54 results on '"Ware RE"'
Search Results
2. Hydroxyurea reduces infections in children with sickle cell anemia in Uganda.
- Author
-
Namazzi R, Bond C, Conroy AL, Datta D, Tagoola A, Goings MJ, Jang JH, Ware RE, Opoka R, and John CC
- Subjects
- Child, Humans, Hydroxyurea therapeutic use, Antisickling Agents therapeutic use, Uganda epidemiology, Anemia, Sickle Cell complications, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell epidemiology, Malaria complications, Malaria drug therapy, Malaria epidemiology
- Abstract
Abstract: After starting hydroxyurea treatment, Ugandan children with sickle cell anemia had 60% fewer severe or invasive infections, including malaria, bacteremia, respiratory tract infections, and gastroenteritis, than before starting hydroxyurea treatment (incidence rate ratio, 0.40 [95% confidence interval, 0.29-0.54]; P < .001)., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Hydroxyurea treatment is associated with lower malaria incidence in children with sickle cell anemia in sub-Saharan Africa.
- Author
-
Olupot-Olupot P, Tomlinson G, Williams TN, Tshilolo L, Santos B, Smart LR, McElhinney K, Howard TA, Aygun B, Stuber SE, Lane A, Latham TS, and Ware RE
- Subjects
- Humans, Child, Hydroxyurea adverse effects, Incidence, Splenomegaly epidemiology, Splenomegaly drug therapy, Africa South of the Sahara epidemiology, Anemia, Sickle Cell complications, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell epidemiology, Malaria drug therapy, Malaria epidemiology, Malaria prevention & control
- Abstract
Realizing Effectiveness Across Continents with Hydroxyurea (REACH, NCT01966731) provides hydroxyurea at maximum tolerated dose (MTD) for children with sickle cell anemia (SCA) in sub-Saharan Africa. Beyond reducing SCA-related clinical events, documented treatment benefits include ∼50% reduction in malaria incidence. To identify associations and propose mechanisms by which hydroxyurea could be associated with lower malaria rates, infections were recorded across all clinical sites (Angola, Democratic Republic of Congo, Kenya, and Uganda). Hazard ratios (HR) with 95% confidence intervals (CIs) for baseline demographics, and time-varying laboratory and clinical parameters were estimated in a modified Cox gap-time model for repeated events. Over 3387 patient-years of hydroxyurea treatment, 717 clinical malaria episodes occurred in 336 of 606 study participants; over half were confirmed by blood smear and/or rapid diagnostic testing with 97.8% Plasmodium falciparum. In univariate analysis limited to 4 confirmed infections per child, malaria risk was significantly associated with absolute neutrophil count (ANC), splenomegaly, hemoglobin, and achieving MTD; age, malaria season, MTD dose, fetal hemoglobin, α-thalassemia, and glucose-6-phosphate dehydrogenase deficiency had no effect. In multivariable regression of confirmed infections, ANC was significant (HR, 1.37 per doubled value; 95% CI, 1.10-1.70; P = .0052), and ANC values <3.0 × 109/L were associated with lower malaria incidence. Compared with nonpalpable spleen, 1- to 4-cm splenomegaly also was associated with higher malaria risk (HR, 2.01; 95% CI, 1.41-2.85; P = .0001). Hydroxyurea at MTD is associated with lower malaria incidence in SCA through incompletely defined mechanisms, but treatment-associated mild myelosuppression with ANC <3.0 × 109/L is salutary. Splenomegaly is an unexplained risk factor for malaria infections among children with SCA in Africa., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
4. Reproductive equity: preserve the reserve.
- Author
-
Quinn CT and Ware RE
- Subjects
- Female, Humans, Ovary, Reproduction
- Published
- 2022
- Full Text
- View/download PDF
5. Increased oxygen affinity: to have and to hold.
- Author
-
Quinn CT and Ware RE
- Subjects
- Oxygen
- Published
- 2021
- Full Text
- View/download PDF
6. Microscope diagnosis of MYH9-related thrombocytopenia.
- Author
-
Sadaf A and Ware RE
- Subjects
- Adolescent, Female, Humans, Menorrhagia complications, Menorrhagia genetics, Menorrhagia pathology, Mutation, Missense, Thrombocytopenia complications, Thrombocytopenia pathology, Myosin Heavy Chains genetics, Thrombocytopenia genetics
- Published
- 2021
- Full Text
- View/download PDF
7. There's safety in numbers.
- Author
-
Ware RE and Dertinger SD
- Subjects
- Humans, Anemia, Sickle Cell, Hydroxyurea
- Published
- 2021
- Full Text
- View/download PDF
8. Your tired, your poor, your huddled masses.
- Author
-
Ware RE and Thornburg CD
- Subjects
- Emergency Service, Hospital, Fatigue, Humans, Anemia, Sickle Cell, Refugees
- Abstract
Competing Interests: Conflict-of-interest disclosure: The authors declare no competing financial interests.
- Published
- 2019
- Full Text
- View/download PDF
9. Novel use Of Hydroxyurea in an African Region with Malaria (NOHARM): a trial for children with sickle cell anemia.
- Author
-
Opoka RO, Ndugwa CM, Latham TS, Lane A, Hume HA, Kasirye P, Hodges JS, Ware RE, and John CC
- Subjects
- Anemia, Sickle Cell blood, Antisickling Agents therapeutic use, Blood Cell Count, Child, Preschool, Double-Blind Method, Endemic Diseases, Female, Fetal Hemoglobin metabolism, Humans, Incidence, Infant, Male, Prospective Studies, Treatment Outcome, Uganda epidemiology, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell epidemiology, Hydroxyurea therapeutic use, Malaria epidemiology
- Abstract
Hydroxyurea treatment is recommended for children with sickle cell anemia (SCA) living in high-resource malaria-free regions, but its safety and efficacy in malaria-endemic sub-Saharan Africa, where the greatest sickle-cell burden exists, remain unknown. In vitro studies suggest hydroxyurea could increase malaria severity, and hydroxyurea-associated neutropenia could worsen infections. NOHARM (Novel use Of Hydroxyurea in an African Region with Malaria) was a randomized, double-blinded, placebo-controlled trial conducted in malaria-endemic Uganda, comparing hydroxyurea to placebo at 20 ± 2.5 mg/kg per day for 12 months. The primary outcome was incidence of clinical malaria. Secondary outcomes included SCA-related adverse events (AEs), clinical and laboratory effects, and hematological toxicities. Children received either hydroxyurea (N = 104) or placebo (N = 103). Malaria incidence did not differ between children on hydroxyurea (0.05 episodes per child per year; 95% confidence interval [0.02, 0.13]) vs placebo (0.07 episodes per child per year [0.03, 0.16]); the hydroxyurea/placebo malaria incidence rate ratio was 0.7 ([0.2, 2.7]; P = .61). Time to infection also did not differ significantly between treatment arms. A composite SCA-related clinical outcome (vaso-occlusive painful crisis, dactylitis, acute chest syndrome, splenic sequestration, or blood transfusion) was less frequent with hydroxyurea (45%) than placebo (69%; P = .001). Children receiving hydroxyurea had significantly increased hemoglobin concentration and fetal hemoglobin, with decreased leukocytes and reticulocytes. Serious AEs, sepsis episodes, and dose-limiting toxicities were similar between treatment arms. Three deaths occurred (2 hydroxyurea, 1 placebo, and none from malaria). Hydroxyurea treatment appears safe for children with SCA living in malaria-endemic sub-Saharan Africa, without increased severe malaria, infections, or AEs. Hydroxyurea provides SCA-related laboratory and clinical efficacy, but optimal dosing and monitoring regimens for Africa remain undefined. This trial was registered at www.clinicaltrials.gov as #NCT01976416., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
10. Sickle cell disease.
- Author
-
Ware RE, de Montalembert M, Tshilolo L, and Abboud MR
- Subjects
- Anemia, Sickle Cell complications, Anemia, Sickle Cell diagnosis, Antisickling Agents therapeutic use, Blood Transfusion methods, Cerebrovascular Disorders prevention & control, Chronic Disease, Early Diagnosis, Female, Genetic Therapy methods, Global Burden of Disease, Hemolysis, Hemolytic Plaque Technique, Humans, Hydroxyurea therapeutic use, Iron Overload therapy, Point-of-Care Systems, Pregnancy, Pregnancy Complications, Hematologic prevention & control, Stem Cell Transplantation methods, Stroke etiology, Stroke prevention & control, Anemia, Sickle Cell therapy
- Abstract
Sickle cell disease is a common and life-threatening haematological disorder that affects millions of people worldwide. Abnormal sickle-shaped erythrocytes disrupt blood flow in small vessels, and this vaso-occlusion leads to distal tissue ischaemia and inflammation, with symptoms defining the acute painful sickle-cell crisis. Repeated sickling and ongoing haemolytic anaemia, even when subclinical, lead to parenchymal injury and chronic organ damage, causing substantial morbidity and early mortality. Currently available treatments are limited to transfusions and hydroxycarbamide, although stem cell transplantation might be a potentially curative therapy. Several new therapeutic options are in development, including gene therapy and gene editing. Recent advances include systematic universal screening for stroke risk, improved management of iron overload using oral chelators and non-invasive MRI measurements, and point-of-care diagnostic devices. Controversies include the role of haemolysis in sickle cell disease pathophysiology, optimal management of pregnancy, and strategies to prevent cerebrovascular disease., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
11. Sickle cell anemia in sub-Saharan Africa: advancing the clinical paradigm through partnerships and research.
- Author
-
McGann PT, Hernandez AG, and Ware RE
- Subjects
- Africa South of the Sahara epidemiology, Anemia, Sickle Cell therapy, Female, Humans, Male, Anemia, Sickle Cell diagnosis, Anemia, Sickle Cell epidemiology
- Published
- 2017
- Full Text
- View/download PDF
12. Pilot of the Chronic Disease Self-Management Program for Adolescents and Young Adults With Sickle Cell Disease.
- Author
-
Crosby LE, Joffe NE, Peugh J, Ware RE, and Britto MT
- Subjects
- Adolescent, Adult, Chronic Disease, Cohort Studies, Feasibility Studies, Female, Follow-Up Studies, Humans, Male, Pilot Projects, Quality of Life psychology, Self-Management psychology, Young Adult, Anemia, Sickle Cell therapy, Attitude to Health, Self-Management methods
- Abstract
Purpose: This study evaluated the feasibility of a group self-management intervention, the well-established Stanford Chronic Disease Self-Management Program (CDSMP), for adolescents and young adults (AYA) with sickle cell disease (SCD)., Methods: A total of 22 AYA participants with SCD, ages 16-24 years, completed self-efficacy and quality of life measures before the CDSMP, after, and 3 and 6 months later., Results: This AYA cohort showed significant improvements in self-efficacy (primary outcome) after the intervention. Analyses of follow-up data revealed a medium effect of the CDSMP on patient activation 3 months post although this was not sustained. Participants were highly satisfied, but only 64% completed the program., Conclusions: This study demonstrates that the CDSMP is acceptable and has the ability to improve self-efficacy. Additional research is needed to determine feasibility and evaluate health outcomes for AYA with SCD., (Copyright © 2016 Society for Adolescent Health and Medicine. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
13. Hydroxycarbamide versus chronic transfusion for maintenance of transcranial doppler flow velocities in children with sickle cell anaemia-TCD With Transfusions Changing to Hydroxyurea (TWiTCH): a multicentre, open-label, phase 3, non-inferiority trial.
- Author
-
Ware RE, Davis BR, Schultz WH, Brown RC, Aygun B, Sarnaik S, Odame I, Fuh B, George A, Owen W, Luchtman-Jones L, Rogers ZR, Hilliard L, Gauger C, Piccone C, Lee MT, Kwiatkowski JL, Jackson S, Miller ST, Roberts C, Heeney MM, Kalfa TA, Nelson S, Imran H, Nottage K, Alvarez O, Rhodes M, Thompson AA, Rothman JA, Helton KJ, Roberts D, Coleman J, Bonner MJ, Kutlar A, Patel N, Wood J, Piller L, Wei P, Luden J, Mortier NA, Stuber SE, Luban NLC, Cohen AR, Pressel S, and Adams RJ
- Subjects
- Adolescent, Anemia, Sickle Cell physiopathology, Blood Flow Velocity, Cerebrovascular Circulation physiology, Child, Child, Preschool, Combined Modality Therapy, Drug Substitution, Female, Humans, Male, Stroke etiology, Treatment Outcome, Ultrasonography, Doppler, Transcranial, Anemia, Sickle Cell drug therapy, Antisickling Agents therapeutic use, Blood Transfusion methods, Hydroxyurea therapeutic use
- Abstract
Background: For children with sickle cell anaemia and high transcranial doppler (TCD) flow velocities, regular blood transfusions can effectively prevent primary stroke, but must be continued indefinitely. The efficacy of hydroxycarbamide (hydroxyurea) in this setting is unknown; we performed the TWiTCH trial to compare hydroxyurea with standard transfusions., Methods: TWiTCH was a multicentre, phase 3, randomised, open-label, non-inferiority trial done at 26 paediatric hospitals and health centres in the USA and Canada. We enrolled children with sickle cell anaemia who were aged 4-16 years and had abnormal TCD flow velocities (≥ 200 cm/s) but no severe vasculopathy. After screening, eligible participants were randomly assigned 1:1 to continue standard transfusions (standard group) or hydroxycarbamide (alternative group). Randomisation was done at a central site, stratified by site with a block size of four, and an adaptive randomisation scheme was used to balance the covariates of baseline age and TCD velocity. The study was open-label, but TCD examinations were read centrally by observers masked to treatment assignment and previous TCD results. Participants assigned to standard treatment continued to receive monthly transfusions to maintain 30% sickle haemoglobin or lower, while those assigned to the alternative treatment started oral hydroxycarbamide at 20 mg/kg per day, which was escalated to each participant's maximum tolerated dose. The treatment period lasted 24 months from randomisation. The primary study endpoint was the 24 month TCD velocity calculated from a general linear mixed model, with the non-inferiority margin set at 15 cm/s. The primary analysis was done in the intention-to-treat population and safety was assessed in all patients who received at least one dose of assigned treatment. This study is registered with ClinicalTrials.gov, number NCT01425307., Findings: Between Sept 20, 2011, and April 17, 2013, 159 patients consented and enrolled in TWiTCH. 121 participants passed screening and were then randomly assigned to treatment (61 to transfusions and 60 to hydroxycarbamide). At the first scheduled interim analysis, non-inferiority was shown and the sponsor terminated the study. Final model-based TCD velocities were 143 cm/s (95% CI 140-146) in children who received standard transfusions and 138 cm/s (135-142) in those who received hydroxycarbamide, with a difference of 4·54 (0·10-8·98). Non-inferiority (p=8·82 × 10(-16)) and post-hoc superiority (p=0·023) were met. Of 29 new neurological events adjudicated centrally by masked reviewers, no strokes were identified, but three transient ischaemic attacks occurred in each group. Magnetic resonance brain imaging and angiography (MRI and MRA) at exit showed no new cerebral infarcts in either treatment group, but worsened vasculopathy in one participant who received standard transfusions. 23 severe adverse events in nine (15%) patients were reported for hydroxycarbamide and ten serious adverse events in six (10%) patients were reported for standard transfusions. The most common serious adverse event in both groups was vaso-occlusive pain (11 events in five [8%] patients with hydroxycarbamide and three events in one [2%] patient for transfusions)., Interpretation: For high-risk children with sickle cell anaemia and abnormal TCD velocities who have received at least 1 year of transfusions, and have no MRA-defined severe vasculopathy, hydroxycarbamide treatment can substitute for chronic transfusions to maintain TCD velocities and help to prevent primary stroke., Funding: National Heart, Lung, and Blood Institute, National Institutes of Health., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
14. Magnetic resonance imaging/angiography and transcranial Doppler velocities in sickle cell anemia: results from the SWiTCH trial.
- Author
-
Helton KJ, Adams RJ, Kesler KL, Lockhart A, Aygun B, Driscoll C, Heeney MM, Jackson SM, Krishnamurti L, Miller ST, Sarnaik SA, Schultz WH, and Ware RE
- Subjects
- Adolescent, Anemia, Sickle Cell therapy, Blood Flow Velocity, Blood Transfusion, Brain blood supply, Brain pathology, Cerebrovascular Circulation, Child, Child, Preschool, Female, Functional Neuroimaging, Humans, Hydroxyurea therapeutic use, Male, Prognosis, Secondary Prevention, Stroke prevention & control, Young Adult, Anemia, Sickle Cell complications, Anemia, Sickle Cell diagnosis, Magnetic Resonance Angiography, Magnetic Resonance Imaging, Stroke diagnosis, Stroke etiology, Ultrasonography, Doppler, Transcranial
- Abstract
The Stroke With Transfusions Changing to Hydroxyurea (SWiTCH) trial compared standard (transfusions/chelation) to alternative (hydroxyurea/phlebotomy) treatment to prevent recurrent stroke and manage iron overload in children chronically transfused over 7 years before enrollment. Standardized brain magnetic resonance imaging/magnetic resonance angiography (MRA) and transcranial Doppler (TCD) exams were performed at entry and exit, with a central blinded review. A novel MRA vasculopathy grading scale demonstrated frequent severe baseline left/right vessel stenosis (53%/41% ≥Grade 4); 31% had no vessel stenosis on either side. Baseline parenchymal injury was prevalent (85%/79% subcortical, 53%/37% cortical, 50%/35% subcortical and cortical). Most children had low or uninterpretable baseline middle cerebral artery TCD velocities, which were associated with worse stenoses (incidence risk ratio [IRR] = 5.1, P ≤ .0001 and IRR = 4.1, P < .0001) than normal velocities; only 2% to 12% had any conditional/abnormal velocity. Patients with adjudicated stroke (7) and transient ischemic attacks (19 in 11 standard/8 alternative arm subjects) had substantial parenchymal injury/vessel stenosis. At exit, 1 child (alternative arm) had a new silent infarct, and another had worse stenosis. SWiTCH neuroimaging data document severe parenchymal and vascular abnormalities in children with SCA and stroke and support concerns about chronic transfusions lacking effectiveness for preventing progressive cerebrovascular injury. The novel SWiTCH vasculopathy grading scale warrants validation testing and consideration for use in future clinical trials. This trial was registered at www.clinicaltrials.gov as #NCT00122980., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
15. Genetic mapping and exome sequencing identify 2 mutations associated with stroke protection in pediatric patients with sickle cell anemia.
- Author
-
Flanagan JM, Sheehan V, Linder H, Howard TA, Wang YD, Hoppe CC, Aygun B, Adams RJ, Neale GA, and Ware RE
- Subjects
- Adolescent, Anemia, Sickle Cell genetics, Child, Child, Preschool, Cohort Studies, Exome, Genome-Wide Association Study, Golgi Matrix Proteins, Humans, Mutation, Risk Factors, Anemia, Sickle Cell complications, Membrane Proteins genetics, Phosphoric Diester Hydrolases genetics, Polymorphism, Single Nucleotide, Pyrophosphatases genetics, Stroke etiology, Stroke genetics
- Abstract
Stroke is a devastating complication of sickle cell anemia (SCA), occurring in 11% of patients before age 20 years. Previous studies of sibling pairs have demonstrated a genetic component to the development of cerebrovascular disease in SCA, but few candidate genetic modifiers have been validated as having a substantial effect on stroke risk. We performed an unbiased whole-genome search for genetic modifiers of stroke risk in SCA. Genome-wide association studies were performed using genotype data from single-nucleotide polymorphism arrays, whereas a pooled DNA approach was used to perform whole-exome sequencing. In combination, 22 nonsynonymous variants were identified and represent key candidates for further in-depth study. To validate the association of these mutations with the risk for stroke, the 22 candidate variants were genotyped in an independent cohort of control patients (n = 231) and patients with stroke (n = 57) with SCA. One mutation in GOLGB1 (Y1212C) and another mutation in ENPP1 (K173Q) were confirmed as having significant associations with a decreased risk for stroke. These mutations were discovered and validated by an unbiased whole-genome approach, and future studies will focus on how these functional mutations may lead to protection from stroke in the context of SCA.
- Published
- 2013
- Full Text
- View/download PDF
16. Impact of hydroxyurea on clinical events in the BABY HUG trial.
- Author
-
Thornburg CD, Files BA, Luo Z, Miller ST, Kalpatthi R, Iyer R, Seaman P, Lebensburger J, Alvarez O, Thompson B, Ware RE, and Wang WC
- Subjects
- Acute Chest Syndrome chemically induced, Acute Chest Syndrome diagnosis, Acute Chest Syndrome epidemiology, Anemia, Sickle Cell complications, Anemia, Sickle Cell epidemiology, Antisickling Agents adverse effects, Antisickling Agents therapeutic use, Child, Preschool, Double-Blind Method, Drug-Related Side Effects and Adverse Reactions epidemiology, Female, Hospitalization statistics & numerical data, Humans, Infant, Inflammation chemically induced, Inflammation diagnosis, Inflammation epidemiology, Male, Pain chemically induced, Pain diagnosis, Pain epidemiology, Placebos, Anemia, Sickle Cell drug therapy, Hydroxyurea adverse effects, Hydroxyurea therapeutic use
- Abstract
The Pediatric Hydroxyurea Phase 3 Clinical Trial (BABY HUG) was a phase 3 multicenter, randomized, double-blind, placebo-controlled clinical trial of hydroxyurea in infants (beginning at 9-18 months of age) with sickle cell anemia. An important secondary objective of this study was to compare clinical events between the hydroxyurea and placebo groups. One hundred and ninety-three subjects were randomized to hydroxyurea (20 mg/kg/d) or placebo; there were 374 patient-years of on-study observation. Hydroxyurea was associated with statistically significantly lower rates of initial and recurrent episodes of pain, dactylitis, acute chest syndrome, and hospitalization; even infants who were asymptomatic at enrollment had less dactylitis as well as fewer hospitalizations and transfusions if treated with hydroxyurea. Despite expected mild myelosuppression, hydroxyurea was not associated with an increased risk of bacteremia or serious infection. These data provide important safety and efficacy information for clinicians considering hydroxyurea therapy for very young children with sickle cell anemia. This clinical trial is registered with the National Institutes of Health (NCT00006400, www.clinicaltrials.gov).
- Published
- 2012
- Full Text
- View/download PDF
17. Red blood cell alloimmunization in sickle cell disease: pathophysiology, risk factors, and transfusion management.
- Author
-
Yazdanbakhsh K, Ware RE, and Noizat-Pirenne F
- Subjects
- Anemia, Sickle Cell epidemiology, Anemia, Sickle Cell physiopathology, Humans, Risk Factors, Anemia, Sickle Cell immunology, Blood Group Incompatibility immunology, Blood Transfusion, Erythrocytes immunology, Isoantigens immunology
- Abstract
Red blood cell transfusions have reduced morbidity and mortality for patients with sickle cell disease. Transfusions can lead to erythrocyte alloimmunization, however, with serious complications for the patient including life-threatening delayed hemolytic transfusion reactions and difficulty in finding compatible units, which can cause transfusion delays. In this review, we discuss the risk factors associated with alloimmunization with emphasis on possible mechanisms that can trigger delayed hemolytic transfusion reactions in sickle cell disease, and we describe the challenges in transfusion management of these patients, including opportunities and emerging approaches for minimizing this life-threatening complication.
- Published
- 2012
- Full Text
- View/download PDF
18. Silent cerebral infarcts: a review on a prevalent and progressive cause of neurologic injury in sickle cell anemia.
- Author
-
DeBaun MR, Armstrong FD, McKinstry RC, Ware RE, Vichinsky E, and Kirkham FJ
- Subjects
- Adolescent, Adult, Anemia, Sickle Cell complications, Anemia, Sickle Cell diagnostic imaging, Anemia, Sickle Cell psychology, Asymptomatic Diseases epidemiology, Cerebral Infarction diagnostic imaging, Child, Child, Preschool, Disease Progression, Female, Humans, Male, Nervous System Diseases diagnostic imaging, Nervous System Diseases etiology, Prevalence, Radiography, Risk Factors, Young Adult, Anemia, Sickle Cell epidemiology, Cerebral Infarction epidemiology, Cerebral Infarction etiology, Nervous System Diseases epidemiology
- Abstract
Silent cerebral infarct (SCI) is the most common form of neurologic disease in children with sickle cell anemia (SCA). SCI is defined as abnormal magnetic resonance imaging (MRI) of the brain in the setting of a normal neurologic examination without a history or physical findings associated with an overt stroke. SCI occurs in 27% of this population before their sixth, and 37% by their 14th birthdays. In adults with SCA, the clinical history of SCI is poorly defined, although recent evidence suggests that they too may have ongoing risk of progressive injury. Risk factors for SCI include male sex, lower baseline hemoglobin concentration, higher baseline systolic blood pressure, and previous seizures. Specific morbidity associated with SCI includes a decrement in general intellectual abilities, poor academic achievement, progression to overt stroke, and progressive SCI. In addition, children with previous stroke continue to have both overt strokes and new SCI despite receiving regular blood transfusion therapy for secondary stroke prevention. Studies that only include overt stroke as a measure of CNS injury significantly underestimate the total cerebral injury burden in this population. In this review, we describe the epidemiology, natural history, morbidity, medical management, and potential therapeutic options for SCI in SCA.
- Published
- 2012
- Full Text
- View/download PDF
19. Stroke With Transfusions Changing to Hydroxyurea (SWiTCH).
- Author
-
Ware RE and Helms RW
- Subjects
- Adolescent, Anemia, Sickle Cell mortality, Anemia, Sickle Cell therapy, Child, Female, Humans, Male, Maximum Tolerated Dose, Phlebotomy, Prognosis, Stroke mortality, Survival Rate, Treatment Outcome, Anemia, Sickle Cell complications, Antisickling Agents therapeutic use, Hydroxyurea therapeutic use, Secondary Prevention, Stroke drug therapy, Stroke etiology, Transfusion Reaction
- Abstract
Stroke is a devastating complication of sickle cell anemia (SCA) with high recurrence if untreated. Chronic transfusions reduce recurrent strokes but have associated morbidities including iron overload. Stroke With Transfusions Changing to Hydroxyurea (SWiTCH) was a multicenter phase 3 randomized trial comparing standard treatment (transfusions/chelation) to alternative treatment (hydroxyurea/phlebotomy) for children with SCA, stroke, and iron overload. SWiTCH was a noninferiority trial with a composite primary end point, allowing an increased stroke risk but requiring superiority for removing iron. Subjects on standard treatment received monthly transfusions plus daily deferasirox iron chelation. Subjects on alternative treatment received hydroxyurea plus overlap transfusions during dose escalation to maximum tolerated dose (MTD), followed by monthly phlebotomy. Subjects on standard treatment (N = 66) maintained 30% sickle hemoglobin (HbS) and tolerated deferasirox at 28.2 ± 6.0 mg/kg/d. Subjects on alternative treatment (N = 67) initiated hydroxyurea and 60 (90%) reached MTD at 26.2 ± 4.9 mg/kg/d with 29.1% ± 6.7% fetal hemoglobin (HbF). Adjudication documented no strokes on transfusions/chelation but 7 (10%) on hydroxyurea/phlebotomy, still within the noninferiority stroke margin. The National Heart, Lung, and Blood Institute closed SWiTCH after interim analysis revealed equivalent liver iron content, indicating futility for the composite primary end point. Transfusions and chelation remain a better way to manage children with SCA, stroke, and iron overload.
- Published
- 2012
- Full Text
- View/download PDF
20. Hydroxyurea therapy of a murine model of sickle cell anemia inhibits the progression of pneumococcal disease by down-modulating E-selectin.
- Author
-
Lebensburger JD, Howard T, Hu Y, Pestina TI, Gao G, Johnson M, Zakharenko SS, Ware RE, Tuomanen EI, Persons DA, and Rosch JW
- Subjects
- Anemia, Sickle Cell complications, Anemia, Sickle Cell metabolism, Animals, Antisickling Agents therapeutic use, Child, Disease Models, Animal, E-Selectin blood, E-Selectin genetics, Female, Humans, Immunohistochemistry, Lung drug effects, Lung microbiology, Lung pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Fluorescence, Neutrophils drug effects, Neutrophils pathology, Pneumonia, Pneumococcal complications, Survival Analysis, Anemia, Sickle Cell drug therapy, E-Selectin metabolism, Hydroxyurea therapeutic use, Pneumonia, Pneumococcal prevention & control
- Abstract
Sickle cell anemia is characterized by chronic hemolysis coupled with extensive vascular inflammation. This inflammatory state also mechanistically promotes a high risk of lethal, invasive pneumococcal infection. Current treatments to reduce vaso-occlusive complications include chronic hydroxyurea therapy to induce fetal hemoglobin. Because hydroxyurea also reduces leukocytosis, an understanding of the impact of this treatment on pneumococcal pathogenesis is needed. Using a sickle cell mouse model of pneumococcal pneumonia and sepsis, administration of hydroxyurea was found to significantly improve survival. Hydroxyurea treatment decreased neutrophil extravasation into the infected lung coincident with significantly reduced levels of E-selectin in serum and on pulmonary epithelia. The protective effect of hydroxyurea was abrogated in mice deficient in E-selectin. The decrease in E-selectin levels was also evident in human sickle cell patients receiving hydroxyurea therapy. These data indicate that in addition to induction of fetal hemoglobin, hydroxyurea attenuates leukocyte-endothelial interactions in sickle cell anemia, resulting in protection against lethal pneumococcal sepsis.
- Published
- 2012
- Full Text
- View/download PDF
21. Epigenetic and molecular profiles of erythroid cells after hydroxyurea treatment in sickle cell anemia.
- Author
-
Walker AL, Steward S, Howard TA, Mortier N, Smeltzer M, Wang YD, and Ware RE
- Subjects
- Adult, Antisickling Agents therapeutic use, Child, CpG Islands drug effects, DNA Methylation drug effects, Erythrocytes drug effects, Erythrocytes physiology, Fetal Hemoglobin metabolism, Gene Expression Profiling, Humans, MicroRNAs genetics, Prospective Studies, Reticulocytes drug effects, Reticulocytes physiology, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell genetics, Epigenesis, Genetic drug effects, Erythroid Cells drug effects, Fetal Hemoglobin genetics, Hydroxyurea therapeutic use
- Abstract
Hydroxyurea has been shown to be efficacious for the treatment of sickle cell anemia (SCA), primarily through the induction of fetal hemoglobin (HbF). However, the exact mechanisms by which hydroxyurea can induce HbF remain incompletely defined, although direct transcriptional effects and altered cell cycle kinetics have been proposed. In this study, we investigated potential epigenetic and alternative molecular mechanisms of hydroxyurea-mediated HbF induction by examining methylation patterns within the (G)γ-globin promoter and miRNA expression within primary CD71(+) erythrocytes of patients with SCA, both at baseline before beginning hydroxyurea therapy and after reaching maximum tolerated dose (MTD). Using both cross-sectional analysis and paired-sample analysis, we found that the highly methylated (G)γ-globin promoter was inversely correlated to baseline HbF levels, but only slightly altered by hydroxyurea treatment. Conversely, expression of several specific miRNAs was significantly increased after hydroxyurea treatment, and expression of miR-26b and miR-151-3p were both associated with HbF levels at MTD. The significant associations identified in these studies suggest that methylation may be important for regulation of baseline HbF, but not after hydroxyurea treatment, whereas changes in miRNA expression may be associated with hydroxyurea-mediated HbF induction. This study was registered at ClinicalTrials.gov (NCT00305175).
- Published
- 2011
- Full Text
- View/download PDF
22. Pharmacokinetics, pharmacodynamics, and pharmacogenetics of hydroxyurea treatment for children with sickle cell anemia.
- Author
-
Ware RE, Despotovic JM, Mortier NA, Flanagan JM, He J, Smeltzer MP, Kimble AC, Aygun B, Wu S, Howard T, and Sparreboom A
- Subjects
- Adolescent, Biomarkers, Pharmacological analysis, Biomarkers, Pharmacological metabolism, Child, Child, Preschool, DNA Mutational Analysis, Dose-Response Relationship, Drug, Female, Humans, Hydroxyurea administration & dosage, Hydroxyurea pharmacology, Male, Maximum Tolerated Dose, Polymorphism, Single Nucleotide, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell genetics, Hydroxyurea pharmacokinetics, Hydroxyurea therapeutic use, Pharmacogenetics methods
- Abstract
Hydroxyurea therapy has proven laboratory and clinical efficacies for children with sickle cell anemia (SCA). When administered at maximum tolerated dose (MTD), hydroxyurea increases fetal hemoglobin (HbF) to levels ranging from 10% to 40%. However, interpatient variability of percentage of HbF (%HbF) response is high, MTD itself is variable, and accurate predictors of hydroxyurea responses do not currently exist. HUSTLE (NCT00305175) was designed to provide first-dose pharmacokinetics (PK) data for children with SCA initiating hydroxyurea therapy, to investigate pharmacodynamics (PD) parameters, including HbF response and MTD after standardized dose escalation, and to evaluate pharmacogenetics influences on PK and PD parameters. For 87 children with first-dose PK studies, substantial interpatient variability was observed, plus a novel oral absorption phenotype (rapid or slow) that influenced serum hydroxyurea levels and total hydroxyurea exposure. PD responses in 174 subjects were robust and similar to previous cohorts; %HbF at MTD was best predicted by 5 variables, including baseline %HbF, whereas MTD was best predicted by 5 variables, including serum creatinine. Pharmacogenetics analysis showed single nucleotide polymorphisms influencing baseline %HbF, including 5 within BCL11A, but none influencing MTD %HbF or dose. Accurate prediction of hydroxyurea treatment responses for SCA remains a worthy but elusive goal.
- Published
- 2011
- Full Text
- View/download PDF
23. A pilot study of subcutaneous decitabine in β-thalassemia intermedia.
- Author
-
Olivieri NF, Saunthararajah Y, Thayalasuthan V, Kwiatkowski J, Ware RE, Kuypers FA, Kim HY, Trachtenberg FL, and Vichinsky EP
- Subjects
- Adult, Azacitidine administration & dosage, Cell Differentiation drug effects, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases deficiency, DNA Methylation drug effects, Decitabine, Erythrocytes drug effects, Female, Hemoglobins metabolism, Humans, Injections, Subcutaneous, Male, Middle Aged, Pilot Projects, Prognosis, Young Adult, beta-Thalassemia genetics, Antimetabolites, Antineoplastic administration & dosage, Azacitidine analogs & derivatives, Gene Expression Regulation drug effects, beta-Thalassemia drug therapy, gamma-Globins genetics
- Abstract
Ineffective erythropoiesis, the hallmark of β-thalassemia, is a result of α/non-α globin chain imbalance. One strategy to redress globin-chain imbalance is to induce γ-globin gene (HBG) expression. Repression of HBG in adult erythroid cells involves DNA methylation and other epigenetic changes. Therefore, the cytosine analog decitabine, which can deplete DNA methyltransferase 1 (DNMT1), can potentially activate HBG. In 5 patients with β-thalassemia intermedia, a dose and schedule of decitabine intended to deplete DNMT1 without causing significant cytotoxicity (0.2 mg/kg subcutaneous 2 times per week for 12 weeks) increased total hemoglobin from 7.88 ± 0.88 g/dL to 9.04 ± 0.77 g/dL (P = .004) and absolute fetal hemoglobin from 3.64 ± 1.13 g/dL to 4.29 ± 1.13 g/dL (P = .003). Significant favorable changes also occurred in indices of hemolysis and red blood cell densitometry. Consistent with a noncytotoxic, differentiation altering mechanism of action, the major side effect was an asymptomatic increase in platelet counts without erythrocyte micronucleus or VDJ recombination assay evidence of genotoxicity. This study was registered at www.clinicaltrials.gov as #NCT00661726.
- Published
- 2011
- Full Text
- View/download PDF
24. Thrombopoiesis: new ITP paradigm?
- Author
-
Despotovic JM and Ware RE
- Subjects
- Humans, Receptors, Thrombopoietin agonists, Purpura, Thrombocytopenic, Idiopathic drug therapy, Purpura, Thrombocytopenic, Idiopathic physiopathology, Receptors, Fc therapeutic use, Recombinant Fusion Proteins therapeutic use, Thrombopoiesis drug effects, Thrombopoiesis physiology, Thrombopoietin therapeutic use
- Published
- 2011
- Full Text
- View/download PDF
25. Genetic predictors for stroke in children with sickle cell anemia.
- Author
-
Flanagan JM, Frohlich DM, Howard TA, Schultz WH, Driscoll C, Nagasubramanian R, Mortier NA, Kimble AC, Aygun B, Adams RJ, Helms RW, and Ware RE
- Subjects
- Adolescent, Anemia, Sickle Cell complications, Anemia, Sickle Cell diagnosis, Child, Child, Preschool, Cohort Studies, Female, Glucosephosphate Dehydrogenase genetics, Glucosephosphate Dehydrogenase Deficiency complications, Glucosephosphate Dehydrogenase Deficiency diagnosis, Glucosephosphate Dehydrogenase Deficiency genetics, Humans, Male, Polymorphism, Single Nucleotide physiology, Risk Factors, Stroke diagnosis, Stroke etiology, beta-Globins genetics, Anemia, Sickle Cell genetics, Genetic Markers physiology, Genetic Predisposition to Disease, Stroke genetics
- Abstract
Stroke is a devastating complication of sickle cell anemia (SCA), affecting 5% to 10% of patients before adulthood. Several candidate genetic polymorphisms have been proposed to affect stroke risk, but few have been validated, mainly because previous studies were hampered by relatively small sample sizes and the absence of additional patient cohorts for validation testing. To verify the accuracy of proposed genetic modifiers influencing stroke risk in SCA, we performed genotyping for 38 published single nucleotide polymorphisms (SNPs), as well as α-thalassemia, G6PD A(-) variant deficiency, and β-globin haplotype in 2 cohorts of children with well-defined stroke phenotypes (130 stroke, 103 nonstroke). Five polymorphisms had significant influence (P < .05): SNPs in the ANXA2, TGFBR3, and TEK genes were associated with increased stroke risk, whereas α-thalassemia and a SNP in the ADCY9 gene were linked with decreased stroke risk. Further investigation at these genetic regions may help define mutations that confer stroke risk or protection in children with SCA.
- Published
- 2011
- Full Text
- View/download PDF
26. Hydroxycarbamide in very young children with sickle-cell anaemia: a multicentre, randomised, controlled trial (BABY HUG).
- Author
-
Wang WC, Ware RE, Miller ST, Iyer RV, Casella JF, Minniti CP, Rana S, Thornburg CD, Rogers ZR, Kalpatthi RV, Barredo JC, Brown RC, Sarnaik SA, Howard TH, Wynn LW, Kutlar A, Armstrong FD, Files BA, Goldsmith JC, Waclawiw MA, Huang X, and Thompson BW
- Subjects
- Acute Chest Syndrome etiology, Acute Chest Syndrome prevention & control, Anemia, Sickle Cell complications, Anemia, Sickle Cell metabolism, Anemia, Sickle Cell pathology, Antisickling Agents adverse effects, Biomarkers blood, Blood Cell Count, Child Development, Female, Glomerular Filtration Rate, Hemoglobins metabolism, Humans, Hydroxyurea adverse effects, Infant, Male, Osmolar Concentration, Pain etiology, Pain prevention & control, Spleen pathology, Spleen physiopathology, Technetium Tc 99m Pentetate metabolism, Treatment Outcome, Ultrasonography, Doppler, Transcranial, United States, Urine chemistry, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell physiopathology, Antisickling Agents therapeutic use, Hydroxyurea therapeutic use
- Abstract
Background: Sickle-cell anaemia is associated with substantial morbidity from acute complications and organ dysfunction beginning in the first year of life. Hydroxycarbamide substantially reduces episodes of pain and acute chest syndrome, admissions to hospital, and transfusions in adults with sickle-cell anaemia. We assessed the effect of hydroxycarbamide therapy on organ dysfunction and clinical complications, and examined laboratory findings and toxic effects., Methods: This randomised trial was undertaken in 13 centres in the USA between October, 2003, and September, 2009. Eligible participants had haemoglobin SS (HbSS) or haemoglobin Sβ(0)thalassaemia, were aged 9-18 months at randomisation, and were not selected for clinical severity. Participants received liquid hydroxycarbamide, 20 mg/kg per day, or placebo for 2 years. Randomisation assignments were generated by the medical coordinating centre by a pre-decided schedule. Identical appearing and tasting formulations were used for hydroxycarbamide and placebo. Patients, caregivers, and coordinating centre staff were masked to treatment allocation. Primary study endpoints were splenic function (qualitative uptake on (99)Tc spleen scan) and renal function (glomerular filtration rate by (99m)Tc-DTPA clearance). Additional assessments included blood counts, fetal haemoglobin concentration, chemistry profiles, spleen function biomarkers, urine osmolality, neurodevelopment, transcranial Doppler ultrasonography, growth, and mutagenicity. Study visits occurred every 2-4 weeks. Analysis was by intention to treat. The trial is registered with ClinicalTrials.gov, number NCT00006400., Findings: 96 patients received hydroxycarbamide and 97 placebo, of whom 83 patients in the hydroxycarbamide group and 84 in the placebo group completed the study. Significant differences were not seen between groups for the primary endpoints (19 of 70 patients with decreased spleen function at exit in the hydroxycarbamide group vs 28 of 74 patients in the placebo group, p=0·21; and a difference in the mean increase in DTPA glomerular filtration rate in the hydroxycarbamide group versus the placebo group of 2 mL/min per 1·73 m(2), p=0·84). Hydroxycarbamide significantly decreased pain (177 events in 62 patients vs 375 events in 75 patients in the placebo group, p=0·002) and dactylitis (24 events in 14 patients vs 123 events in 42 patients in the placebo group, p<0·0001), with some evidence for decreased acute chest syndrome, hospitalisation rates, and transfusion. Hydroxyurea increased haemoglobin and fetal haemoglobin, and decreased white blood-cell count. Toxicity was limited to mild-to-moderate neutropenia., Interpretation: On the basis of the safety and efficacy data from this trial, hydroxycarbamide can now be considered for all very young children with sickle-cell anaemia., Funding: The US National Heart, Lung, and Blood Institute; and the National Institute of Child Health and Human Development., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
27. Biomarkers of splenic function in infants with sickle cell anemia: baseline data from the BABY HUG Trial.
- Author
-
Rogers ZR, Wang WC, Luo Z, Iyer RV, Shalaby-Rana E, Dertinger SD, Shulkin BL, Miller JH, Files B, Lane PA, Thompson BW, Miller ST, and Ware RE
- Subjects
- Anemia, Sickle Cell blood, Anemia, Sickle Cell pathology, Biomarkers metabolism, Erythrocyte Count, Erythrocyte Inclusions pathology, Female, Humans, Infant, Liver metabolism, Liver pathology, Male, Spleen pathology, Anemia, Sickle Cell physiopathology, Spleen physiopathology
- Abstract
We evaluated spleen function in 193 children with sickle cell anemia 8 to 18 months of age by (99m)Tc sulfur-colloid liver-spleen scan and correlated results with clinical and laboratory parameters, including 2 splenic biomarkers: pitted cell counts (PIT) and quantitative Howell-Jolly bodies (HJB) enumerated by flow cytometry. Loss of splenic function began before 12 months of age in 86% of infants in association with lower total or fetal hemoglobin and higher white blood cell or reticulocyte counts, reinforcing the need for early diagnosis and diligent preventive care. PIT and HJB correlated well with each other and liver-spleen scan results. Previously described biomarker threshold values did define patients with abnormal splenic function, but our data suggest that normal spleen function is better predicted by PIT of ≤1.2% or HJB ≤55/10(6) red blood cells and absent function by PIT ≥4.5% or HJB ≥665/10(6). HJB is methodologically advantageous compared with PIT, but both are valid biomarkers of splenic function. This trial was registered at www.clinicaltrials.gov as #NCT00006400., (© 2011 by The American Society of Hematology)
- Published
- 2011
- Full Text
- View/download PDF
28. Pulmonary hypertension and nitric oxide depletion in sickle cell disease.
- Author
-
Bunn HF, Nathan DG, Dover GJ, Hebbel RP, Platt OS, Rosse WF, and Ware RE
- Subjects
- Adult, Anemia, Sickle Cell blood, Anemia, Sickle Cell complications, Anemia, Sickle Cell drug therapy, Animals, Child, Clinical Trials as Topic, Disease Models, Animal, Echocardiography, Doppler, Endothelium, Vascular physiopathology, False Positive Reactions, Female, Hemoglobins analysis, Hemoglobins chemistry, Hemoglobinuria, Paroxysmal complications, Hemolysis, Humans, Hydroxyurea pharmacology, Hydroxyurea therapeutic use, Hypertension, Pulmonary blood, Hypertension, Pulmonary diagnostic imaging, Hypertension, Pulmonary physiopathology, L-Lactate Dehydrogenase blood, Leg Ulcer etiology, Leg Ulcer physiopathology, Male, Microcirculation, Multicenter Studies as Topic, Nitric Oxide administration & dosage, Nitric Oxide blood, Nitric Oxide physiology, Nitric Oxide therapeutic use, Priapism etiology, Priapism physiopathology, Thromboembolism etiology, Thromboembolism physiopathology, Tricuspid Valve Insufficiency diagnostic imaging, Tricuspid Valve Insufficiency etiology, Tricuspid Valve Insufficiency physiopathology, Anemia, Sickle Cell physiopathology, Hypertension, Pulmonary etiology, Models, Biological, Nitric Oxide deficiency
- Abstract
During the past decade a large body of experimental and clinical studies has focused on the hypothesis that nitric oxide (NO) depletion by plasma hemoglobin in the microcirculation plays a central role in the pathogenesis of many manifestations of sickle cell disease (SCD), particularly pulmonary hypertension. We have carefully examined those studies and believe that the conclusions drawn from them are not adequately supported by the data. We agree that NO depletion may well play a role in the pathophysiology of other hemolytic states such as paroxysmal nocturnal hemoglobinuria, in which plasma hemoglobin concentrations are often at least an order of magnitude greater than in SCD. Accordingly, we conclude that clinical trials in SCD designed to increase the bioavailability of NO or association studies in which SCD clinical manifestations are related to plasma hemoglobin via its surrogates should be viewed with caution.
- Published
- 2010
- Full Text
- View/download PDF
29. How I use hydroxyurea to treat young patients with sickle cell anemia.
- Author
-
Ware RE
- Subjects
- Adolescent, Child, Clinical Trials as Topic, Humans, Anemia, Sickle Cell drug therapy, Antisickling Agents therapeutic use, Hydroxyurea therapeutic use
- Abstract
Hydroxyurea has many characteristics of an ideal drug for sickle cell anemia (SCA) and provides therapeutic benefit through multiple mechanisms of action. Over the past 25 years, substantial experience has accumulated regarding its safety and efficacy for patients with SCA. Early proof-of-principle studies were followed by prospective phase 1/2 trials demonstrating efficacy in affected adults, then adolescents and children, and more recently infants and toddlers. The phase 3 National Heart, Lung and Blood Institute-sponsored Multicenter Study of Hydroxyurea trial proved clinical efficacy for preventing acute vaso-occlusive events in severely affected adults. Based on this cumulative experience, hydroxyurea has emerged as an important therapeutic option for children and adolescents with recurrent vaso-occlusive events; recent evidence documents sustained long-term benefits with prevention or reversal of chronic organ damage. Despite abundant evidence for its efficacy, however, hydroxyurea has not yet translated into effective therapy for SCA. Because many healthcare providers have inadequate knowledge about hydroxyurea, patients and families are not offered treatment or decline because of unrealistic fears. Limited support for hydroxyurea by lay organizations and inconsistent medical delivery systems also contribute to underuse. Although questions remain regarding its long-term risks and benefits, current evidence suggests that many young patients with SCA should receive hydroxyurea treatment.
- Published
- 2010
- Full Text
- View/download PDF
30. Assessment of genotoxicity associated with hydroxyurea therapy in children with sickle cell anemia.
- Author
-
Flanagan JM, Howard TA, Mortier N, Avlasevich SL, Smeltzer MP, Wu S, Dertinger SD, and Ware RE
- Subjects
- Adolescent, Child, Child, Preschool, Humans, Micronucleus Tests, Mutation, Time, Anemia, Sickle Cell drug therapy, Antisickling Agents adverse effects, DNA Damage, Hydroxyurea adverse effects, Micronuclei, Chromosome-Defective
- Abstract
Hydroxyurea induces fetal hemoglobin, improves laboratory parameters, and ameliorates clinical complications of sickle cell anemia (SCA), but its long-term efficacy and safety in this patient population remain incompletely defined. Although generally considered non-DNA reactive, an important safety concern is that hydroxyurea may indirectly cause genotoxic damage. To better address this safety issue of hydroxyurea in patients with SCA, we measured the production of micronuclei (MN) in red blood cells (RBCs) as a marker of genotoxicity. Blood samples were collected from children with SCA enrolled in the Hydroxyurea Study of Long-term Effects (ClinicalTrials.gov NCT00305175). Flow cytometry quantified circulating MN-containing erythrocyte sub-populations before and during hydroxyurea exposure. The frequency of micronucleated reticulocytes (MN-CD71(+)) and micronucleated mature erythrocytes (MN-RBC) was then tested for associations with laboratory and clinical data. In cross-sectional analysis of 293 blood samples from 105 children with SCA and a median of 2 years of hydroxyurea therapy, exposure to hydroxyurea was associated with significantly increased frequencies of MN-CD71(+) and MN-RBC compared to baseline. The increases were evident by 3 months of therapy, and did not escalate further with up to 12 years of continuous drug exposure. In prospective longitudinal analysis, substantial inter-individual variation in the effect of hydroxyurea on %MN-CD71(+) was observed that was associated with the expected laboratory effects of hydroxyurea. In conclusion, clinically relevant exposure to hydroxyurea is associated with increased MN production consistent with erythroblast genotoxicity but with substantial inter-patient variability. Associations between increased %MN-CD71(+) and laboratory benefits suggest that hydroxyurea effects on MN production may be related to individual patient sensitivity to hydroxyurea within the bone marrow., (Copyright 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
31. Sickle-cell disease: an ounce of prevention, a pound of cure.
- Author
-
Hankins J and Ware RE
- Subjects
- Africa epidemiology, Anemia, Sickle Cell complications, Anemia, Sickle Cell epidemiology, Bacteremia etiology, Child, Humans, Infant, Mass Screening, United States epidemiology, Anemia, Sickle Cell diagnosis
- Published
- 2009
- Full Text
- View/download PDF
32. R2* magnetic resonance imaging of the liver in patients with iron overload.
- Author
-
Hankins JS, McCarville MB, Loeffler RB, Smeltzer MP, Onciu M, Hoffer FA, Li CS, Wang WC, Ware RE, and Hillenbrand CM
- Subjects
- Adolescent, Adult, Biopsy, Calibration, Child, Female, Ferritins analysis, Ferritins blood, Humans, Iron Overload pathology, Liver pathology, Magnetic Resonance Imaging standards, Male, Radiography, Single-Blind Method, Young Adult, Iron Overload diagnostic imaging, Liver diagnostic imaging, Magnetic Resonance Imaging methods
- Abstract
R2* magnetic resonance imaging (R2*-MRI) can quantify hepatic iron content (HIC) by noninvasive means but is not fully investigated. Patients with iron overload completed 1.5T R2*-MRI examination and liver biopsy within 30 days. Forty-three patients (sickle cell anemia, n = 32; beta-thalassemia major, n = 6; and bone marrow failure, n = 5) were analyzed: median age, 14 years, median transfusion duration, 15 months, average (+/-SD) serum ferritin 2718 plus or minus 1994 ng/mL, and average HIC 10.9 plus or minus 6.8 mg Fe/g dry weight liver. Regions of interest were drawn and analyzed by 3 independent reviewers with excellent agreement of their measurements (intraclass correlation coefficient = 0.98). Ferritin and R2*-MRI were weakly but significantly associated (range of correlation coefficients among the 3 reviewers, 0.41-0.48; all P < .01). R2*-MRI was strongly associated with HIC for all 3 reviewers (correlation coefficients, 0.96-0.98; all P < .001). This high correlation confirms prior reports, calibrates R2*-MRI measurements, and suggests its clinical utility for predicting HIC using R2*-MRI. This study was registered at www.clinicaltrials.gov as #NCT00675038.
- Published
- 2009
- Full Text
- View/download PDF
33. Defective binding of factor XI-N248 to activated human platelets.
- Author
-
Sun MF, Ho D, Martincic D, Ware RE, Walsh PN, and Gailani D
- Published
- 2007
- Full Text
- View/download PDF
34. Hydroxyurea therapy lowers transcranial Doppler flow velocities in children with sickle cell anemia.
- Author
-
Zimmerman SA, Schultz WH, Burgett S, Mortier NA, and Ware RE
- Subjects
- Anemia, Sickle Cell complications, Anemia, Sickle Cell diagnostic imaging, Anemia, Sickle Cell therapy, Blood Transfusion, Child, Child, Preschool, Female, Humans, Male, Maximum Tolerated Dose, Middle Cerebral Artery diagnostic imaging, Prospective Studies, Stroke diagnostic imaging, Stroke etiology, Stroke prevention & control, Anemia, Sickle Cell physiopathology, Antisickling Agents administration & dosage, Blood Flow Velocity drug effects, Hydroxyurea administration & dosage, Stroke physiopathology, Ultrasonography, Doppler, Transcranial
- Abstract
Hydroxyurea has hematologic and clinical efficacy in sickle cell anemia (SCA), but its effects on transcranial Doppler (TCD) flow velocities remain undefined. Fifty-nine children initiating hydroxyurea therapy for clinical severity had pretreatment baseline TCD measurements; 37 with increased flow velocities (> or = 140 cm/s) were then enrolled in an institutional review board (IRB)-approved prospective phase 2 trial with TCD velocities measured at maximum tolerated dose (MTD) and one year later. At hydroxyurea MTD (mean +/- 1 SD = 27.9 +/- 2.7 mg/kg per day), significant decreases were observed in the right middle cerebral artery (MCA) (166 +/- 27 cm/s to 135 +/- 27 cm/s, P < .001) and left (MCA) (168 +/- 26 cm/s to 142 +/- 27 cm/s, P < .001) velocities. The magnitude of TCD velocity decline was significantly correlated with the maximal baseline TCD value. At hydroxyurea MTD, 14 of 15 children with conditional baseline TCD values improved, while 5 of 6 with abnormal TCD velocities whose families refused transfusions became less than 200 cm/s. TCD changes were sustained at follow-up. These prospective data indicate that hydroxyurea can significantly decrease elevated TCD flow velocities, often into the normal range. A multicenter trial is warranted to determine the efficacy of hydroxyurea for the management of increased TCD values, and ultimately for primary stroke prevention in children with SCA.
- Published
- 2007
- Full Text
- View/download PDF
35. Long-term hydroxyurea therapy for infants with sickle cell anemia: the HUSOFT extension study.
- Author
-
Hankins JS, Ware RE, Rogers ZR, Wynn LW, Lane PA, Scott JP, and Wang WC
- Subjects
- Blood Platelets cytology, Child, Child, Preschool, Clinical Trials as Topic, Dose-Response Relationship, Drug, Erythrocyte Indices, Female, Hematologic Tests, Hemoglobins chemistry, Homozygote, Humans, Hydroxyurea blood, Hydroxyurea metabolism, Infant, Leukocytes cytology, Liver pathology, Male, Reticulocytes cytology, Spleen pathology, Time Factors, Anemia, Sickle Cell drug therapy, Hydroxyurea pharmacology
- Abstract
The long-term efficacy and toxicity of hydroxyurea for infants are undefined, and its role in preventing organ dysfunction is unknown. Short-term feasibility of hydroxyurea administration, toxicities, hematologic effects, and effect on spleen function in infants with sickle cell anemia (SCA) were reported (Hydroxyurea Safety and Organ Toxicity [HUSOFT] trial). These infants completing 2 years of hydroxyurea therapy (20 mg/kg/d) were offered study extension with dose escalation to 30 mg/kg/d. Patients were monitored with laboratory tests and biannual imaging studies. Hematologic indices were compared with predicted age-specific values and event rates compared with historic rates. All 21 subjects completing the original trial enrolled in the extension study: median age, 3.4 years old (range, 2.6 to 4.4 years); 12 females; 20 with Hb SS, 1 with Hb S/beta0-thalassemia. Seventeen patients completed 4 years of hydroxyurea, and 11 completed 6 years. After 4 years, hydroxyurea was associated with increased hemoglobin concentration, percentage of fetal hemoglobin (Hb F), and mean corpuscular volume (MCV) and decreased reticulocytes, white blood cells (WBCs), and platelets (P < .01). Patients experienced 7.5 acute chest syndrome (ACS) events per 100 person-years, compared with 24.5 events per 100 person-years among historic controls (P = .001). Treated patients had better spleen function than expected and improved growth rates. Infants with SCA tolerate prolonged hydroxyurea therapy with sustained hematologic benefits, fewer ACS events, improved growth, and possibly preserved organ function.
- Published
- 2005
- Full Text
- View/download PDF
36. Sustained long-term hematologic efficacy of hydroxyurea at maximum tolerated dose in children with sickle cell disease.
- Author
-
Zimmerman SA, Schultz WH, Davis JS, Pickens CV, Mortier NA, Howard TA, and Ware RE
- Subjects
- Adolescent, Adult, Anemia, Sickle Cell genetics, Antisickling Agents adverse effects, Child, Child, Preschool, Cohort Studies, Female, Fetal Hemoglobin, Genotype, Humans, Hydroxyurea adverse effects, Infant, Leukocyte Count, Male, Treatment Outcome, Anemia, Sickle Cell drug therapy, Antisickling Agents administration & dosage, Hydroxyurea administration & dosage
- Abstract
Hydroxyurea improves hematologic parameters for children with sickle cell disease (SCD), but its long-term efficacy at maximum tolerated dose (MTD) has not been determined. Between 1995 and 2002, hydroxyurea therapy was initiated for 122 pediatric patients with SCD including 106 with homozygous sickle cell anemia (HbSS), 7 with sickle hemoglobin C (HbSC), 7 with sickle/beta-thalassemia (HbS/ beta-thalassemia [6 HbS/beta0, 1 HbS/beta+]), and 2 with sickle hemoglobin OArab (HbS/OArab). Median age at initiation of therapy was 11.1 years. Hydroxyurea was escalated to MTD, with an average dose of 25.4 +/- 5.4 mg/kg per day; the average duration of hydroxyurea therapy has been 45 +/- 24 months (range, 6-101 months). Hydroxyurea was discontinued for 15 (12%) children with poor compliance. Mild transient neutropenia occurred, but no hepatic or renal toxicity was noted. Hydroxyurea therapy led to significant increases in hemoglobin level, mean corpuscular volume, and fetal hemoglobin (HbF) level, whereas significant decreases occurred in reticulocyte, white blood cell, and platelet counts and serum bilirubin levels. Children with variant SCD genotypes also had hematologic responses to hydroxyurea. HbF induction has been sustained for up to 8 years without adverse effects on growth or increased numbers of acquired DNA mutations. Long-term hydroxyurea therapy at MTD is well tolerated by pediatric patients with SCD and has sustained hematologic efficacy with apparent long-term safety.
- Published
- 2004
- Full Text
- View/download PDF
37. UGT1A promoter polymorphisms influence bilirubin response to hydroxyurea therapy in sickle cell anemia.
- Author
-
Heeney MM, Howard TA, Zimmerman SA, and Ware RE
- Subjects
- Anemia, Sickle Cell complications, Antisickling Agents administration & dosage, Bilirubin blood, Child, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Genotype, Hemolysis drug effects, Hemolysis genetics, Humans, Hydroxyurea administration & dosage, Male, Promoter Regions, Genetic, Reticulocyte Count, Retrospective Studies, Treatment Outcome, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell genetics, Antisickling Agents therapeutic use, Glucuronosyltransferase genetics, Hydroxyurea therapeutic use, Polymorphism, Genetic
- Abstract
Hydroxyurea therapy reduces hemolysis and decreases serum bilirubin levels in children and adults with sickle cell anemia (SCA) and may therefore help prevent the development of cholelithiasis in this patient population. We recently reported that a promoter polymorphism in the uridine diphosphoglucuronate glucuronosyltransferase 1A (UGT1A) gene affects steady-state bilirubin levels and the incidence of gallstones in children with SCA. We have now analyzed the influence of the UGT1A genotype on the therapeutic response to hydroxyurea. A large cohort of children with SCA taking hydroxyurea therapy at the maximum tolerated dose demonstrated significant reductions in hemolysis independent of UGT1A promoter polymorphism genotype, but the hydroxyurea-related decreases in serum bilirubin levels were significantly different. Children with the wild-type 6/6 UGT1A genotype demonstrated normalized bilirubin levels with hydroxyurea therapy, but children with the heterozygous 6/7 or abnormal 7/7 genotypes did not. Children with the abnormal 7/7 genotype, which confers the phenotype of Gilbert syndrome, had bilirubin levels greater than 3 mg/dL despite full-dose hydroxyurea therapy. These data indicate the UGT1A promoter polymorphism is a powerful nonglobin genetic modifier in SCA that influences serum bilirubin both at baseline and on hydroxyurea therapy. UGT1A promoter polymorphisms may therefore influence the ability of hydroxyurea to prevent gallstone formation in patients with SCA.
- Published
- 2003
- Full Text
- View/download PDF
38. Predictors of fetal hemoglobin response in children with sickle cell anemia receiving hydroxyurea therapy.
- Author
-
Ware RE, Eggleston B, Redding-Lallinger R, Wang WC, Smith-Whitley K, Daeschner C, Gee B, Styles LA, Helms RW, Kinney TR, and Ohene-Frempong K
- Subjects
- Antisickling Agents administration & dosage, Antisickling Agents adverse effects, Child, Hemoglobins analysis, Humans, Hydroxyurea administration & dosage, Hydroxyurea adverse effects, Leukocyte Count, Maximum Tolerated Dose, Patient Compliance, Platelet Count, Reticulocyte Count, Treatment Outcome, Anemia, Sickle Cell blood, Anemia, Sickle Cell drug therapy, Antisickling Agents therapeutic use, Fetal Hemoglobin analysis, Hydroxyurea therapeutic use
- Abstract
In the phase I/II pediatric hydroxyurea safety trial (HUG-KIDS), school-aged children with sickle cell anemia receiving hydroxyurea at the maximally tolerated dose (MTD) had variable increases in the percentage of fetal hemoglobin (%HbF). To identify predictors of the HbF response to hydroxyurea therapy, baseline clinical and laboratory values (age, sex, hemoglobin concentration, %HbF, reticulocytes, white blood cell [WBC], platelets, and serum chemistries), as well as treatment variables (number of toxicities, noncompliance, MTD dose, and MTD blood counts) were analyzed in 53 HUG-KIDS children who achieved MTD. Baseline %HbF values (P =.001), baseline hemoglobin concentration (P =.01), MTD dose (P =.02), and compliance (P =.02) were significantly associated with a higher %HbF at MTD; in contrast, age, sex, number of toxicities, and other baseline hematologic parameters were not. After adjusting for variations in baseline %HbF, the baseline reticulocyte count (P =.05) and baseline WBC count (P =.05) were also significantly associated with a higher %HbF at MTD. Hydroxyurea-induced increases in the hemoglobin concentration and mean corpuscular volume (both higher absolute values at MTD and larger positive changes from baseline values), as well as hydroxyurea-induced decreases in reticulocytes and WBC count, were significantly associated with a higher %HbF at MTD. These data suggest that selected baseline laboratory parameters, a higher MTD dose with attention to compliance, and greater therapy-related changes in blood counts may predict the HbF response to hydroxyurea therapy for children with sickle cell anemia. The HbF response to hydroxyurea is variable and complex, however, and even children with low baseline %HbF values can develop substantial increases in %HbF at MTD.
- Published
- 2002
- Full Text
- View/download PDF
39. Defective binding of factor XI-N248 to activated human platelets.
- Author
-
Sun MF, Baglia FA, Ho D, Martincic D, Ware RE, Walsh PN, and Gailani D
- Subjects
- Amino Acid Substitution, Cells, Cultured, Chromogenic Compounds, Factor IX metabolism, Factor XI drug effects, Factor XI Deficiency etiology, Factor XI Deficiency genetics, Fibroblasts, Humans, Kinetics, Protein Binding, Recombinant Proteins genetics, Recombinant Proteins metabolism, Thrombin pharmacology, Blood Platelets metabolism, Factor XI genetics, Factor XI metabolism
- Abstract
Variants of factor XI containing Gln226 to Arg (Q226 to R) and Ser248 to Asn (S248 to N) substitutions were first identified in an African American family with a history of excessive bleeding. The substitutions have recently been identified in unrelated individuals, suggesting they are relatively common. Both amino acids are located in the third apple domain of factor XI, an area implicated in binding interactions with factor IX and activated platelets. Recombinant factor XI-R226 and factor XI-N248 were compared with wild-type factor XI in assays for factor IX activation or platelet binding. Factor XI-R226 activates factor IX with a Michaelis-Menten constant (K(m)) about 5-fold greater than wild-type protein. The catalytic efficiency of factor IX activation is similar to wild-type protein, however, due to an increase in the turnover number (k(cat)) for the reaction. Iodinated factor XI-N248 binds to activated platelets with a dissociation constant (K(d)) more than 5-fold higher than wild-type protein (55 nM and 10 nM, respectively). Activation of factor XI-N248 by thrombin in the presence of activated platelets is slower and does not progress to the same extent as activation of the wild-type protein under similar conditions. Factor XI-N248 activates factor IX normally in a purified protein system and has relatively normal activity in activated partial thromboplastin time (aPTT) assays. Factor XI-N248 is the first factor XI variant described with a clear functional difference compared with wild-type protein. Importantly, the defect in platelet binding would not be detected by routine clinical evaluation with an aPTT assay.
- Published
- 2001
- Full Text
- View/download PDF
40. Efficient retrovirus-mediated PIG-A gene transfer and stable restoration of GPI-anchored protein expression in cells with the PNH phenotype.
- Author
-
Nishimura Ji, Phillips KL, Ware RE, Hall S, Wilson L, Gentry TL, Howard TA, Murakami Y, Shibano M, Machii T, Gilboa E, Kanakura Y, Takeda J, Kinoshita T, Rosse WF, and Smith CA
- Subjects
- 3T3 Cells, Animals, B-Lymphocytes metabolism, Bone Marrow Cells metabolism, Cell Line, Cell Line, Transformed, Gene Expression, Genetic Vectors, Hematopoietic Stem Cells metabolism, Hemoglobinuria, Paroxysmal blood, Hemoglobinuria, Paroxysmal metabolism, Hemolysis, Herpesvirus 4, Human, Membrane Proteins deficiency, Membrane Proteins physiology, Mice, Mutation, Nerve Growth Factor analysis, Nerve Growth Factor genetics, Phenotype, Glycosylphosphatidylinositols genetics, Hemoglobinuria, Paroxysmal genetics, Membrane Proteins genetics, Retroviridae genetics, Transfection
- Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a clonal hematopoietic stem cell disorder characterized by complement-mediated hemolysis due to deficiencies of glycosylphosphatidylinositol-anchored proteins (GPI-APs) in subpopulations of blood cells. Acquired mutations in the X-linked phosphatidylinositol glycan-class A (PIG-A) gene appear to be the characteristic and pathogenetic cause of PNH. To develop a gene therapy approach for PNH, a retroviral vector construct, termed MPIN, was made containing the PIG-A complementary DNA along with an internal ribosome entry site and the nerve growth factor receptor (NGFR) as a selectable marker. MPIN transduction led to efficient and stable PIG-A and NGFR gene expression in a PIG-A-deficient B-cell line (JY5), a PIG-A-deficient K562 cell line, an Epstein-Barr virus-transformed B-cell line (TK-14(-)) established from a patient with PNH, as well as peripheral blood (PB) mononuclear cells from a patient with PNH. PIG-A expression in these cell lines stably restored GPI-AP expression. MPIN was transduced into bone marrow mononuclear cells from a patient with PNH, and myeloid/erythroid colonies and erythroid cells were derived. These transduced erythroid cells restored surface expression of GPI-APs and resistance to hemolysis. These results indicate that MPIN is capable of efficient and stable functional restoration of GPI-APs in a variety of PIG-A-deficient hematopoietic cell types. Furthermore, MPIN also transduced into PB CD34(+) cells from a normal donor, indicating that MPIN can transduce primitive human progenitors. These findings set the stage for determining whether MPIN can restore PIG-A function in multipotential stem cells, thereby providing a potential new therapeutic option in PNH.
- Published
- 2001
- Full Text
- View/download PDF
41. Recombinant human acid alpha-glucosidase enzyme therapy for infantile glycogen storage disease type II: results of a phase I/II clinical trial.
- Author
-
Amalfitano A, Bengur AR, Morse RP, Majure JM, Case LE, Veerling DL, Mackey J, Kishnani P, Smith W, McVie-Wylie A, Sullivan JA, Hoganson GE, Phillips JA 3rd, Schaefer GB, Charrow J, Ware RE, Bossen EH, and Chen YT
- Subjects
- Age Factors, Animals, Blotting, Western, CHO Cells, Cricetinae, Enzyme-Linked Immunosorbent Assay, Glycogen metabolism, Heart physiology, Heart Diseases genetics, Heart Diseases prevention & control, Humans, Infant, Muscle, Skeletal metabolism, Muscle, Skeletal physiology, Muscles pathology, Myocardium metabolism, Phenotype, Radiography, Thoracic, Time Factors, X-Rays, Glycogen Storage Disease Type II therapy, Recombinant Proteins therapeutic use, alpha-Glucosidases therapeutic use
- Abstract
Purpose: Infantile glycogen storage disease type II (GSD-II) is a fatal genetic muscle disorder caused by deficiency of acid alpha-glucosidase (GAA). The purpose of this study was to investigate the safety and efficacy of recombinant human GAA (rhGAA) enzyme therapy for this fatal disorder., Methods: The study was designed as a phase I/II, open-label, single-dose study of rhGAA infused intravenously twice weekly in three infants with infantile GSD-II. rhGAA used in this study was purified from genetically engineered Chinese hamster ovary (CHO) cells overproducing GAA. Adverse effects and efficacy of rhGAA upon cardiac, pulmonary, neurologic, and motor functions were evaluated during 1 year of the trial period. The primary end point assessed was heart failure-free survival at 1 year of age. This was based on historical control data that virtually all patients died of cardiac failure by 1 year of age., Results: The results of more than 250 infusions showed that rhGAA was generally well tolerated. Steady decreases in heart size and maintenance of normal cardiac function for more than 1 year were observed in all three infants. These infants have well passed the critical age of 1 year (currently 16, 18, and 22 months old) and continue to have normal cardiac function. Improvements of skeletal muscle functions were also noted; one patient showed marked improvement and currently has normal muscle tone and strength as well as normal neurologic and Denver developmental evaluations. Muscle biopsies confirmed that dramatic reductions in glycogen accumulation had occurred after rhGAA treatment in this patient., Conclusions: This phase I/II first study of recombinant human GAA derived from CHO cells showed that rhGAA is capable of improving cardiac and skeletal muscle functions in infantile GSD-II patients. Further study will be needed to assess the overall potential of this therapy.
- Published
- 2001
- Full Text
- View/download PDF
42. Acquired DNA mutations associated with in vivo hydroxyurea exposure.
- Author
-
Hanft VN, Fruchtman SR, Pickens CV, Rosse WF, Howard TA, and Ware RE
- Subjects
- Adult, Anemia, Sickle Cell blood, Anemia, Sickle Cell genetics, Antineoplastic Agents adverse effects, Child, DNA drug effects, Humans, Hypoxanthine Phosphoribosyltransferase blood, Leukocytes, Mononuclear drug effects, Myeloproliferative Disorders blood, Myeloproliferative Disorders genetics, Recombination, Genetic drug effects, Anemia, Sickle Cell drug therapy, Antisickling Agents adverse effects, DNA genetics, Hydroxyurea adverse effects, Hypoxanthine Phosphoribosyltransferase genetics, Mutagenesis, Mutagens, Myeloproliferative Disorders drug therapy
- Abstract
Hydroxyurea (HU) is an effective therapeutic agent for patients with myeloproliferative disorders (MPDs) or sickle cell disease (SCD). Short-term HU toxicities primarily include transient myelosuppression, but long-term HU risks have not been defined. The mutagenic and carcinogenic potential of HU is not established, although HU has been associated with an increased risk of leukemia in some patients with MPD. In this study, 2 assays were used to quantitate acquired somatic DNA mutations in peripheral blood mononuclear cells (PBMCs) after in vivo HU exposure. The HPRT assay measures hypoxanthine phosphoribosyl transferase (hprt) mutations, while the VDJ assay identifies "illegitimate" T-cell receptor Vgamma-Jbeta interlocus recombination events. PBMCs were analyzed from patients with MPD, adults and children with SCD, and normal controls. MPD patients with prolonged HU exposure had numbers of DNA mutations equivalent to patients with low HU exposure or controls. Similarly, adults with SCD had equivalent numbers of DNA mutations regardless of HU exposure. Children with SCD and 30-month HU exposure had equivalent hprt(-) mutations but significantly more VDJ mutations (1.82 +/- 1.20 events per microg DNA) than children with 7-month HU exposure (1.58 +/- 0.87 events) or no HU exposure (1.06 +/- 0.45 events), P =.04 by analysis of variance. Taken together, these data suggest that the mutagenic and carcinogenic potential of in vivo HU therapy is low. Although increased numbers of illegitimate VDJ recombination events do not directly portend leukemia, young patients with SCD and HU exposure should be monitored serially for increases in DNA mutations.
- Published
- 2000
43. Hydroxyurea as an alternative to blood transfusions for the prevention of recurrent stroke in children with sickle cell disease.
- Author
-
Ware RE, Zimmerman SA, and Schultz WH
- Subjects
- Administration, Oral, Adolescent, Adult, Child, Child, Preschool, Humans, Infant, Prospective Studies, Stroke etiology, Treatment Outcome, Anemia, Sickle Cell complications, Antisickling Agents administration & dosage, Blood Transfusion, Hydroxyurea administration & dosage, Stroke prevention & control
- Abstract
Children with sickle cell disease (SCD) and stroke receive chronic transfusions to prevent stroke recurrence. Transfusion risks including infection, erythrocyte allosensitization, and iron overload suggest a need for alternative therapies. We previously used hydroxyurea (HU) and phlebotomy in two young adults with SCD and stroke as an alternative to transfusions. We have now prospectively discontinued transfusions in 16 pediatric patients with SCD and stroke. Reasons to discontinue transfusions included erythrocyte alloantibodies or autoantibodies, recurrent stroke on transfusions, iron overload, noncompliance, and deferoxamine allergy. HU was started at 15 mg/kg/d and escalated to 30 mg/kg/d based on hematologic toxicity. Patients with iron overload underwent phlebotomy. The children have been off transfusions 22 months, (range, 3 to 52 months). Their average HU dose is 24.9 +/- 4.2 mg/kg/d, hemoglobin concentration is 9.4 +/- 1.3 g/dL, and mean corpuscular volume (MCV) is 112 +/- 9 fL. Maximum percentage fetal hemoglobin (%HbF) is 20.6% +/- 8.0% and percentage HbF-containing erythrocytes (%F cells) is 79.3% +/- 14.7%. Fourteen patients underwent phlebotomy with an average of 8,993 mL (267 mL/kg) removed. Serum ferritin has decreased from 2,630 to 424 ng/mL, and 4 children have normal ferritin values. Three patients (19%) had neurological events considered recurrent stroke, each 3 to 4 months after discontinuing transfusions, but before maximal HU effects. These preliminary data suggest some children with SCD and stroke may discontinue chronic transfusions and use HU therapy to prevent stroke recurrence. Phlebotomy is well-tolerated and significantly reduces iron overload. Modifications in HU therapy to raise HbF more rapidly might increase protection against stroke recurrence.
- Published
- 1999
44. Safety of hydroxyurea in children with sickle cell anemia: results of the HUG-KIDS study, a phase I/II trial. Pediatric Hydroxyurea Group.
- Author
-
Kinney TR, Helms RW, O'Branski EE, Ohene-Frempong K, Wang W, Daeschner C, Vichinsky E, Redding-Lallinger R, Gee B, Platt OS, and Ware RE
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Drug Monitoring, Female, Humans, Male, Treatment Outcome, Anemia, Sickle Cell drug therapy, Antisickling Agents administration & dosage, Antisickling Agents adverse effects, Hydroxyurea administration & dosage, Hydroxyurea adverse effects
- Abstract
Previous studies have determined the short-term toxicity profile, laboratory changes, and clinical efficacy associated with hydroxyurea (HU) therapy in adults with sickle cell anemia. The safety and efficacy of this agent in pediatric patients with sickle cell anemia has not been determined. Children with sickle cell anemia, age 5 to 15 years, were eligible for this multicenter Phase I/II trial. HU was started at 15 mg/kg/d and escalated to 30 mg/kg/d unless the patient experienced laboratory toxicity. Patients were monitored by 2-week visits to assess compliance, toxicity, clinical adverse events, growth parameters, and laboratory efficacy associated with HU treatment. Eighty-four children were enrolled between December 1994 and March 1996. Sixty-eight children reached maximum tolerated dose (MTD) and 52 were treated at MTD for 1 year. Significant hematologic changes included increases in hemoglobin concentration, mean corpuscular volume, mean corpuscular hemoglobin, and fetal hemoglobin parameters, and decreases in white blood cell, neutrophil, platelet, and reticulocyte counts. Laboratory toxicities typically were mild, transient, and were reversible upon temporary discontinuation of HU. No life-threatening clinical adverse events occurred and no child experienced growth failure. This Phase I/II trial shows that HU therapy is safe for children with sickle cell anemia when treatment was directed by a pediatric hematologist. HU in children induces similar laboratory changes as in adults. Phase III trials to determine if HU can prevent chronic organ damage in children with sickle cell anemia are warranted.
- Published
- 1999
45. Identification of mutations and polymorphisms in the factor XI genes of an African American family by dideoxyfingerprinting.
- Author
-
Martincic D, Zimmerman SA, Ware RE, Sun MF, Whitlock JA, and Gailani D
- Subjects
- Adult, Alleles, Amino Acid Substitution, Cells, Cultured, Child, DNA Fingerprinting, DNA Mutational Analysis, Dideoxynucleosides, Dimerization, Exons genetics, Factor XI Deficiency ethnology, Female, Genetic Predisposition to Disease, Hemorrhagic Disorders etiology, Heterozygote, Humans, Male, Polymorphism, Single-Stranded Conformational, Recombinant Fusion Proteins metabolism, Black or African American, Black People genetics, Factor XI genetics, Factor XI Deficiency genetics, Mutation, Missense
- Abstract
Congenital deficiency of factor XI is a rare condition associated with a mild to moderate bleeding diathesis that is most commonly found in persons of Jewish ancestry. The disorder has been reported sporadically in a number of other ethnic groups, but rarely in the black population. We report on the genetic analysis of the factor XI genes of two African American patients: a 9-year-old boy (the propositus) with mild factor XI deficiency and his mother. Both individuals have lifelong histories of excessive bleeding. Dideoxyfingerprinting, a technique combining components of single-strand conformational polymorphism analysis and dideoxy-chain termination sequencing, was used in the analysis. Both patients were found to be heterozygous for a mutation changing serine 248 to asparagine [corrected], whereas the propositus was heterozygous for an additional mutation on the paternal allele changing glutamine 226 to arginine. Both mutations reside in the third apple domain of the factor XI heavy chain, an area that has been shown to contain binding sites for factor IX, platelets, and glycosaminoglycans. Previously reported mutations in the factor XI gene seem to cause deficiency primarily by reducing protein expression. Because both alleles in the propositus contain amino acid substitutions, the significant amount of circulating factor XI in his plasma must be comprised entirely of abnormal molecules. Factor XI circulates as a homodimer, and the presence of mutations in both alleles of the factor XI gene suggests that his bleeding disorder is caused in part by the effect of the two abnormal gene products forming dimers in different combinations. Three neutral (not associated with amino acid changes) DNA polymorphisms were also identified in the two subjects: a C to T change at nucleotide 472 in exon 5, A to G at nucleotide 844 in exon 8, and T to C at nucleotide 1234 in exon 11. Analysis of a random sample of normal volunteers showed that these polymorphisms are relatively common, with allele frequencies of 7.4%, 19%, and 18%, respectively. This suggests that there is considerable genetic heterogeneity in the factor XI gene., (Copyright 1998 by The American Society of Hematology)
- Published
- 1998
46. The PIG-A mutation and absence of glycosylphosphatidylinositol-linked proteins do not confer resistance to apoptosis in paroxysmal nocturnal hemoglobinuria.
- Author
-
Ware RE, Nishimura J, Moody MA, Smith C, Rosse WF, and Howard TA
- Subjects
- Adolescent, Adult, Aged, Anemia, Aplastic blood, Antibodies, Monoclonal pharmacology, Apoptosis radiation effects, B-Lymphocytes radiation effects, Cell Line, Clone Cells pathology, Culture Media, Serum-Free pharmacology, DNA, Complementary genetics, Erythrocytes pathology, Female, Gamma Rays, Granulocytes chemistry, Hematopoietic Stem Cells pathology, Hemoglobinuria, Paroxysmal pathology, Humans, Male, Membrane Proteins deficiency, Membrane Proteins physiology, Middle Aged, Recombinant Fusion Proteins physiology, Selection, Genetic, Transfection, X Chromosome genetics, fas Receptor immunology, Apoptosis genetics, B-Lymphocytes metabolism, Glycosylphosphatidylinositols deficiency, Granulocytes pathology, Hemoglobinuria, Paroxysmal genetics, Membrane Proteins genetics
- Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a clonal stem cell disorder characterized by complement-mediated hemolysis and deficient hematopoiesis. The development of PNH involves an acquired mutation in the X-linked PIG-A gene, which leads to incomplete bioassembly of glycosylphosphatidylinositol (GPI) anchors and absent or reduced surface expression of GPI-linked proteins. The origin and mechanisms by which the PNH clone becomes dominant are not well understood, but recently resistance to apoptosis has been postulated. To test the hypothesis that the PIG-A mutation and absence of GPI-linked surface proteins directly confer resistance to apoptosis, we isolated peripheral granulocytes from 26 patients with PNH and 20 normal controls and measured apoptosis induced by serum starvation. Granulocytes from patients with PNH were relatively resistant to apoptosis (38.8% +/- 14.1%) as compared with granulocytes from controls (55.0% +/- 12.0%, P < .001). However, this resistance to apoptosis was not related to the dominance of the PNH clone because patients with a low percentage of GPI-deficient granulocytes had a similar rate of apoptosis as those with a high percentage of GPI-deficient granulocytes. Similarly, the resistance to granulocyte apoptosis was not influenced by the degree of neutropenia or a prior history of aplastic anemia. To investigate formally the importance of GPI-linked surface proteins in apoptosis, we introduced the PIG-A cDNA sequence into the JY5 GPI-negative B-lymphoblastoid cell line using two different methods: (1) stable transfection of a plasmid containing PIG-A, and (2) stable transduction of a retroviral vector containing PIG-A. We then measured rates of apoptosis induced either by Fas antibody, serum starvation, or gamma-irradiation. With each stimulus, apoptosis of JY5 with stable surface expression of GPI-linked proteins was not statistically different from the parent JY5 cell line or the JY25 (GPI-positive) cell line. Our data confirm that granulocytes from patients with PNH have a relative resistance to apoptosis as compared with normal granulocytes. However, this resistance does not vary with the level of expression of GPI-linked proteins, and stable introduction of PIG-A cDNA with correction of GPI-linked surface expression does not change the rate of apoptosis. Taken together, our data do not support the hypothesis that the PIG-A mutation and absence of GPI-linked surface proteins directly confer resistance to apoptosis in PNH. We conclude that the resistance to apoptosis in PNH is not related to the PIG-A mutation, indicating that other factors must be important in the origin of this phenomenon and the clonal dominance observed in PNH.
- Published
- 1998
47. Molecular basis of the heterogeneity of expression of glycosyl phosphatidylinositol anchored proteins in paroxysmal nocturnal hemoglobinuria.
- Author
-
Endo M, Ware RE, Vreeke TM, Singh SP, Howard TA, Tomita A, Holguin MH, and Parker CJ
- Subjects
- Adult, Anemia, Aplastic complications, Base Sequence, Clone Cells metabolism, DNA, Complementary genetics, Dosage Compensation, Genetic, Female, Hemoglobinuria, Paroxysmal complications, Hemoglobinuria, Paroxysmal metabolism, Humans, Membrane Proteins physiology, Molecular Sequence Data, Phenotype, Glycosylphosphatidylinositols biosynthesis, Hemoglobinuria, Paroxysmal genetics, Membrane Proteins genetics, Mosaicism, Mutation, T-Lymphocytes metabolism
- Abstract
The purpose of these studies was to determine the molecular basis of the phenotypic mosaicism that is a defining feature of paroxysmal nocturnal hemoglobinuria (PNH). Analysis of T cell clones from a female patient revealed four distinct phenotypes based on surface expression of glycosyl phosphatidylinositol-anchored proteins (GPI-AP). When PIG-A (the gene that is mutant in PNH) from these clones was analyzed, four discrete somatic mutations were identified. Analysis of X chromosomal inactivation among the abnormal T cell clones was consistent with polyclonality. Together, these studies demonstrate that the phenotypic mosaicism that is characteristic of PNH is a consequence of genotypic mosaicism and that, at least in this case, PNH is a polyclonal rather than a monoclonal disease. That four distinct somatic mutations were present in a single patient suggests that in conditions that predispose to PNH PIG-A may be hypermutable.
- Published
- 1996
48. The molecular basis of paroxysmal nocturnal hemoglobinuria.
- Author
-
Rosse WF and Ware RE
- Subjects
- Acetylglucosamine metabolism, Animals, CD55 Antigens metabolism, Carbohydrate Sequence, Complement Activation, Endoplasmic Reticulum metabolism, Glycosylphosphatidylinositols physiology, Hematopoiesis, Hemoglobinuria, Paroxysmal metabolism, Humans, Lymphocytes metabolism, Membrane Proteins deficiency, Membrane Proteins metabolism, Membrane Proteins physiology, Mice, Mice, Mutant Strains, Models, Biological, Molecular Sequence Data, Mutation, Phosphatidylinositol Diacylglycerol-Lyase, Phosphatidylinositols metabolism, Phosphoric Diester Hydrolases metabolism, RNA Splicing, RNA, Messenger genetics, RNA, Messenger metabolism, X Chromosome, Glycosylphosphatidylinositols deficiency, Hemoglobinuria, Paroxysmal genetics, Membrane Proteins genetics, Protein Processing, Post-Translational
- Published
- 1995
49. Immunophenotypic analysis of reticulocytes in paroxysmal nocturnal hemoglobinuria.
- Author
-
Ware RE, Rosse WF, and Hall SE
- Subjects
- Adolescent, Adult, Antigens, CD metabolism, CD59 Antigens, Female, Glycosylphosphatidylinositols metabolism, Hemoglobinuria, Paroxysmal pathology, Humans, Immunophenotyping, Male, Membrane Glycoproteins metabolism, Middle Aged, Hemoglobinuria, Paroxysmal immunology, Reticulocytes immunology
- Abstract
The hematologic disorder paroxysmal nocturnal hemoglobinuria (PNH) occurs following an acquired somatic mutation in the Piga gene within a bone marrow stem cell. The progeny of this mutated cell cannot synthesize glycosylphosphatidylinositol (GPI) anchors, with a resultant deficiency in surface expression of all GPI-linked proteins. The protean clinical manifestations of PNH presumably result from the deficiency of these GPI-linked surface proteins. To explain the observation that neutrophils are affected at a significantly higher percentage than circulating erythrocytes and to analyze the proliferative rates of erythroid production in PNH, we studied 25 patients using flow cytometry. The fluorescent dye thiazole orange was used to detect reticulocytes, and CD59 monoclonal antibody was used to identify GPI-deficient cells. In contrast to the mature circulating erythrocytes, the percentage of abnormal reticulocytes was similar to the percentage of affected neutrophils. However, the vast majority of reticulocytes was completely GPI-deficient, ie, were type III cells, even in patients with only modest numbers of circulating type III erythrocytes. In addition, greater than 5% type II reticulocytes were identified in only 3 patients, although greater than 5% type II mature erythrocytes were identified in 10 of 25 patients. The results show that the erythroid and neutrophil bone marrow precursors have an equivalent proliferative advantage in PNH. The data also have important implications for the origin of type-II erythrocytes in PNH.
- Published
- 1995
50. Chromosomal assignment of genes involved in glycosylphosphatidylinositol anchor biosynthesis: implications for the pathogenesis of paroxysmal nocturnal hemoglobinuria.
- Author
-
Ware RE, Howard TA, Kamitani T, Chang H-M, Yeh ETH, and Seldin MF
- Subjects
- Animals, Base Sequence, Crossing Over, Genetic, Humans, Meiosis, Mice, Mice, Inbred C3H, Molecular Sequence Data, Chromosome Mapping, Glycosylphosphatidylinositols biosynthesis, Hemoglobinuria, Paroxysmal etiology
- Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematologic disorder that affects both sexes equally. The biochemical defect in PNH resides in the incomplete enzymatic assembly of glycosylphosphatidylinositol (GPI) anchors used for surface protein attachment. In all PNH patients tested to date, the biosynthetic defect occurs at the addition of N-acetyl-glucosamine to the phosphatidylinositol molecule (class A defect). A human cDNA, Piga, that repairs cell lines with the class A GPI-anchor biosynthetic defect has been recently cloned. Mapping of Piga to the X chromosome suggests that a single acquired mutation within Piga could alter GPI-anchor synthesis and result in PNH. However, this finding does not explain why all PNH patients have the class A defect. In the current study, the chromosomal assignment of Piga, as well as of Pigf and Pigh, two additional genes involved in GPI-anchor biosynthesis, has been established using a mouse interspecific backcross mapping technique. In contrast to Piga, both human and mouse Pigf and Pigh genes map to autosomes. The location of Pigf and Pigh suggests that mutations on both alleles of these autosomal genes would be necessary to produce PNH. This helps to explain the predominant class A defect in PNH.
- Published
- 1994
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.