Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood Cells, Molecules, and Diseases, Elsevier BV, Vol. 87 ( 2021-03), p. 102534-
    Type of Medium: Online Resource
    ISSN: 1079-9796
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 1462186-1
    detail.hit.zdb_id: 1237083-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 3652-3652
    Abstract: Background: Hydroxyurea is the primary disease modifying therapy for patients with sickle cell anemia (SCA). The clinical and laboratory benefits of hydroxyurea are the greatest when escalated to the maximum tolerated dose (MTD). The process of dose escalation to MTD requires expertise and can be tedious, often taking 6-12 months to titrate to the optimal dose. In addition, due in part to inter-patient variability in hydroxyurea pharmacokinetics (PK), the MTD varies among patients with a range of 15-35 mg/kg/day. We utilized a population PK model in combination with Bayesian estimation and a sparse sampling strategy, to individualize dosing of children starting hydroxyurea treatment. Methods: The Therapeutic Response Evaluation and Adherence Trial (TREAT, ClinicalTrials.gov NCT02286154) is a prospective study of hydroxyurea for children with SCA. The primary objective is to develop and evaluate a population PK-based model to predict hydroxyurea MTD through an individualized dosing strategy. A sparse sampling approach was developed to allow practical sampling from young children with SCA. The sampling strategy includes administering a single oral 20 mg/kg dose followed by collection of small quantities of blood (~100uL) at three post-dosing time points (15-20 minutes, 50-60 minutes, and ~3 hours). Baseline labs (including liver and renal function) are typically collected by venipuncture, while the other two samples are drawn by fingerstick or heelstick. Plasma hydroxyurea concentrations are measured using HPLC using an internal standard of methylurea. Using the population PK model with Bayesian estimation and hydroxyurea concentrations measured at the three specified time points, hydroxyurea exposure is estimated using specialized therapeutic drug monitoring software (MWPharm, Mediware, Prague, Czech Republic). Using the area under the curve (AUC0-inf) estimated by the model, we calculate a starting dose that is predicted to achieve an AUC of 115 ug*h/mL, which was the mean AUC value at MTD for a large cohort of children from a previous study (Dong M et al. Br J Clin Pharmacol 2016). The primary objective is to select a starting dose that is close to actual MTD, to reduce the time to maximum therapeutic effect and need for dose modifications before achieving MTD. Results: From December 2014 through June 2016, 20 children taking taking hydroxyurea for the first time were enrolled in TREAT. Seventeen of the 20 participants had all 3 post-treatment PK samples collected and processed to allow calculation of an individualized PK-based dose, while 3 had difficulties in sampling or processing that prevented a safe PK-guided dose recommendation. These 3 participants were started at the standard hydroxyurea dose of 20 mg/kg/day. The Table summarizes baseline characteristics for the initial study population, notable for a very young starting age with 13/20 (65%) less than two years of age. Twelve children with PK-based initial dosing have been treated with hydroxyurea for at least six months. Despite the young starting age, after six months of hydroxyurea, children have documented increases in total Hb (1.4+/-1.9 g/dL), HbF (11.3+/-6.4%), and MCV (15+/-8 fL) and decreases in absolute reticulocyte count (-217+/-128 x 109/L) and absolute neutrophil count (-1.0+/-1.9 x 109/L). In 9 of 12 participants, the PK-guided initial dose remained the best clinical dose at six months without significant dose changes except for minor adjustments for weight. Two patients required a single dose escalation due to inadequate marrow suppression, while one required a dose hold and decrease due to neutropenia during and following a viral infection. Conclusions: These data demonstrate that a sparse sampling approach, requiring only 3 blood samples over 3 hours, is able to accurately estimate hydroxyurea exposure in children with SCA. Hydroxyurea exposure, as defined by AUC, was similar with this sparse sampling approach as previous studies that relied upon a more standard and prolonged PK sampling approach. This population PK model is then able to predict a safe starting dose of hydroxyurea that approximates the actual MTD, with clinically significant improvements in laboratory parameters following six months of therapy. This individualized PK-guided dosing regimen should simplify hydroxyurea dosing and reduce the time interval to reach MTD and maximal clinical benefits. Table Table. Disclosures Kalfa: Baxter/Baxalta/Shire: Research Funding. Quinn:Silver Lake Research Corporation: Consultancy; Amgen: Research Funding; Eli Lilly: Research Funding. Ware:Nova Laboratories: Consultancy; Addmedica: Research Funding; Global Blood Therapeutics: Consultancy; Bayer Pharmaceuticals: Consultancy; Biomedomics: Research Funding; Bristol Myers Squibb: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: The Lancet, Elsevier BV, Vol. 387, No. 10019 ( 2016-02), p. 661-670
    Type of Medium: Online Resource
    ISSN: 0140-6736
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 2067452-1
    detail.hit.zdb_id: 3306-6
    detail.hit.zdb_id: 1476593-7
    SSG: 5,21
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3-3
    Abstract: Transcranial Doppler (TCD) screening in children with sickle cell anemia (SCA) identifies abnormally elevated cerebral artery flow velocities that confer an elevated risk for primary stroke. Chronic transfusions offer effective stroke prophylaxis in this setting, but must be continued indefinitely and lead to transfusional iron overload. An alternative treatment strategy that offers similar effective protection against primary stroke, and provides control of iron overload, is needed. TCD With Transfusions Changing to Hydroxyurea (TWiTCH, NCT01425307) was an NHLBI-funded Phase III multicenter randomized clinical trial comparing 24-months of standard treatment (transfusions) to alternative treatment (hydroxyurea) in children with SCA and abnormal TCD velocities. All eligible children had received at least 12 months of transfusions. TWiTCH had a non-inferiority trial design; the primary study endpoint was the 24-month TCD velocity obtained from a linear mixed model, controlling for baseline (enrollment) values, with a non-inferiority margin of 15 cm/sec. The transfusion arm maintained children at HbS 〈 30%; an elevated liver iron concentration (LIC) identified by R2 MRI FerriScan® was managed with chelation. The hydroxyurea arm included an overlap period with transfusions until a stable maximum tolerated dose (MTD) of hydroxyurea was reached; transfusions were then replaced by serial phlebotomy to reduce iron overload. In both arms, TCD velocities were obtained every 12 weeks and reviewed centrally, with local investigators masked to the results. A centralized TCD alert algorithm monitored changes from enrollment velocities. A total of 159 children were enrolled but 38 failed screening due primarily to severe vasculopathy on brain MRA or inadequate TCD exams; 121 children were randomized (61 to transfusions, 60 to hydroxyurea) with balanced characteristics including enrollment maximum TCD velocities (145 ± 21 versus 145 ± 26 cm/sec), age, duration of transfusions, serum ferritin, and LIC. Study participants randomized to transfusions maintained an average HbS 〈 30% throughout the study, while those on hydroxyurea reached MTD after 7 ± 2 months at an average dose of 27 mg/kg/day, with expected hematological changes including HbF ~25% throughout the treatment period. After 37% of the participants exited the study, a scheduled interim analysis suggested the primary study endpoint was likely to be achieved. NHLBI allowed the study to continue until 50% of the children exited, at which time the statistical analysis was confirmed and the study was terminated; all remaining participants moved to the exit phase. The final analysis included 42 on the transfusion arm who completed all treatment, 11 with truncated treatment, and 8 withdrawn; the hydroxyurea arm included 41 who completed all treatment, 13 with truncated treatment, and 6 withdrawn. The final calculated TCD velocities (mean ± standard error) in the transfusion and hydroxyurea arms were 143 ± 1.6 and 138 ± 1.6 cm/sec, respectively; by intention-to-treat analysis, the p-value for non-inferiority = 8.82 x 10-16 and by post-hoc analysis the p-value for superiority = 0.046. Among 29 new neurological events, all centrally adjudicated by masked reviewers, there were no strokes but 6 transient ischemic attacks (3 in each arm). One child (transfusion arm) was withdrawn per the TCD alert algorithm after developing on-study TCD velocities 〉 240 cm/sec. Exit brain MRI/MRA exams documented no new parenchymal abnormalities but one child (transfusion arm) developed new vasculopathy. Sickle cell related serious adverse events were more common in the hydroxyurea arm than the transfusion arm (23 to 15), but none was related to study treatment or study procedures. Iron overload improved more in the hydroxyurea arm than in the transfusion arm, with a greater average change in serum ferritin (-1085 compared to -38 ng/mL, p 〈 0.001) and LIC (average -1.9 compared to +2.4 mg/g dry weight liver, p=0.001). In the multicenter Phase III TWiTCH trial, which treated children with SCA and abnormal TCD velocities but without severe MRA vasculopathy, hydroxyurea at MTD was non-inferior and possibly superior to chronic transfusions for maintaining TCD velocities. Serial phlebotomy effectively managed iron overload. Hydroxyurea may represent an effective alternative to indefinite transfusions for the prevention of primary stroke in this high risk population. Disclosures Ware: Eli Lilly: Other: DSMB membership; Bayer Pharmaceuticals: Consultancy; Bristol Myers Squibb: Research Funding; Biomedomics: Research Funding. Off Label Use: Hydroxyurea for children with SCA. Owen:Novartis: Speakers Bureau. Rogers:BioRad Labs: Consultancy; Apopharma: Consultancy; Baxter: Consultancy; Glaxo Smith Kline: Consultancy. Kwiatkowski:Shire Pharmaceuticals and Sideris Pharmaceuticals: Consultancy; Sideris Pharmaceuticals: Consultancy; Novartis: Research Funding; ISIS: Membership on an entity's Board of Directors or advisory committees. Heeney:Sancillio: Consultancy; Eli Lilly: Research Funding. Nottage:Janssen Pharmaceuticals: Employment. Cohen:Novartis: Consultancy; ApoPharma: Other: DSMB member.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: American Journal of Hematology, Wiley, Vol. 94, No. 8 ( 2019-08), p. 871-879
    Abstract: Hydroxyurea is FDA‐approved and now increasingly used for children with sickle cell anemia (SCA), but dosing strategies, pharmacokinetic (PK) profiles, and treatment responses for individual patients are highly variable. Typical weight‐based dosing with step‐wise escalation to maximum tolerated dose (MTD) leads to predictable laboratory and clinical benefits, but often takes 6 to 12 months to achieve. The Therapeutic Response Evaluation and Adherence Trial (TREAT, NCT02286154) was a single‐center study designed to prospectively validate a novel personalized PK‐guided hydroxyurea dosing strategy with a primary endpoint of time to MTD. Enrolled participants received a single oral 20 mg/kg dose of hydroxyurea, followed by a sparse PK sampling approach with three samples collected over three hours. Analysis of individual PK data into a population PK model generated a starting dose that targets the MTD. The TREAT cohort (n = 50) was young, starting hydroxyurea at a median age of 11 months (IQR 9‐26 months), and PK‐guided starting doses were high (27.7 ± 4.9 mg/kg/d). Time to MTD was 4.8 months (IQR 3.3‐9.3), significantly shorter than comparison studies (p  〈  0.0001), thus meeting the primary endpoint. More remarkably, the laboratory response for participants starting with a PK‐guided dose was quite robust, achieving higher hemoglobin (10.1 ± 1.3 g/dL) and HbF (33.3 ± 9.1%) levels than traditional dosing. Though higher than traditional dosing, PK‐guided doses were safe without excess hematologic toxicities. Our data suggest early initiation of hydroxyurea, using a personalized dosing strategy for children with SCA, provides laboratory and clinical response beyond what has been seen historically, with traditional weight‐based dosing.
    Type of Medium: Online Resource
    ISSN: 0361-8609 , 1096-8652
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2019
    detail.hit.zdb_id: 1492749-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 131, No. 2 ( 2021-1-19)
    Type of Medium: Online Resource
    ISSN: 0021-9738 , 1558-8238
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2021
    detail.hit.zdb_id: 2018375-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2022-01-24)
    Abstract: M-CSF receptor signaling supports the development and survival of mononuclear phagocytes and is thought to play a role in post burn anemia by promoting myeloid lineage bias. We found M-CSF secretion was increased in burn patients and a murine model of post burn ACI, so we neutralized M-CSF in ACI mice to determine if erythropoiesis was improved. Instead, M-CSF blockade further impaired erythropoiesis and erythroid cells access to iron. M-CSF blockade enhanced inflammatory cytokine secretion, further increased systemic neutrophil counts, and led to tissue iron sequestration that was dependent, in part, on augmented IL-6 secretion which induced hepcidin. Deleterious effects of post burn M-CSF blockade were associated with arrest of an iron recycling gene expression signature in the liver and spleen that included Spi-C transcription factor and heme oxygenase-1, which promote heme metabolism and confer a non-inflammatory tone in macrophages. Hepatic induction of these factors in ACI mice was consistent with a recovery of ferroportin gene expression and reflected an M-CSF dependent expansion and differentiation of Spi-C+ monocytes into Kupffer cells. Together, this data indicates M-CSF secretion supports a homeostatic iron recycling program that plays a key role in the maintenance of erythroid cells access to iron following burn injury.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2615211-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 108, No. 12 ( 2006-12-01), p. 3637-3645
    Abstract: Actin oligomers are a significant structural component of the erythrocyte cytoskeleton. Rac1 and Rac2 GTPases regulate actin structures and have multiple overlapping as well as distinct roles in hematopoietic cells; therefore, we studied their role in red blood cells (RBCs). Conditional gene targeting with a loxP-flanked Rac1 gene allowed Crerecombinase–induced deletion of Rac1 on a Rac2 null genetic background. The Rac1–/–;Rac2–/– mice developed microcytic anemia with a hemoglobin drop of about 20% and significant anisocytosis and poikilocytosis. Reticulocytes increased more than 2-fold. Rac1–/–;Rac2–/– RBCs stained with rhodamine-phalloidin demonstrated F-actin meshwork gaps and aggregates under confocal microscopy. Transmission electron microscopy of the cytoskeleton demonstrated junctional aggregates and pronounced irregularity of the hexagonal spectrin scaffold. Ektacytometry confirmed that these cytoskeletal changes in Rac1–/–;Rac2–/– erythrocytes were associated with significantly decreased cellular deformability. The composition of the cytoskeletal proteins was altered with an increased actin-to-spectrin ratio and increased phosphorylation (Ser724) of adducin, an F-actin capping protein. Actin and phosphorylated adducin of Rac1–/–;Rac2–/– erythrocytes were more easily extractable by Triton X-100, indicating weaker association to the cytoskeleton. Thus, deficiency of Rac1 and Rac2 GTPases in mice alters actin assembly in RBCs and causes microcytic anemia with reticulocytosis, implicating Rac GTPases as dynamic regulators of the erythrocyte cytoskeleton organization.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2211-2211
    Abstract: Sickle nephropathy (SN) is a common cause of morbidity and mortality in sickle cell disease (SCD), begins with hyposthenuria in childhood, and progresses to albuminuria, focal sclerosing glomerulo-sclerosis (FSGS), glomerular hypofiltration, and end stage renal disease in 30-50% adults The precise molecular mechanisms underlying SN are largely unexplored, as SN has been presumed to result from sickling associated ischemia/necrosis. Herein, we explored mechanisms of sickle renal pathologies utilizing the Berkeley sickle mouse model (HbS mice). We show that HbS mice develop renal pathologies similar to human SN, with hyposthenuria, progressive albuminuria, FSGS and nephron loss. HbS mice 4-8 weeks of age have high GFR compared to WT mice, that rapidly declines to subnormal levels by 16-24 weeks of age. We next explored the role of increased oxidant stress in mediating SN. We recently showed that sickle RBC are likely major contributors of reactive oxygen species (ROS) in SCD, and these high levels of ROS in RBC are also generated enzymatically by NADPH oxidase (George, et al Blood 2013). We now show that SCD-associated ROS initiate pathologically significant processes, including increased conversion of oxidized angiotensinogen (ANG) to angiotensin II (AT), and secondary AT receptor 1 (AT1R)-mediated generation of TGFβ1 in the HbS kidneys, which then phosphorylates Smad 2/3. We tested if activated Renin-Angiotensin-system (RAS) -AT1R-mediated TGFβ1 signaling causes albuminuria and FSGS in HbS mice. We blocked the AT1R with losartan, or its ligand AT by an angiotensin converting enzyme inhibitor, Captopril, starting at an early age (4wk) for 6-12 months. This prevented albuminuria and FSGN development in HbS mice. However, sickle hyposthenuria was worsened with losartan, and was even more severe with captopril. These data suggest that excessive AT1R signaling causes sickle glomerulopathy, and AT1R promotes urine concentrating ability; however, the captopril effect suggests that AT binds another receptor to further mediate urine concentrating ability. Increased RAS signaling is known to mediate glomerulopathy in other diseases, but its role in urine concentration has not been described. AT can also bind AT2 receptor that has been identified as a renoprotective receptor. We therefore investigated the role of AT1R and AT2R in sickle glomerulopathy and hyposthenuria by transplanting bone marrow from HbS mice into WT mice, AT1R-/- mice (HbS/AT1R-/-) and AT2R-/- mice (HbS/AT2R-/-) and followed them for 6-12 months. Bone marrow from WT mice was concurrently transplanted into WT, AT1R and AT2R deficient mice as controls. HbS/WT mice developed similar SN as in HbS mice with progressive albuminuria and hyposthenuria, the former reversible with losartan and captopril, and the latter worsened by these drugs as described above. However, HbS/AT1R mice were protected from development of albuminuria and FSGS, had reduced active TGFβ1 and PSmad-2/3, unlike HbS/WT mice, but developed significant hyposthenuria, which was worse than HbS/WT mice, and reminiscent of the effect of losartan. The HbS/AT2R mice also developed significantly worse hyposthenuria than HbS/WT mice, and were additionally not protected from albuminuria. These data suggest both AT1R and AT2R mediate urine concentrating ability, an effect blocked more effectively by captopril than losartan. AT1R signaling is known to activate NADPH oxidase to generate ROS. Indeed, mice placed on Captopril and Losartan had reduced ROS in RBC and platelets (cell types known to express AT1R) and kidneys, and consequently reduced RAS activation (significantly less oxidized ANG and AT), breaking the ROS-RAS-AT1R feedback loop. Significantly higher RBC and platelet ROS, oxidized ANG, and AT levels were also confirmed in patients with SCD as compared to their unaffected sibling controls. In summary, our data show that SN occurs from two distinct mechanisms – a) glomerulopathy that results in albuminuria, glomerulosclerosis and renal failure, which occurs primarily from increased AT1R signaling, and b) a tubulopathy, that results in inability to concentrate urine, and is worsened by AT1R signaling blockade, and AT2R signaling protects tubules against worsening hyposthenuria. Targeted therapies that block AT1R signaling but increase AT2R signaling may improve both glomerular and tubular pathologies in SCD and can now be explored. 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: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 976-976
    Abstract: Abstract 976 Introduction: Erythrocyte cytoskeleton disorders, a common cause of hereditary hemolytic anemia, consist of a genetically and phenotypically variable group of diseases that include hereditary spherocytosis (HS), elliptocytosis (HE), pyropoikilocytosis (HPP), and stomatocytosis (HSt) syndromes. The diagnosis is most commonly based on the morphology of the red blood cells (RBCs) on the blood smear and on ektacytometry and osmotic fragility studies. However, in infants and children with transfusion-dependent hemolytic anemia these studies are challenging to obtain since the patient has mostly transfused RBCs. Molecular diagnosis has not been easily attainable so far due to the number and large size of the genes involved in pathogenesis and due to the fact that each kindred has frequently a private mutation in the responsible gene(s) (Gallagher, Hematology, 2005). Methods and Results: We have developed a high-throughput assay for the diagnosis of known and discovery of new genetic mutations causing erythrocyte cytoskeleton disorders. The complete exon sequences of 24 genes encoding cytoskeleton proteins (spectrin a-chain (SPTA1), spectrin b-chain (SPTB), ankyrin 1 (ANK1), band 3 (SLC4A1), protein 4.1 (EPB41), protein 4.2 (EPB42), adducins (ADD), dematin (EPB49), tropomyosins (TPM), tropomodulins (TMOD), Rh-associated glycoprotein (RhAG), erythrocyte protein p55 (MPP1), stomatin (EPB72), Glut1-glucose transporter (SLC2A1), and K-Cl-cotransporter (SLC12A4, SLC12A6, SLC12A7) were determined using a Next Generation Sequencing technology. Analysis was performed on eleven patients diagnosed with erythrocyte cytoskeleton disorder along with a negative control. Informed consent was obtained from all subjects under an Institutional Review Board-approved protocol. The RainDance Technologies RDT1000 instrument was used for target enrichment covering the exons, 20 bases of exon/intron junctions, and 500 bases up and downstream of the 24 genes of interest. The products were then sequenced on an Illumina HiSeq2000 system. Bioinformatic analysis was performed in a blinded fashion as to the disease characteristics and inheritance mode of the patient using the GATK software package from the Broad Institute (DePristo et al, Nature Genetics, 2011). Mutations predicted to have significant impact to the corresponding protein structure and therefore likely to cause disease were identified in 9 out of the 11 patients (Table 1). Shared pathogenic mutation(s) were identified blindly in family members, e.g. siblings HS-S1 and HS-S2, or parents sHS-M, sHS-F with child sHS. Two EPB49 mutations predicted to impair gene function were identified in an infant with clinical picture of HPP, implicating dematin in HPP pathogenesis. Immunoblotting of RBC membranes from this patient demonstrated decreased dematin. Conclusions: Next generation sequencing for the genetically variable erythrocyte cytoskeleton disorders can provide a cost-effective and faster patient diagnosis compared with a gene-by-gene approach and it is a feasible diagnostic method in a transfusion-dependent child. Moreover, a precise genetic diagnosis can facilitate natural history studies to understand genotype-phenotype correlations in the erythrocyte cytoskeleton disorders and offer valuable insights into the structure-function relationship of the erythrocyte cytoskeleton proteins. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages