Genetic Investigation of the Role of GDF11 in the Treatment of β-Thalassemia and MDS

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2439-2439 ◽  
Author(s):  
Paraskevi Rea Oikonomidou ◽  
Ping La ◽  
Ritama Gupta ◽  
Vania Lo Presti ◽  
Carla Casu ◽  
...  

Abstract The current treatment of β-thalassemia only partially mitigates the phenotype of the disease, making the need for novel therapeutic agents imperative. The investigational drug Luspatercept (ACE-536) is a ligand trap that contains the modified extracellular domain of activin receptor IIB (ACVR2B) and induces red blood cell production in an erythropoietin independent fashion. ACE-536 binds with high affinity to members of the transforming growth factor (TGF) β superfamily and therefore alters activin/GDF signaling through the intracellular SMAD complex. In search of the specific ligands, recent studies in a mouse model of β thalassemia intermedia identified growth differentiation factor 11 (GDF11) as a possible target of the drug. It has been proposed that GDF11 is overexpressed in thalassemic erythroblasts and inhibits terminal erythroid maturation via SMAD complex phosphorylation. A negative role of GDF11 in erythropoiesis has been postulated also in myelodysplastic syndrome (MDS). We recapitulate, by a genetic approach, the phenotype of thalassemic and MDS mice treated with RAP-536, the murine counterpart of ACE-536. We generated and analyzed animals with GDF11 deletion in erythroid cells (Hbbth3/+ Gdf11fl/flEpoR-Cre and NUP98-HOXD13 Gdf11fl/flEpoR-Cre) and in all hematopoietic tissues (Hbbth3/+Gdf11fl/flVav-Cre and NUP98-HOXD13 Gdf11fl/flVav-Cre). We did not detect any changes in red blood cell number, reticulocyte count, hemoglobin or hematocrit levels compared to thalassemic or MDS mice in absence of the floxed gene. Focusing on thalassemic mice, administration of RAP-536 significantly improved the anemia and other hematopoietic parameters in the peripheral blood, decreased spleen size and ameliorated ineffective erythropoiesis as indicated by an increased ratio of mature to immature splenic erythroblasts analyzed by flow cytometry. Similar endpoints were seen comparing floxed and non-floxed animals treated with RAP-536. Therefore, the lack of GDF11 in erythroid or bone marrow derived cells did not prevent a response to the drug. To assess the effect of a pancellular absence of GDF11, we are currently in process of generating a model of β-thalassemia with total Gdf11 deletion. To investigate the possible effects of RAP-536, we treated erythroid cells derived from normal or thalassemic patients with the drug. Erythroid cell viability, number, differentiation and cell cycle remained unvaried. Of note, we did not detect significant expression of GDF11/Gdf11 in human and mouse erythroid cells. To investigate the effect of an exogenous source of GDF11 production on erythroid cells, we treated murine erythroleukemia (MEL) cells with recombinant GDF11. Upon treatment we observed phosphorylation of the SMAD2/3 complex by western blot. This effect was hindered by co-treatment of GDF11 with RAP-536. We further assessed the effects of RAP-536 on the mouse erythroid transcriptome, using RNA seq analysis in splenic erythroid populations. After administration of a single dose of RAP-536, thalassemic mice were euthanized. We used flow cytometry to identify possible alterations on differentiating erythroid populations in the spleen. Notably, between 60 and 72h we observed reduced numbers of basophilic and increased numbers of polychromatophilic erythroblasts. Analysis at 60h revealed that signal transducer and activator of transcription 5a (Stat5a), cyclin-dependent kinase 6 (Cdk6) and other cell cycle-related and metabolic genes were increased in the basophilic erythroid progenitors treated with RAP-536. This effect suggests that RAP-536 promotes proliferation and/or differentiation of erythroblasts. Thus, our genetic analyses suggest that lack of GDF11 may be required but not sufficient to improve erythropoiesis. Furthermore, erythroid cells do not produce but can respond to exogenous GDF11, likely synthesized by non-erythroid cells and under conditions of ineffective erythropoiesis. Even though we detected in vitro effects, these may not mimic physiological effects, as experimental conditions may not correlate with GDF11 concentrations in vivo. As additional ligands have been proposed (such as GDF8 and Activin B), our future studies will focus on the potential role of these molecules. Altogether, these results reveal a potential alternative target of action for ACE-536 and may lead to the discovery of new therapeutic molecules. Disclosures Suragani: Acceleron Pharma: Employment, Equity Ownership. Kumar:Acceleron Pharma: Employment, Equity Ownership.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3796-3796 ◽  
Author(s):  
Rajasekhar NVS Suragani ◽  
Robert Li ◽  
Dianne Sako ◽  
Asya Grinberg ◽  
R. Scott Pearsall ◽  
...  

Abstract Abstract 3796 Myelodysplastic syndromes (MDS) are a group of hematopoietic stem cell disorders characterized by peripheral blood cytopenias such as anemia, neutropenia or thrombocytopenia. Ineffective erythropoiesis due to increased proliferation and abortive maturation of precursors leads to severe anemia, the most common cytopenia observed in MDS syndromes. Despite elevated erythropoietin (EPO) and erythroid hyperplasia, MDS patients are often given recombinant EPO therapy to stimulate erythropoiesis. However, only a small proportion of patients respond to EPO therapy. Frequent blood transfusions as supportive care result in iron overloading and recently iron overloading is also linked to enhanced progression to AML. Therefore, alternative therapies are necessary to treat anemia in MDS patients. Signaling by members of the TGFβ superfamily are known regulators of erythropoiesis. We developed ACE-536, a ligand trap consisting of a modified activin receptor Type IIB extracellular domain linked to a human Fc domain. In vitro assays revealed that ACE-536 inhibits smad 2/3 ligands of the signaling pathway but not smad 1/5/8 ligands. Dose dependent studies using ACE-536 in mice, rats and monkeys revealed that ACE-536 treatment resulted in increased red blood parameters but did not affect other cell types. These data suggests that ACE-536 inhibits smad 2/3 phosphorylation modulating the expression of downstream genes involved in erythroid development pathway. BFU-E and CFU-E colony formation assays from bone marrow and spleen in mice following ACE-536 treatment revealed that ACE-536 did not affect the proliferation stages of erythropoiesis. In mice, terminal erythroid differentiation analysis by flow cytometry at 72hrs following RAP-536 (10mg/kg) treatment demonstrated decreased basophilic and increased ortho- and poly-chromatophilic erythroblasts and reticulocytes compared to VEH treatment. Cell cycle analysis of bone marrow and splenic erythroblasts counterstained with BrdU and 7-AAD after RAP-536 (10mg/kg, for 24 hours) or VEH treatment to EPO pre-treated (1500 units/kg, for 40 hours) mice (N=5/group) revealed that EPO+RAP-536 treatment resulted in significant decrease in S-phase and increase in G1/G2-phases of cell cycle compared to EPO+VEH treatment. In addition, EPO+RAP-536 treatment resulted in a greater increase in RBC parameters than either of the treatments alone. Together, these results demonstrate that ACE-536 increases red blood cell formation by promoting maturation of late stage erythroblasts. We then investigated the effect of ACE-536 on anemia in NUP98-HOXD13 (NHD13) transgenic murine model of MDS. NHD13 mice develop anemia, neutropenia and lymphopenia, with normal or hyper cellular bone marrow. A Majority of the mice die by 14 months due to severe pancytopenia or progression to acute myeloid leukemia. In this study, mice were divided into three groups based on age. Early (∼4 months old), mid (∼8 months old) and late stage (∼10 months) groups were randomized and dosed with either RAP-536 at 10 mg/kg or VEH twice per week for 6–8 weeks. NHD13 mice in each group had severe anemia characterized by reduced RBC, Hemoglobin and HCT and compared to wild-type littermates prior to treatment. Treatment of RAP-536 for 6–8 weeks significantly increased RBC parameters and reversed anemia at all stages. Peripheral blood smear analysis revealed no indication of increased leukemic progression due to RAP-536 treatment. Cell differential and flow cytometric evaluation of erythroid precursors from bone marrow demonstrated decreased erythroid precursors and hyperplasia after RAP-536 treatment compared to vehicle treated control. Our data demonstrate that RAP-536 can increase hematology parameters by enhancing maturation of terminally differentiated red blood cells. We have shown RAP-536 corrects ineffective erythropoiesis, decreases erythroid hyperplasia and normalizes myeloid: erythroid ratios without enhanced progression to AML in a murine MDS model. Therefore ACE-536 may represent a novel treatment for anemia associated with MDS, particularly in patients that are refractory to EPO therapy. ACE-536 has completed Phase I clinical trials in healthy human volunteers and Phase II study in MDS patients is planned. Disclosures: Suragani: Acceleron Pharma Inc: Employment, Equity Ownership. Li:Acceleron Pharma Inc: Employment, Equity Ownership. Sako:Acceleron Pharma Inc: Employment, Equity Ownership. Grinberg:Acceleron Pharma Inc: Employment, Equity Ownership. Pearsall:Acceleron Pharma Inc: Employment, Equity Ownership. Kumar:Acceleron Pharma Inc: Employment, Equity Ownership.


2006 ◽  
Vol 291 (4) ◽  
pp. H1679-H1685 ◽  
Author(s):  
Johnson Haynes ◽  
Boniface Obiako ◽  
Judy A. King ◽  
Raymond B. Hester ◽  
Solomon Ofori-Acquah

Activated neutrophils (ANs) increase sickle red blood cell (SRBC) retention/adhesion in the pulmonary circulation. This study investigates the role of neutrophil activation and SRBC retention/adhesion in the pulmonary circulation through a mechanism that involves increasing phosphatidylserine (PS) exposure on the external membrane surface of the SRBCs (PS-exposed). With the use of flow cytometry, double-labeling studies were performed with a calcium-dependent phospholipid-binding protein, annexin V-fluorescein isothiocyanate fluorescence, and the erythroid-specific marker glycophorin A to assess for the percentage of PS-exposed normal and SRBCs at baseline and after coincubation with ANs. Additional studies were performed that assessed retention/adhesion of SRBCs in the isolated rat lung using51Cr-labeled SRBC alone, SRBC + AN, SRBC + AN + zileuton, and SRBC + AN + annexin V. Specific activities of lung and perfusate were measured, and the number of retained SRBCs per gram lung was calculated. Flow cytometry demonstrated that ANs increased the percentage of PS-exposed normal and SRBCs. The 5-lipoxygenase inhibitor zileuton attenuated AN-mediated increases in PS-exposed SRBCs and decreased SRBC retention/adherence in the lung on histological sections. Similarly, in the isolated perfused lung and in histological lung sections, retention/adherence of SRBCs cloaked with annexin V was attenuated in the presence of ANs. We conclude that ANs enhance the adhesion of SRBCs to vascular endothelium by increasing red blood cell membrane externalization of PS. Zileuton attenuation of AN-mediated SRBC PS externalization suggests that a 5-lipoxygenase product(s), secreted by the AN, plays a vital role in altering the adhesive properties of PS-exposed SRBCs to vascular endothelium.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1366-1366 ◽  
Author(s):  
Watkins M Krystal ◽  
Russell Walker ◽  
Nathan Fishkin ◽  
Charlene Audette ◽  
Yelena Kovtun ◽  
...  

Abstract IMGN779 is a CD33-targeting ADC consisting of a humanized anti-CD33 antibody, Z4681A, conjugated to DGN462, a novel DNA-alkylating agent, through a cleavable disulfide linker, sulfo-SPDB. CD33 is broadly expressed on leukemic blasts of patients with AML, making it a promising target for AML therapy. DGN462 is a member of the novel IGN class of DNA-acting cytotoxic agents, that consists of an indolino-benzodiazepine dimer containing a mono-imine moiety. Potent killing of AML tumor cells by DGN462 and IMGN779 has previously been demonstrated in vitro (K. Whiteman et. al, ASH 2014 #2321). Here we describe studies elucidating the mechanism of action of this novel payload in the AML cell lines: MV4-11, HL60, and EOL-1. The ability of DGN462 to alkylate DNA was demonstrated in an AML cell line by isolation and analysis of the genomic DNA from cells. DGN462 was found to co-purify with DNA, demonstrating that it covalently reacts with cellular DNA. A comet assay was performed confirming that DGN462 does not cross-link DNA. H2AX is known to be phosphorylated on serine 139 in response to DNA damage. Using flow cytometry, we assessed the phosphorylation of H2AX in AML cell lines following exposure to DGN462 and IMGN779. Increases in phosphorylated H2AX levels were detected as early as 4 hours following exposure to DGN462-SMe and 12 hours following exposure to IMGN779. Additional flow cytometry analysis showed that exposure to DGN462-SMe and IMGN779 leads to S-phase accumulation, G2/M arrest, followed by induction of apoptotic markers (cleaved PARP and cleaved Caspase-3) at later time points. Ex vivo studies using AML patient samples also showed elevation of phosphorylated H2AX and an increase in apoptosis in myeloid blasts following exposure to DGN462-SMe and IMGN779. As a demonstration of proof-of-mechanism in AML disease models, our results suggest that cell killing by IMGN779 is mediated by DNA damage, as a consequence of DNA alkylation. The DNA damage response is accompanied by cell cycle arrest, which leads to apoptosis. This relationship will be further explored in additional AML preclinical models to support the use of phosphorylated H2AX as a pharmacodynamic biomarker for IMGN779 activity in future clinical studies. Disclosures Krystal: ImmunoGen, Inc.: Employment, Equity Ownership. Walker:ImmunoGen, Inc.: Employment, Equity Ownership. Fishkin:ImmunoGen, Inc.: Employment, Equity Ownership. Audette:ImmunoGen, Inc.: Employment, Equity Ownership. Kovtun:ImmunoGen, Inc.: Employment, Equity Ownership. Romanelli:ImmunoGen, Inc.: Employment, Equity Ownership.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3812-3812
Author(s):  
Yesim Aydinok ◽  
Antonio Piga ◽  
Raffaella Origa ◽  
Nina Mufti ◽  
Anna Erikson ◽  
...  

Abstract Introduction: Transfusion dependent thalassemia (TDT) requires regular transfusion of red cell concentrates (RBCC) to prevent the complications of anemia and excessive erythroid expansion. Despite donor testing, long-term transfusion has a substantial cumulative risk of transfusion-transmitted infection (TTI) due to undetected viruses, bacteria, and protozoa. Splenectomized (-S) TDT patients may have greater TTI morbidity than patients with spleens (+S); but may benefit from reduced use of red blood cell concentrates (RBCC) and reduced transfusion iron (Fe) loading. Pathogen reduction (PR) of RBCC with amustaline-glutathione (A-GSH) offers potential to reduce the risk of TTI. Objectives: To determine, the impact of PR-RBCC on hemoglobin (Hb) use, transfused Fe burden, incidence of RBC antibodies, and safety in -S and +S TDT patients. Methods: TDT patients at 3 sites, not stratified by spleen status, were prospectively enrolled in a two- period cross-over study randomized by sequence for RBCC preparation. Leukocyte reduced PR-RBCC (Test) were treated with 0.2mM amustaline and 20 mM GSH, re-suspended in saline-adenine-glucose mannitol (SAGM), and stored up to 35 days at 4°C. Leukocyte reduced conventional RBCC (Control) were suspended in SAGM and stored for up to 35 days at 4°C. Patients received 6 transfusions in each treatment sequence of Test or Control RBCC over ~ 5 months. Clinicians, blinded to RBCC Hb content and treatment sequence, ordered RBCC to maintain targeted pre-transfusion Hb thresholds of ~ 9-10 g/dL. Transfusion intervals or number of RBCC transfused were adjusted for clinical management. The primary efficacy outcome was assessed by non-inferiority (NI) analysis for Hb use (expressed as g/kg body weight/ day) using a pre-specified NI margin (≤ 15% of the observed Control mean). Results : Overall, mean (SD) Hb content (g) of 1024 Test RBCC = 54.6±5.9 (range: 39-73) and of 1008 Control RBCC = 55.6 ± 5.9 (range: 35-74) and varied widely. By intent-to-treat (ITT), 80 patients (40 +S and 40 -S) were transfused. For ITT patients (Table), the baseline Hb level (BL-Hb, g/dL) at first transfusion of Control periods was significantly lower than at Test periods; but the mean number of RBCC transfused, RBCC storage days, total Hb dose (g), and transfusion intervals were not significantly different for Test and Control. ITT analysis for all transfusion episodes showed Hb use for Test RBCC (0.110 g/kg/d) was not different from Control RBCC (0.112 g/kg/day). Non-inferiority was demonstrated (T-C = - 0.002 g/kg/d: 95% CI: -0.005, 0.001). ITT Test patients received a slightly lower mean total Hb dose (- 14g), and mean pre-transfusion Hb levels declined after 6 transfusions (9.4 to 8.8 g/dL). -S patients had lower BL-Hb levels (g/dL) than S+ patients in Test (9.2 vs 9.7) and Control (8.8 vs 9.2) periods (Table). -S patients received a lower mean total Hb dose of Test than Control RBCC (p=0.019); and had a decline in mean pre-transfusion Hb levels during Test periods (from 9.2 to 8.7 g/dL). Transfusion intervals were significantly longer for -S patients than +S patients with both Test and Control RBCC (p< 0.001 by 2-sample t test, respectively); and -S patients had lower Hb use than +S patients. However, Hb use of Test and Control RBCC was comparable within -S and + S cohorts (Table). Transfused Fe was less for -S patients for Test and Control RBCC. During 6 Test and 3 Control treatment periods, 8 patients (6 -S, 2 +S) had worsening anemia with pre-transfusion Hb levels (6.0-7.8 g/dL) substantially below the targeted transfusion threshold, but without evidence of hemolysis. Each of these patients received one or more Hb doses below the average RBCC transfusion episode dose (Test: 114.5 g) or (Control: 116.7 g); and 3 patients had concurrent infections. None of 80 patients had evidence of increased RBC clearance, developed antibodies to PR-RBCC, or had treatment emergent RBC alloantibodies in either treatment period. There were no differences in the overall safety profiles for Test and Control RBCC. Conclusions: Amustaline-GSH PR treatment of RBCCs offers the potential to reduce TTI risk without impacting Hb use or Fe burden in TDT. However, Hb content of Test and Control RBCC varies widely and may contribute to unexpected changes in pre-transfusion Hb levels. Spleen status affected Hb use comparably for PR-RBCC and Control RBCC, and remains an important factor in assessing transfusion requirements and Fe loading. Table. Table. Disclosures Aydinok: TERUMO: Research Funding; Cerus: Honoraria, Research Funding; CRISPR Tech: Other: DMC; Protagonist: Other: SSC; La Jolla Pharmaceuticals: Research Funding; Celgene: Research Funding; Novartis: Research Funding, Speakers Bureau. Piga:Apopharma: Honoraria, Research Funding; Celgene Corp: Membership on an entity's Board of Directors or advisory committees, Research Funding; La Jolla: Membership on an entity's Board of Directors or advisory committees, Research Funding; Bluebird Bio: Honoraria; Acceleron: Research Funding; Novartis: Research Funding. Origa:Novartis: Honoraria; Bluebird Bio: Consultancy; Cerus Corporation: Research Funding; Apopharma: Honoraria. Mufti:Cerus Corporation: Employment, Equity Ownership. Erikson:Cerus Corporation: Employment, Equity Ownership. North:Cerus Corporation: Employment, Equity Ownership. Waldhaus:Cerus Corporation: Employment, Equity Ownership. Ernst:Cerus Corporation: Employment, Equity Ownership. Lin:Cerus Corporation: Employment, Equity Ownership. Huang:Cerus Corporation: Employment, Equity Ownership. Benjamin:Cerus Corporation: Employment, Equity Ownership. Corash:Cerus Corporation: Employment, Equity Ownership.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3108-3108
Author(s):  
Hugo Germain ◽  
Sihem Tarfi ◽  
Nicolas Freynet ◽  
Andrea Toma ◽  
Bouchra Badaoui ◽  
...  

Abstract Introduction: The IMDSFlow working group reported recently the usefulness of immunophenotypic analysis of erythroid dysplasia in myelodysplastic syndromes (MDS) (Westers, 2017). This multicenter study revealed that analysis of CD36 and CD71 expression on nucleated erythroid cells as coefficient of variation (CV), in combination with CD71 fluorescence intensity and the percentage of CD117+ erythroid progenitors provided the best discrimination between MDS and non-clonal cytopenias. These four parameters were analyzed on bone marrow samples after red blood cell lysis (RBC) procedure. Since this latter could also remove some nucleated erythroid cells, it was proposed to use a nuclear dye in a whole no-lysis bone marrow strategy for analysis of CD36 and CD71 expression (Mathis, 2013). So far, no study has compared the four ELN dyserythropoiesis parameters after or without lysis procedure. Objective:We aimed to evaluate whether no-lysis procedure can lead to a better assessment of dyserythropoiesis by flow cytometry compared to lysis procedure by preserving erythroblast cells. Methods: One hundred patients referred to our laboratory for bone marrow investigations between November 2017 and June 2018 were included in this prospective study. Nineteen patients were used as control samples without cytopenia whereas 81 presented at least one cytopenia. Complete blood count parameters as well as morphological and cytogenetic analyses were used to classify these patients as 25 MDS or 56 non-MDS, referred thereafter as pathological controls. Bone marrow specimens were collected on EDTA and were processed within 4h following aspiration. A similar amount of each sample was stained in parallel with the same antibody panel (CD36, CD71, CD117 and CD45), yet according to two different protocols, one with CyTrak orange without lysis procedure and one with RBC lysis using VersaLyse (Beckman-Coulter). Data were acquired using a Navios cytometer and analyzed using the Kaluza software (BC). Geometrical means of fluorescence (GMFI) of CD71 as well as CD36 and CD71 coefficients of variation and the percentage of CD117+ erythroid progenitors were collected in order to calculate the ELN dyserythropoiesis score as previously described (see Figure 1 A-B for erythroblast selection). Results: Firstly, we compared the percentages of erythroblasts obtained with the no-lysis and the RBC lysis strategies as reported to the total nucleated cells analyzed (corresponding to the CD45 positive cells in addition to erythroblasts selected as CD71 and CD36 positive cells). Surprisingly, we found that the lysis protocol led to a higher percentage of erythroblasts than the no-lysis protocol (19.0±11.8% vs 15.4±10.8%; p<0.0001), yet both flow cytometry approaches gave lower percentages than morphology (30.4±12.5%; p<0.0001) as expected. Samples stained according to the two different protocols were analyzed with the same calibrated cytometer and the same settings for all markers (especially CD71 and CD36), allowing comparison of the raw data. The four parameters of the ELN score were significantly different between the two protocols for the 100 patients analyzed: both CV of CD36 and CD71 and % CD117 progenitors were higher with no-lysis protocol while GMFI CD71 was lower (Figure 1 C-F). We then calculated the ELN score with the recommended weighted manner: 4 points for increase in CD36 CV, 3 points for increase in CD71 CV, 2 points for decreased CD71 MFI and 2 points in the case of decreased or increased percentage of CD117+ erythroid cells. For each of the four parameters, cut-offs for the identification of MDS-associated aberrancies were defined against the 10th and/or 90th percentiles of the values obtained either from control samples (Figure 1 G) or from pathological controls (Figure 1 H) for both lysis and no-lysis protocols. With cut-offs defined against control samples, both specificity and sensitivity were better for lysis vs no-lysis protocol (93.3% vs 88.0% and 20.0% vs 12.0%, respectively). With cut-offs defined against pathological controls, again both specificity and sensitivity were higher for lysis vs no-lysis protocol (94.7% vs 88.0% and 20.0% vs 8.0%, respectively). Conclusion: Our results demonstrate that the lysis protocol leads to better specificity and sensitivity of the ELN dyserythropoiesis score than the no-lysis protocol with cut-offs defined against either control samples or pathological controls. Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2109-2109
Author(s):  
Shuo Wang ◽  
Huihui Li ◽  
Lisa Sarran ◽  
Gordana Olbina ◽  
Mark Westerman ◽  
...  

Abstract Abstract 2109 Hemoglobin (Hb) below 12.5g/dL accounts for more than 40% of all blood donor deferrals, and no standardized message is provided to these deferred donors. Furthermore, many donors who meet Hb standards are iron depleted. In light of this, the FDA requested comments on possibly changing the minimal acceptable Hb and/or the inter-donation interval. Here we explore several parameters of iron utilization after red blood cell (RBC) donation to understand its relationship to erythropoiesis. Hepcidin is the central regulator of iron absorption, suppressed by low iron stores. We hypothesize that hepcidin values would identify donors with normal Hb levels and low iron stores and thus identify those who would benefit from iron replacement to prevent future low Hb deferral. We enrolled eligible repeat male double RBC donors randomized to receive placebo (n=5) or iron supplementation (n=5) using 90 mg of elemental iron (feosol caplets). Donors were enrolled on day of donation and were invited to return at 2–4 day intervals during the 14 days following donation to compare with pre-donation markers of erythropoiesis and iron metabolism. In addition, buffy coats were collected on day 0 and again 7–10 days following donation (n=6) to quantify by flow cytometry circulating CD34+ cells as well as numbers of common myeloid progenitors (CMPs) and megakaryocyte erythroid progenitors (MEPs) in study donors. Comparing iron supplemented to placebo receiving volunteers, ferritin was higher at 14 days in iron supplemented donors (18.6±3.9 vs 12.0±1.6 ng/mL) and corresponds with 52% vs. 36% of levels prior to donation relative to placebo (P=0.05). Furthermore, serum hepcidin nadirs earlier and rebounds sooner relative to placebo (15.4±3.2 vs. 6.8±2.1 ng/mL, P=0.05). Serum iron (150±37.1 vs. 60±5.8 mg/dL, P=0.04) and transferrin saturation (39.8±9.8 vs. 16.2±2.1%, P=0.05) are also increased relative to placebo by day 10 following donation. Although we found no difference in recovery of RBC count, hemoglobin, or hematocrit following donation, a trend toward a more sustained elevation in reticulocyte count is present relative to placebo (158±12.1 vs. 118±17.3 × 109 cells/mL, P=0.07). CD34+ cells in circulation are increased following donation in all donors. Furthermore, only donors with baseline ferritin levels >20 ng/mL increased MEPs following donation. These later findings are consistent with expectations of iron regulatory mechanisms influencing erythropoiesis. Taken together, these data suggest that a complex interplay between iron regulatory and erythropoietic mechanisms occurs during recovery from blood donation. Although it is currently unclear how iron regulation affects erythropoiesis, a larger (double) sample size will aid in the generation of criteria for donor iron supplementation. Disclosures: Olbina: Intrinsic LifeSciences LLC: Employment, Equity Ownership; Hepcidin C-ELISA:. Westerman:Intrinsic LifeSciences LLC: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Hepcidin C-ELISA: Patents & Royalties; Centocor-Ortho R&D, Inc.: Honoraria.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 482-482
Author(s):  
David B. Stagg ◽  
Sara Gardenghi ◽  
Stefano Rivella ◽  
Nancy C. Andrews ◽  
Karin E. Finberg

Abstract Abstract 482 β-thalassemia is a disorder of ineffective erythropoiesis in which oxidative damage caused by unpaired α-globin chains leads to erythroid apoptosis, increased proliferation of erythroid precursors, and impaired erythroid differentiation. Patients develop systemic iron overload that is caused by red blood cell transfusions and by insufficient inhibition of gastrointestinal iron absorption by the iron regulatory hormone hepcidin. Previously we reported that homozygous genetic loss of Tmprss6, a hepatic transmembrane serine protease that inhibits hepcidin expression by the liver, led to hepcidin elevation and systemic iron deficiency in Hbbth3/+ mice, a model of β-thalassemia intermedia. Interestingly, we also found that while maintaining similar hemoglobin levels, Hbbth3/+mice with homozygous loss of Tmprss6 showed a significant reduction in splenomegaly and marked improvement in peripheral red blood cell (RBC) morphology. Here, we investigated the effects of genetic loss of Tmprss6 on erythropoiesis in Hbbth3/+ mice. In mice of different Tmprss6-Hbb genotypes, we used flow cytometry to quantify the proportion of total bone marrow cells of the erythroid lineage by measuring expression of TER119, an antigen expressed from the pro-erythroblast through the mature erythrocyte stage. Additionally, within the TER119+ population, we quantified the different erythroblast subpopulations by analyzing the intensity of forward scatter and CD44 expression. Compared to wild type (Tmprss6+/+Hbb+/+) controls, Hbbth3/+ mice with 2 wild-type Tmprss6 alleles (Tmprss6+/+Hbbth3/+) showed a significant increase in the proportion of total erythroid cells in the bone marrow, significant increases in the proportion of immature erythroid precursors (basophilic and polychromatic erythroblasts) within the erythroid population, and a significant decrease in the proportion of mature RBCs, resulting in a marrow profile consistent with ineffective erythropoiesis. In Hbbth3/+ mice with homozygous Tmprss6 disruption (Tmprss6−/−Hbbth3/+), the proportion of immature erythroid precursors (basophilic and polychromatic erythroblasts) within the erythroid population remained significantly elevated; however, the proportion of total erythroid cells in the bone marrow was no longer increased. Compared to Tmprss6+/+Hbbth3/+ mice, Tmprss6−/−Hbbth3/+ mice showed a significant increase in the proportion of mature RBCs; this was accompanied by a reduction in reactive oxygen species (ROS) production (as assessed by the indicator CM-H2DCFDA) and apoptotic cells (as assessed by annexin V binding) within both the orthochromatic erythroblast/reticulocyte and mature red cell subpopulations. Additionally, compared to Tmprss6+/+Hbbth3/+ mice, Tmprss6−/−Hbbth3/+ mice showed a marked reduction in α-globin precipitates in membrane fractions prepared from peripheral RBCs. Interestingly, when normalized to α-globin mRNA expression, bone marrow mRNA encoding α-hemoglobin stabilizing protein (AHSP), an α-globin chaperone, was significantly higher in Tmprss6−/−Hbbth3/+ mice compared to Tmprss6+/+Hbbth3/+ mice, compatible with the known stabilization of AHSP mRNA under low iron conditions. Together, these findings suggest a model in which genetic loss of Tmprss6 in Hbbth3/+ mice leads to a systemically iron-deficient state in which reduced iron availability to erythroid precursors leads to stabilization of free α-globin chains, a reduction in both ROS formation and erythroid apoptosis, and ultimately more effective erythropoiesis. In the context of previous findings, these results indicate that hepcidin-elevating strategies based on pharmacological inhibition of Tmprss6 might alter the clinical phenotype of β-thalassemia not only by reducing systemic iron loading but also by altering erythroid maturation. Disclosures: Rivella: Novartis Pharmaceuticals: Consultancy; Biomarin: Consultancy; Merganser Biotech: Consultancy, Equity Ownership, Research Funding; Isis Pharma: Consultancy, Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2856-2856
Author(s):  
Wolfgang Kern ◽  
Manja Meggendorfer ◽  
Seishi Ogawa ◽  
Claudia Haferlach ◽  
Susanne Schnittger ◽  
...  

Abstract Introduction: The diagnostic approach for suspected myelodysplastic syndromes (MDS) is evolving and flow cytometry and molecular genetics are increasingly considered to be applied in addition to cytomorphology and cytogenetics. While reports on comparisons of flow cytometric findings with results of cytomorphology and cytogenetics are available, data on comparisons between results obtained by flow cytometry and molecular genetics, however, have not yet been presented in detail. Aims: 1) To assess the correlation between flow cytometric findings on MDS-specific aberrant antigen expression and the presence of molecular mutations in patients with cytomorphologically proven MDS. 2) To determine the respective impact of flow cytometric findings and of molecular mutations on survival in patients with MDS. Patients and methods: In 256 patients (male/female, 161/95; median age 72 years, range 24-90) with proven MDS (137 low-risk MDS, 119 RAEB1/2) we compared data on aberrantly expressed antigens (AEA) determined according to ELN guidelines (Westers, Leukemia 2012) to the previously published mutational status of 104 genes (Haferlach, Leukemia 2014). Results: Median numbers (ranges) of AEA were 0 (0-3) in myeloid progenitors, 2 (0-4) in granulocytes, 1 (0-5) in monocytes and 0 (0-1) in erythroid cells. Median number of mutation was 2 (0-7). The number of AEA in myeloid progenitors, granulocytes and monocytes increased with increasing number of mutations (r=0.257, p<0.001). Accordingly, in cases with ≥3 mutations the number of AEA in myeloid progenitors, granulocytes and monocytes was higher than in cases with ≤2 mutations (mean±SD, 3.9±1.9 vs. 3.0±2.0, p=0.001). This correlation was significant also when considering granulocytes as a single cell compartment (r=0.308, p<0.001) but non-significant trends only for myeloid progenitors and monocytes. No such correlation was observed for erythroid cells. Specifically, mutations in each of the genes TET2, ASXL1, SRSF2, STAG2, ZRSR2 or NF1 were associated with significantly higher numbers of AEA in ≥1 cell compartment. Cases with mutations in ≥1 of these genes (n=145), as compared to those without these 6 mutations (n=111), had higher numbers of AEA in myeloid progenitors (0.4±0.7 vs. 0.2±0.5, p=0.037), granulocytes (2.0±1.1 vs. 1.4±1.1, p<0.001) and monocytes (1.5±1.3 vs. 1.0±1.0, p=0.002). Consequently, the difference in the total of AEA was even larger (3.9±2.0 vs. 2.7±1.9, p<0.001). Regarding scoring points according to IPSS-R, there was a significant correlation with the number of AEA in granulocytes (r=0.189, p=0.004) as well as with the number of AEA in monocytes (r=0.159, p=0.017). Consequently, there was also a significant correlation between the IPSS-R scoring points and the number of AEA in myeloid progenitors, granulocytes and monocytes (r=0.227, p=0.001). Overall survival was impacted by the presence of mutations in ≥1 of the genes TP53, EZH2, ETV6, RUNX1 and ASXL1 (p<0.001, HR 2.9) published by Bejar (NEJM 2011) as well as by the presence of ≥3 AEA in myeloid progenitors, granulocytes and monocytes (p=0.015, HR 1.7) and by IPSS-R (p<0.001, HR 1.4). Multivariate analysis considering mutations and AEA revealed an independent significance for both of them (mutations, p<0.001, HR 2.9; AEA, p=0.017, HR 1.7). However, inclusion of also IPSS-R as a covariate resulted in a trend only for AEA (p=0.16, HR 1.4) and independent significance for mutations (p<0.001, HR 2.3) and IPSS-R (p<0.001, HR 1.3). Conclusions: This data demonstrates that the degree of flow cytometric findings on MDS-related aberrant antigen expression correlates with the number of molecular mutations as well as with the IPSS-R. The present result therefore further support the consideration of both flow cytometry and molecular genetics for the diagnostic work-up of MDS in an integrated approach in combination with cytomorphology and cytogenetics. Disclosures Kern: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Meggendorfer:MLL Munich Leukemia Laboratory: Employment. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4548-4548 ◽  
Author(s):  
Ann Barbier ◽  
Bruce Silver ◽  
Elizabeth Merica ◽  
Marvin Cohen ◽  
Charles Kung ◽  
...  

Abstract INTRODUCTION: Pyruvate kinase (PK) deficiency is a congenital hemolytic anemia caused by deficient function of the PK enzyme. This condition is currently managed with supportive care, including transfusions, folate supplementation, splenectomy, and reducing iron overload, but there are no treatments specifically targeting the underlying metabolic defect. AG-348 is a novel, orally available, small molecule activator of wild-type and mutant red blood cell PK. METHODS: Results of single-ascending dose (SAD) and multiple-ascending dose (MAD) studies in healthy adult volunteers (Yang H et al. Blood 2014;124[21]:4007; Yang H et al. Haematologica 2015;100[s1]:Abs S138) indicated good safety and tolerability and predictable pharmacokinetics at doses that demonstrated significant pharmacodynamic responses of increased whole blood adenosine triphosphate (ATP) and decreased 2,3-diphosphoglycerate (2,3-DPG) levels. In pre-clinical studies AG-348 inhibited the enzyme aromatase, and sex hormone profiles in the MAD study showed signs of aromatase inhibition. RESULTS: DRIVE PK is a multicenter, international, randomized, open-label, two-arm dose ranging trial of AG-348 in adults with PK deficiency. DRIVE PK is currently open for enrollment. Eligible patients are randomized to either low dose (50 mg BID) or high dose (300 mg BID) arms and receive treatment for 6 months, with the option of extended treatment. Patients are stratified by genotype to allow for genotype-phenotype correlations. A third dose may be added based on the observations made in the two original arms. The primary objective is to evaluate the safety and tolerability of AG-348. Secondary objectives include evaluation of pharmacokinetics and pharmacodynamics, as measured by AG-348, ATP and 2,3-DPG levels. A range of biochemical markers of clinical benefit will be assessed, including hemoglobin levels, reticulocyte count, bilirubin, erythropoietin, ferritin, transferrin saturation, and haptoglobin. PK activity, glycolytic flux assay and PK protein levels in blood will be analyzed as exploratory endpoints. The trial population consists of adult patients with PK deficiency with hemoglobin levels at screening of ≤12.0 g/dL (men) or ≤11.0 g/dL (women), who have received no more than three units of red blood cells in the 12 months preceding the first dose of AG-348, and no transfusion within 4 months. Full inclusion/exclusion criteria can be found on www.clinicaltrials.gov, NCT02476916. CONCLUSION: DRIVE PK, an ongoing phase 2 trial of the PK activator, AG-348, is the first interventional trial to target the underlying metabolic defect in patients with PK deficiency. This flexible trial with two dose arms, and potential for a third dose arm, is designed to evaluate safety and tolerability, pharmacokinetic and pharmacodynamic responses, as well as hematological markers of red blood cell metabolism. Disclosures Barbier: Agios: Employment, Equity Ownership. Silver:Agios: Consultancy. Merica:Agios Pharmaceuticals: Employment, Equity Ownership. Cohen:Agios: Consultancy. Kung:Agios: Employment, Equity Ownership. Yang:Agios Pharmaceuticals: Employment, Equity Ownership. Grace:Agios: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Agresta:Agios: Employment, Equity Ownership.


2017 ◽  
Vol 82 (6) ◽  
pp. 894-896
Author(s):  
Jean L Raphael ◽  

Sign in / Sign up

Export Citation Format

Share Document