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. 139, No. 16 ( 2022-04-21), p. 2427-2440
    Abstract: Inherited bone marrow failure syndromes (IBMFSs) are a group of disorders typified by impaired production of 1 or several blood cell types. The telomere biology disorders dyskeratosis congenita (DC) and its severe variant, Høyeraal-Hreidarsson (HH) syndrome, are rare IBMFSs characterized by bone marrow failure, developmental defects, and various premature aging complications associated with critically short telomeres. We identified biallelic variants in the gene encoding the 5′-to-3′ DNA exonuclease Apollo/SNM1B in 3 unrelated patients presenting with a DC/HH phenotype consisting of early-onset hypocellular bone marrow failure, B and NK lymphopenia, developmental anomalies, microcephaly, and/or intrauterine growth retardation. All 3 patients carry a homozygous or compound heterozygous (in combination with a null allele) missense variant affecting the same residue L142 (L142F or L142S) located in the catalytic domain of Apollo. Apollo-deficient cells from patients exhibited spontaneous chromosome instability and impaired DNA repair that was complemented by CRISPR/Cas9-mediated gene correction. Furthermore, patients’ cells showed signs of telomere fragility that were not associated with global reduction of telomere length. Unlike patients’ cells, human Apollo KO HT1080 cell lines showed strong telomere dysfunction accompanied by excessive telomere shortening, suggesting that the L142S and L142F Apollo variants are hypomorphic. Collectively, these findings define human Apollo as a genome caretaker and identify biallelic Apollo variants as a genetic cause of a hitherto unrecognized severe IBMFS that combines clinical hallmarks of DC/HH with normal telomere length.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    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. 118, No. 21 ( 2011-11-18), p. 2790-2790
    Abstract: Abstract 2790 Background: TKIs were initially developed as targeted therapies that would solely interfere with a “specific” aberrant signaling pathway in malignant cells. However, we and others showed that the EGFR TKI erlotinib (Erlo) has in vivo and in vitro efficacy in MDS and AML (Boehrer et al., Blood, 2008). We also previously observed, in a preliminary study, the potentiation of apoptosis upon combination of Erlo with azacitidine (Aza), but not with decitabine, in HL-60 cells and in cells from a few (five) MDS/AML patients (ASH 2010, 974). We decided to expand this pre-clinical study to define the potential interest of combining different TKI (Dasatinib (Dasa), Sorafenib (Sora) or Erlo) with Aza, now a reference first line treatment in higher risk MDS (Lancet Oncol, 2009). Methods: Erlo (10μM), Dasa (500nM), or Sora (5μM) were combined to Aza (1μM) and apoptosis over-time (24, 48 and 72h) quantified by FACS analysis following DioC3(6)/PI staining in different MDS/AML-derived cell lines (MOLM-13, SKM-1, MV4-11, Kasumi-1). Quantification of apoptosis at 48h or 72h was recapitulated ex vivo in CD34+ cells from patients with MDS (n=12), AML (n=14) or AML post MDS (n=5). For each single drug, as well as the respective combinationsErlo, Aza and Erlo+Aza, the capacity to induce cell cycle arrest (PI staining), cytotoxicity (MTT assay) and decrease of proliferation (Click-it EdU Assay) was assessed concomitantly. Differentiation was assessed by staining with CD11b on day 3 (FACS), evaluation of ROS production at 24/48h by FACS staining with CM-DFCDA (oxidative stress indicator) and HE (Hydroethidine). Functional relevance of apoptosis-related signaling pathways was determined by co-incubation of Aza (and drugs combinations with biochemical inhibitors against MAPK: JNK (SP600125, 10μM), p-38MAPK (SB203580, 10μM) and MEK (U0126, 5μM). Immunoblot analyses of caspase-3, PARP, Mcl-1 and Bcl-xl were performed at 24h and 48h. Results: Whereas co-incubation of Dasa or Sora with Aza did not increase the degree of apoptosis observed with Aza alone, combination of Erlo with Aza had synergistic effects already observed at 24h, an effect that increased over-time (SKM-1 72h (mean PI+ cells, n=3), Erlo: 15%, Aza: 40%, Erlo+Aza: 81% - MOLM-13 72h, Erlo: 21%, Aza: 25%, Erlo+Aza: 64%). The % of cells with intact metabolic activity at 72h (determined by the MTT assay using 2.5μM Erlo and 0.5μM Aza) decreased from 83% (Erlo) and 79% (Aza) to 23% (Erlo+Aza) in SKM-1 (similar results for MOLM-13 cells). To determine if Erlo also impacts on apoptosis in CD34+ cells from patients with MDS or AML, we screened 31 samples and observed a synergistic effect in 5/31 samples (2/12 MDS, 2/14 LAM, 1/5 AML post MDS) and an additive effect in 8/31 samples. Induction of apoptosis was not preceded by differentiation (no increase in CD11b) or production of ROS. On the other hand, analysis of cell cycle distribution at 24h showed that apoptosis was accompanied by an increase in G0/G1 arrest and a decrease in the % of cells in S phase upon incubation with the combination of Erlo+Aza (G0/G1 (mean, n=2) in SKM-1, DMSO: 51%, Erlo: 52%, Aza: 61%, Erlo+Aza: 78%, G0/G1 in MOLM-13, DMSO: 40%, Erlo: 51%, Aza: 38%, Erlo+Aza: 65%) confirmed by the study of newly synthesized DNA before induction of apoptosis (15h) with an alternative BrdU test (Click-EdU assay). Indeed, in SKM-1 cells, proliferation was decreased by 20% upon Erlo or Aza and by 80% with the association. Moreover, cell cycle arrest was associated with an increase in the % of apoptotic cells with a sub-diploid DNA content (about 10% for Erlo or Aza and 32% for Erlo+Aza in SKM-1. Immunoblot analyses confirmed an increase in cleaved PARP and caspase-3 upon incubation with Aza +Erlo as compared to the single agents, as well as a decrease of the anti-apoptotic protein Mcl-1 and Bcl-xl. Incubation of Aza with SP600125 induced the same extent of apoptosis observed with Erlo+Aza, whereas no effect was observed with the p-38MAPK inhibitor SB203580 or the MEK inhibitor U0126. In addition, Erlo decreased phosphorylation of JNK on Thr183/Tyr185, suggesting a potential implication of the JNK pathway in the mechanism of apoptosis. Conclusions: In this study, Erlo was the only TKI tested able to increase sensitivity towards Aza in MDS/AML cell lines and in patient-derived cells. These results suggest a potential clinical interest of combining Aza to Erlo in MDS/AML. Disclosures: Fenaux: Celgene: Honoraria, Research Funding.
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 768-768
    Abstract: Spleen dysfunction and susceptibility to pneumococcal infection is a well known feature in homozygous sickle cell disease (HbSS), whilst to date splenic function in hemoglobin SC disease (HbSC) has been poorly investigated. The aim of this study was to analyze spleen function in children with HbSC disease using a high-throughput validated method (1) and to examine if the current recommendations regarding pneumococcal risk are appropriate in this population. Spleen function was evaluated using a flow cytometry quantification of red blood cells (RBCs) with Howell-Jolly bodies (HJBs), in a cross-sectional study of patients at steady state during an outpatient visit in an expert center. Quantification of HJB-RBCs was performed in children with HbSC disease aged & lt; 10 years and compared to children with HbSS disease or healthy children of the same age groups, or splenectomized children. Additional exploratory analysis was performed according to age (under or above the age of 5 years old) and treatment group (hydroxyurea). The median (Q1-Q3) HJB-RBCs count was 16 (11-28.25) /100.000 RBCs in 40 HbSC children (Figure 1). This result was not statistically different from the control group of 22 healthy children (p=0.96) nor in subgroups & lt; or ≥ 5 years old, indicating that children with HbSC under 10 years have a preserved splenic function. Expectedly, the HJB-RBCs counts differed significantly from splenectomized children (419 (296-489)/100.000 RBCs, n=15, p & lt;0.0001). By contrast, among the 53 HbSS children, the median HJB-RBCs count was 134 (29-216) /100.000 RBCs, differing significantly from HbSC children (p & lt;0.0001). In HbSS children, HJB-RBCs counts increased significantly with age (r=0.30, p=0.03), showing important variability among subjects but did not reach the level found in splenectomized patients suggesting that complete loss of spleen function occurs presumably later in a majority of children in this population. Treatment with hydroxyurea did not significantly impact HJB-RBCs counts in a subgroup analysis in HbSS children. The result of this study suggests that spleen function in children under 10 years old with HbSC is not altered. The routine administration of prophylactic penicillin to young children with SC disease may therefore be questioned. Similarly, fever in children with HbSC under 3 years old may not require parenteral antibiotics as it is generally currently recommended by analogy to children with HbSS. Functional or anatomical asplenia in children with HbSC is delayed compared to those with HbSS at least after the first decade of life. Future large cohort studies using similar methodology will allow better evaluation of the pneumococcal risk in adolescents and adults with Hb SC disease. Bibliography (1) El Hoss S, Dussiot M, Renaud O, Brousse V, El Nemer W. A novel non-invasive method to measure splenic filtration function in humans. Haematologica. oct 2018;103(10):e436-9. Figure 1 Figure 1. Disclosures El Nemer: Hemanext: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    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. 136, No. Supplement 1 ( 2020-11-5), p. 4-5
    Abstract: Objectives: Juvenile myelomonocytic leukemia (JMML) is a rare but aggressive myeloproliferative/myelodysplastic neoplasm affecting infants and young children. The narrow age-window of onset suggests that a prenatal environment is needed for JMML oncogenesis. In search of a transcriptional reminiscence of embryo-fetal characteristics that would confirm this hypothesis, we investigated how the gene expression profile of JMML hematopoietic progenitors compared to their healthy counterpart isolated at different stages of ontogeny. Methods: Hematopoietic stem cell and progenitor cell (HSPC) fractions of JMML (n=16), bone marrow (BM) of healthy children (n=7), fetal liver (FL; n=3) and fetal BM (FBM; n=2) were phenotyped and sorted using signatures validated in the fetal and adult BM (Notta et al, Science 2011). RNAseq was performed using the TruSeq® Stranded Total RNASample preparation kit. Unsupervised hierarchical clustering analysis was done with the Bioconductor edgeR package. Differentially expressed genes were identified with the Bioconductor limma package. Results: To eliminate the impact of variations in the HSPC distribution, the JMML transcriptome was assessed on FACS-sorted common myeloid progenitor (CMP), granulocyte-monocyte progenitor (GMP) and megakaryocyte-erythroid progenitor (MEP) cell fractions from 16 JMML and compared to healthy counterparts at different stages of ontogeny (FL, FBM, age-matched children BM). Unsupervised hierarchical clustering separated the samples into 4 groups (C1-4), primarily according to ontogeny, with 14/15 embryo-fetal fractions in C1 and all healthy post-natal progenitors in C2 (CMP, MEP) or C3 (GMP). Most JMML fractions clustered either with the prenatal fractions (C1; 17/47 fractions from 8/16 patients) or in a separate group containing no healthy sample (C4; 23/47 samples from 10/16 patients). Two groups were defined accordingly: one with JMML resembling embryo-fetal samples ('Fetal-JMML'; n=6/16), and a JMML-specific group ('Onco-JMML'; 8/16). Patients with Onco-JMML tended to be older, with a more severe presentation and elevated fetal hemoglobin levels. All PTPN11-mutated JMML were in this group whereas 5/6 Fetal-JMML had NRAS or KRAS mutations. Analysis of differential gene expression between Fetal and Onco-JMML highlighted 344 up-regulated genes versus 19 up-regulated genes in Onco-JMML. Surprisingly, LIN28B and WT1, both known to activate fetal pathways, were the most up-regulated genes in Onco-JMML. These key transcription factors were deregulated as early as the hematopoietic stem cell (HSC) compartment. Gene Set Enrichment Analysis (GSEA) confirmed enrichment in LIN28B and WT1-related signatures and showed enrichment in an AML signature in Onco-JMML. On the other hand, Fetal-JMML showed striking overexpression of monocytic /dendritic cell markers and inflammasome and innate immunity components. GSEA confirmed the strong monocyte identity of Fetal-JMML progenitors compared to onco-JMML or healthy postnatal progenitors. Part of the monocytic markers 'aberrantly' expressed in JMML progenitors was expressed in healthy fetal progenitors. Analysis of the HSC and multipotent progenitor (MPP) fractions showed that up regulation of monocytic markers was limited to the JMML progeny compartments. As we were able to confirm the transcriptional and functional identity of the sorted progenitors, these data suggest an early monocytic priming in these JMML progenitors, reminiscent of the monocyte-biased myelopoiesis characterizing physiologic fetal hematopoiesis. Conclusion: Our findings give a striking example of how ontogeny-related features are involved in childhood oncogenesis. They highlight a strong but complex link beween JMML and development, with a fetal identity resulting either from retention of a physiologic fetal monocytic signature or from aberrant (re)activation of master oncofetal regulators. Intriguingly, although LIN28B is thought to reprogram hematopoietic progenitors into a fetal-like state, its activation does not lead to an overall fetal profile in JMML, suggesting a regulatory mechanism distinct from that of physiological development. These two ontogeny-based signatures are likely to uncover the biology underlying previous classifiers based on AML-like profile or DNA methylation and suggest that a subset of JMML patient may benefit from immunomodulating therapies. Disclosures Dalle: Bellicum: Consultancy, Honoraria; bluebird bio: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi-Genzyme: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Medac: Consultancy, Honoraria; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Gilead: Honoraria; AbbVie Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Orchard: Consultancy, Honoraria; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees. Baruchel:Jazz Pharmaceuticals: Consultancy, Honoraria; Celgene Corporation: Consultancy, Honoraria; Astra Zeneca: Consultancy; Servier: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Shire: Research Funding; Bellicum: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    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. 118, No. 21 ( 2011-11-18), p. 2564-2564
    Abstract: Abstract 2564 Background: Erlotinib (Erlo) was originally developed as an epidermal growth factor receptor inhibitor, yet it also exerts antileukemic “off-target” effects, in vitro and in vivo in MDS and AML (Boehrer et al., Blood, 2008). In a preliminary pre-clinical study, we observed that Erlo increased chemosensitivity to current AML drugs in different AML cell lines and in ex vivo AML patient cells (n=3) (ASH 2010, 2163). Those first results suggested an implication of ABC-transporters in the potentiation of apoptosis. Here, we bring direct evidence for Erlo's ability to hinder efflux pumps and to decrease their expression on AML cells. Methods: Drug efflux via ABC-transporters (substrate: mitoxantrone-MTZ or doxorubicin-Dox), and specific efflux via P-gp (substrates: DioC23 and Rho-123), MRP (s: Calcein and CDCFDA) and BCRP (s: Hoechst 33342) were quantified by FACS following incubation with 10mM Erlo. Intracellular VP-16) content was quantified by Rapid Resolution Liquid Chromatography (RRLC). Biochemical inhibitors of the respective ABC-transporters (CSA (1μM), verapamil (Vera-10μM), MK571 (10μM), KO143 (500nM) served as positive controls. To assess chemosensitivity, 10mM Erlo was combined to AraC (100nM), Dox (100nM), or VP-16 (1mM) and apoptosis over-time (24, 48, 72h) quantified by DioC3(6)/PI staining. Assessment of sensitivity to the drug combinations listed above were carried out in KG-1 cells, and its more immature variant KG-1a and in ex vivo CD34+ marrow cells from AML patients (AML post MDS n=5, de novo AML n=5). P-gp's ATPase activity was quantified with the luminescence-based Pgp-Gloä Assay System. Surface expression of P-gp was determined by FACS analysis and total protein expression of MRP, BCRP and P-gp by immunoblot analysis. Functional relevance of signaling pathways was tested using the SRC inhibitor PP2 (10μM) and the mTOR inhibitor Rapamicin (10nM). Results: We found that I) Erlo inhibited efflux via P-gp, MRP and BCRP as demonstrated by increased intracellular retention of DioC23/Rho-123, Calcein/CDCFDA and Hoechst 33342, respectively, andby its ability to retain MTX (300nM) and Dox (200nM) intracellularly II) Inhibition of drug efflux was higher in KG-1 than in KG-1a cellss, in agreement with a lower expression of P-gp and BCRP on KG-1a as compared to KG-1 cells; III) Quantification of VP-16 by RRLC after incubation with or without Erlo showed the ability of Erlo to increase intracellular VP-16 contents by approximately 60%; IV) Erlo increased ATPase activity in a dose-dependant manner, supporting the notion that Erlo is a competitive inhibitor of P-gp; IV) Erlo combined to VP-16 induced synergistic effects on apoptosis in KG-1 cells, and to a lesser extent in KG-1a (48h KG-1: Erlo 20%, VP-16 38%, Erlo+VP16 78%, KG-1a 48h: Erlo 10%, VP-16: 12%, Erlo+VP16: 35%); V) 48h of incubation with Erlo reduced cell surface expression of P-gp in KG-1 cells by 50%, whereas total P-gp protein expression remained unchanged, suggesting that Erlo interferes exclusively with the protein form expressed on the cell surface, VI) Decrease of P-gp cell surface expression was recapitulated upon incubation with PP2 (10μM) or Rapamicin (10nM); VII) the combination of Erlo+VP-16 in 10 AML-patient samples induced synergistic effects on apoptosis in 5 of them and additive effects in 3 of them. Conclusions: We here confirm that Erlo increases sensitivity towards chemotherapeutic agents subjected to drug efflux via ABC-transporters and delineate the molecular pathways conveying these effects. Disclosures: Fenaux: Celgene: Honoraria, Research Funding.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 5028-5028
    Abstract: Abstract 5028 Background In MDS and AML, P-gp expression is associated with low response rate to conventional anthracycline-AraC chemotherapy (Lepelley et al., Leukaemia, 1994; Mahadevan and List, Blood, 2004). Since hypomethylating agents, notably AZA, can improve survival and have become a reference first line treatment in high and int 2 risk MDS (Lancet Oncol, 2009), we assessed whether P-gp expression and function in ex vivo MDS cells from patients treated with AZA was associated to clinical response to that drug. Methods Bone marrow (BM) cells from 30 patients with MDS or AML post MDS treated with AZA were studied, at onset of AZA. All patients received AZA for at least one cycle (75 mg/m2/d during 7 days every 28 days), and response was evaluated according to IWG 2006 criteria. Mononuclear cells from samples were isolated using a Ficoll-Paque PLUS density gradient. The evaluation of P-gp expression and functionality was made on CD45dim cells (blast population) and CD45dim CD34+ population, gated by flow cytometry. Quantification of P-gp expression was evaluated by FACS, using a phycoeythrin(PE)-conjugated antibody specific for ABCB1(P-gp)(clone UIC2). P-gp functionality was quantified by flow cytometry evaluation of Rhodamine123 (Rhoda: a specific substrate of P-gp) efflux in the presence or the absence of ciclosporine A (CSA, an inhibitor of P-gp). Patient cells were considered Rhoda pos when the RFI (Relative Fluorescence Intensity) CSA+/CSA- was 〉 1.5. Results WHO diagnosis at onset of AZA was MDS in 19 pts (1 RA, 1 RCMD, 1 LMMC2, 5 RAEB-1, 10 RAEB-2, 1 RAEB-t) and AML post MDS in 11 pts. Median age was 76, M/F: 19/11. Karyotype (IPSS) was fav (n=10), int (n=3) unfav (n=14) and a failure (n=3). In MDS, IPSS was int-1 in 3pts, int-2 in 9 pts and high in 5 pts (and not evaluable in 2). The median number of cycles of AZA administered was 6 [2–19]. Fourteen pts (47%) responded including 6 (20%) CR and 8 hematological improvements (HI). Median OS from initiation of AZA was 418 d. No correlation between response and karyotype was found. P-gp expression was found in 64% of pts, and 65% of the pts had Rhoda pos cells, with some discordant cases for P-gp expression and functionality (including 23% P-gp pos Rhoda neg and 10% P-gp neg Rhoda pos). Karyotype was not correlated to P-gp expression or Rhoda RFI. The response rate was 39% in P-gp pos and 71% in P-gp neg pts (p=0.2) and the CR rate 17% in P-gp pos pts vs 28% in P-gp neg pts (P=0.6) The response rate was 47% in Rhoda pos and 33% in Rhoda neg pts (p=0.6) and the CR rate 23% in Rhoda pos vs 0% in Rhoda neg pts (P=0.26).Median OS was 373 d. in P-gp pos and not reached in P-gp neg pts (p= 0.18), and 428 d in Rhoda pos pts vs 292 d. in Rhoda neg pts (P=0.07). Conclusion Contrary to what is observed with anthracycline based chemotherapy, P-gp expression and functionality did not negatively affect clinical response to AZA in this cohort of MDS and AML post MDS, with even a trend for longer OS in patient with Rhoda pos cells. This difference may further explain the possible efficacy of AZA in chemoresistant cases of MDS and AML post MDS Disclosures: Fenaux: Celgene: Honoraria, Research Funding.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2041-2041
    Abstract: Abstract 2041 INTRODUCTION: Hereditary Spherocytosis (HS) is characterized by weakened vertical linkages between the membrane skeleton and the red blood cells (RBC) lipid bilayer, leading to the release of microparticles (MPs). SDS-PAGE and ektacytometry are the two reference tests to establish the diagnosis. However, SDS-PAGE lacks sensitivity to very mild or asymptomatic HS carriers, and a high reticulocyte count might mask a reduction in ankyrin-1 in SDS-PAGE whereas an ankyrin-1 defect represents 40–65% of HS in the USA and Europe. Ektacytometry is only available in specialised laboratories. Consequently, diagnosis of HS is currently based on a combination of clinical and family histories, physical examination and laboratory data, especially RBC indices and sometimes a hypertonic cryohaemolysis test and eosin-5-maleimide binding in flow cytometry (EMA). Besides the release of MPs due to destabilisation of the lipid bilayer, one previous study showed that in HS, but not in AIHA, the surface area loss is already present at the circulating reticulocyte stage. AIMS: The aim of this study is to use the release of MPs at the reticulocyte stage to develop an algorithm on 2 haematological analysers: XE-2100 and XE-5000 (Sysmex) as simple, fast, accurate, sensitive, and specific diagnostic laboratory test for HS. METHODS: HgB, MCV, MCHC, Reticulocytes (Ret), Immature Reticulocytes Fraction (IRF), Hypochromic RBC (Hypo-He), Microcytic RBC (MicroR) and indices were determined using XE-2100 (n= 15) and XE-5000 (n= 30). %HYPO−He and %HYPER−He are specific to the XE−5000 (Figure 1). We first studied the efficiency of discordance between Ret and IRF on the XE-2100 and XE-5000, the MicroR/Hypo-He ratio on XE-5000 and the combination of those 2 rules forming an algorithm, to screen 45 confirmed HS (22 women, 23 men, mean age 13.1, from 1 day to 76 years old). They were classified as severe (n= 6, Hg 〈 8g/dl), moderate (n= 27, Hg 8–12 g/dl) and mild (n= 12, Hg 〉 12g/dl). We then studied the efficiency of the algorithm to differentiate HS from other haemolytic disorders (n=108), microcytic RBC disorders (n=93), healthy subjects (n=61) and a routine database (n=1230). RESULTS: HS is characterized by a high reticulocyte count (Ret) without an equally elevated Immature Reticulocytes Fraction (IRF) (Figure 1). The algorithm includes a precondition to screen all cases of HS, and a second rule taking into account the degree of anaemia. All 45 HS have reticulocytes 〉 80 G/l and a Ret (G/l)/IRF (%) ratio higher than 7.7. The second rule is different according to HgB level. All trait and mild cases of HS have a Ret/IRF ratio greater than 19. The efficiency of the algorithm to screen trait and mild HS is very interesting, as the diagnosis of trait and mild HS is currently difficult even by SDS-PAGE. The severity of the disease shown by HgB level is due to the intensity of release of MPs, which is reflected at best by MicroR. Thus, for moderate and severe cases, MicroR and MicroR/Hypo-He were combined. The optimal cut-offs for MicroR and MicroR/Hypo-He in cases of HgB between 8 and 12 g/dl, were 3.5% and 2.5 respectively and if HgB was lower than 8 g/dl, were 3.5% and 2.0 respectively. Interestingly, the algorithm was also valid for the 4 neonates and the 4 cases of ABO incompatibility patients. We also showed that this algorithm is much more performant than single parameters (Ret/IRF index) or the existing rules (reticulocytosis, % microcytes, combination of increased Mean Corpuscular Haemoglobin Concentration (MCHC) with increased red distribution width (RDW), combination of increased RDW with hyperdense cells (Hyperchromic cells), combination of increased MCHC with increased hyperdense cells). The Area Under the Curve (AUC), sensitivity, specificity, predictive positive value (PPV) and negative predictive value (NPV) were respectively 0.997, 100%, 99.3%, 75% and 100% for the HS algorithm. Finally, preliminary data on a small group of patients indicate that HS induced the release of significantly more RBC MPs than healthy subjects, paroxysmal nocturn hemoglobinuria and heparin-induced thrombocytopenia. CONCLUSIONS: We have developed a simple and inexpensive algorithm that could be used as an excellent screening device for the diagnosis of HS. This rapid algorithm also works on mild HS, ABO incompatibilities, neonates and overcomes the lack of sensitivity of SDS-PAGE in ankyrin deficiencies. 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: 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. 134, No. Supplement_1 ( 2019-11-13), p. 3876-3876
    Abstract: Objectives: Tisagenlecleucel (CTL019) is a chimeric antigen receptor T cell- therapy that reprograms autologous T cells to target CD19+ leukemia cells, approved in the US (2017) and in the EU (2018). This study reports the feasibility, safety and efficacy of CTL019 in patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) treated in Robert-Debré and Saint-Louis University Hospitals (Assistance Publique-Hôpitaux de Paris/Université de Paris). Methods: Patients (pts) with an apheresis performed between march 1, 2016 and june 15, 2019, included in sponsored-clinical trials or treated within the French compassionate program or with the commercial product, were analyzed. All infused pts received a fludarabine-cyclophosphamide based lymphodepletion before a single infusion of CAR-T cells (2 to 5 x 106 CTL019/kg if less than 50 kg; a fixed dose of 1 to 2.5 x 108 CTL019/kg if 〉 50 kg) Results: 55 pts were referred from 25 French centers. Forty-one pts with a median number of relapses of 2 (range, 1-5) were infused with CTL019 at a median age of 18.2 y (range, 1-29.2). Eight pts were not infused due to progressive disease (n=4), screen failure (n=3) or fatal septic shock (n=1). Six pts were waiting to be infused at time of analysis. Out of the 41 infused pts, 26 had a prior history of allogeneic SCT (63%), 11 had received blinatumomab (27%). Among the 40 pts evaluable at one month post-infusion, 38 were in CR/CRi (95%) (one progression at day 5 after infusion and one toxic death at D6), 35/38 (92.1%) being clone-specific Ig-TCR MRD negative. After 3 months 21 out of 26 evaluable pts (81%) had a negative MRD. The 5 remaining MRD positive pts did relapse. No pt underwent allogeneic HSCT while in CR after CTL019 infusion. Median event free survival (EFS) and overall survival (OS) were not reached with a median follow up of 7.2 months (range, 0.2-36.3). The 18-month OS probability was 80% (95%CI, 58%-92%). The 18-month EFS probability was 58 % (95%CI, 37%-74%). Ten pts relapsed after a median time of 3.4 months (range, 1.9-10): 3 relapses were CD19+ and 5 CD19- (4 out of these 5 pts had a preexposure to blinatumomab), 2 being of undetermined status. Loss of B-cell aplasia (BCA) occurred in 9 pts after a median time of 3 months (range, 2-12), followed by relapse for 2 pts (one concomitant with loss of BCA and one 7 months later). Three pts received a second infusion of CTL019 for loss of BCA with no further expansion of CAR-T cells. Prior treatment with blinatumomab was a significant predictive factor for relapse (HR=6.082, 95%CI, 1.2-30, p=0.0005) in a univariate analysis. There was a trend toward increased risk of relapse with increased disease burden (≥ 5%) before lymphodepletion regimen (HR=2.4, 95%CI, 0.7-8, p=0.14). Twenty-two pts experienced a CRS (≥ grade 3: n=13, all ≥ 10 y). ICU was required for 14 pts (34%). One 29 year-old pt died of an uncontrollable CRS at day 6. Ten pts received tocilizumab, 4 pts siltuximab and 9 pts corticosteroids. Age ≥ 10 y (p=0.04) and a high disease burden just before lymphodepletion (marrow blasts ≥ 5%) (p=0.01) were associated with a higher risk of CRS ≥ grade 3. Nine neurological events have been reported, being reversible except in 2 cases (one death in combination with grade 5 CRS-cf supra-, one HHV6-related encephalitis with neurological sequelae). Among the 9 pts who presented neurological events, 8 experienced CRS grade ≥ 3 (RR=17.2, 95%CI, 3.22-100.3, p=0.0001). By day 28, unresolved neutropenia grade ≥ 3 was reported for 13 pts. G-CSF treatment was required in 21 pts overall. Thrombocytopenia grade ≥3 was reported for 14 pts. Conclusion: CTL019 confirms its efficacy with a high response rate after infusion and very encouraging early outcomes in a cohort of pts heavily pretreated for refractory or multiply relapsed B-ALL. Persistent remissions with a potential for cure were observed without additional HSCT, relapses occurring within the first year after infusion of CTL019. Accurate assessment of a potentially deleterious effect of Blinatumomab preexposure notable on CD19 negative relapse will need more pts and a longer follow-up. Toxicity profile was tolerable and manageable thanks to collaboration between intensivists, neurologists and hematologists. The identification of severe CRS predictive risk factors (high disease burden just before lymphodepletion and age ≥ 10 y) points towards the reinforcement of toxicity monitoring and treatment anticipation in these cases. Disclosures Boissel: NOVARTIS: Consultancy. Baruchel:NOVARTIS: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    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. 131, No. 7 ( 2018-02-15), p. 717-732
    Abstract: Next-generation sequencing broadens the spectrum of germ line mutations in a cohort of patients with likely-inherited BMF. Salient clinical features and distinct natural histories are consistently found in SAMD9L and SAMD9, MECOM/EVI1, and ERCC6L2 disorders.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 119, No. 24 ( 2012-06-14), p. 5722-5730
    Abstract: Desensitization controls G protein–dependent signaling of chemokine receptors. We investigate the physiologic implication of this process for CXCR4 in a mouse model harboring a heterozygous mutation of the Cxcr4 gene, which engenders a desensitization-resistant receptor. Such anomaly is linked to the warts, hypogammaglobulinemia, infections, myelokathexis (WHIM) syndrome, a human rare combined immunodeficiency. Cxcr4+/mutant(1013) mice display leukocytes with enhanced responses to Cxcl12 and exhibit leukopenia as reported in patients. Treatment with CXCL12/CXCR4 antagonists transiently reverses blood anomalies, further demonstrating the causal role of the mutant receptor in the leukopenia. Strikingly, neutropenia occurs in a context of normal bone marrow architecture and granulocyte lineage maturation, indicating a minor role for Cxcr4-dependent signaling in those processes. In contrast, Cxcr4+/1013 mice show defective thymopoiesis and B-cell development, accounting for circulating lymphopenia. Concomitantly, mature T and B cells are abnormally compartmentalized in the periphery, with a reduction of primary follicles in the spleen and their absence in lymph nodes mirrored by an unfurling of the T-cell zone. These mice provide a model to decipher the role of CXCR4 desensitization in the homeostasis of B and T cells and to investigate which manifestations of patients with WHIM syndrome may be overcome by dampening the gain of CXCR4 function.
    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...