.0001). To further assess tumour burden, LTS were sacrificed at different time intervals. No PML-RARα transcripts were detected in PB and BM of any LTS (n=15) suggesting complete molecular remission. On the other hand, while PML-RARα cDNA, analyzed by qPCR in skin, salivary glands, thymus, kidney, muscle, heart, spleen, liver, lung and CNS was negative in all tissues in 10 (67%) LTS, it was positive in 5 (33%) LTS, including 4 in the CNS (2 of them were also positive in the spleen) and 1 in the spleen only. Conclusion: In this preclinical model, analyzed with sensitive molecular assays, while two thirds treated long-term survivors were in molecular remission in PB, BM and other organs studied, about one third still had leukemic cells, mainly in the CNS and to a lesser extent in the spleen. This model could be of interest to better understand relapses in APL patients, especially late and CNS relapses and to evaluate drugs aimed at eliminating those reservoirs of residual cells. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 2614-2614
    Abstract: Abstract 2614 We previously demonstrated that although retinoic acid (RA) has targeted efficacy in Acute Promyelocytic Leukemia (APL), heterogeneity exists leading to the appearance of un-targeted clones at the time of relapse. Characterization of these clones is not yet fully unraveled though we and others have previously highlighted the roles of RARα mutations, pharmacogenomics or APL miRNome. We recently identified that the ERK1/2 pathway synergized with RA to restore the transcriptional activity of RA in resistant APL cells, thus restoring RA induced differentiation (Cassinat et al. Mol Cell Biol 2011). These results suggest that targeting interconnected signaling pathways could optimize differentiation therapy efficacy. To this effect, we studied known signaling pathway activators or inhibitors that could potentiate with RA and identified Lithium chloride (LiCl). Treatment of the ATRA sensitive-APL NB4 cell line with LiCl (25mM) decreases proliferation and increases apoptosis (25% and 40% of Annexin V-positive cells at day 1 and 2 respectively) with evidence of caspase 3 cleavage at day 2. Because NB4 cells fully differentiated with RA alone we were unable to observe any synergy when combined with LiCl. Treatment of the RA-resistant APL UF-1 cell line with RA or LiCl alone does not induce differentiation. Combination of RA+LiCl restores differentiation after 3 days of culture (65% CD11b positive and 55% NBT test positive cells). Similar results were obtained with different GSK3 inhibitors, suggesting that the LiCL effects were in part linked to its well characterized GSK3 inhibitory activity. Interestingly, we noted that LiCl treatment induces rapid phosphorylation of ERK1/2 and pretreatment with the MEK/ERK1/2 inhibitor UO126 fully abolished the differentiation induced by the RA+LiCl combination. The combination restores in UF-1 the expression of RA target genes (such as RARα2) to the same levels obtained in NB4 cells treated by RA alone. The level of luciferase activity of an RA responsive element reporter gene was increased with the RA+LiCl combination compared to RA alone. Both target gene expression and luciferase activiy were abolished after inhibition of the MEK/ERK1/2 pathway. Thus, increase in differentiation of UF-1 cells by RA+LiCl is linked to increased RA transcriptional activation. Similar studies in fresh APL patient cells confirmed both the increase in differentiation and level of RA target gene expression and their inhibition by UO126. Finally, to translate these findings in vivo, we used the APL-transplantable mouse model. Plasma lithium levels in treated mice were measured between 0.6 and 1.05 mmol/l, levels reached in humans. When LiCl was combined with RA we repeatedly observed a pronounced survival advantage compared to mice treated by RA alone as evaluated by Kaplan Meier analysis. In this work we demonstrate that LiCl, a well tolerated agent in humans, has the potential, when combined with RA, to restore RA induced transcriptional activation and differentiation in RA resistant APL cells. Furthermore, this combination also increases RA efficacy in an in vivo APL mouse model. Disclosures: Off Label Use: Lithium is a mood modulator administered for bipolar disorders.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...