Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sharada A. Sarnaik is active.

Publication


Featured researches published by Sharada A. Sarnaik.


The Lancet | 2011

Hydroxycarbamide in very young children with sickle-cell anaemia: a multicentre, randomised, controlled trial (BABY HUG)

Winfred C. Wang; Russell E. Ware; Scott T. Miller; Rathi V. Iyer; James F. Casella; Caterina P. Minniti; Sohail Rana; Courtney D. Thornburg; Zora R. Rogers; Ram Kalpatthi; Julio C. Barredo; R. Clark Brown; Sharada A. Sarnaik; Thomas H. Howard; Lynn W. Wynn; Abdullah Kutlar; F. Daniel Armstrong; Beatrice Files; Jonathan C. Goldsmith; Myron A. Waclawiw; Xiangke Huang; Bruce W. Thompson

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.


The New England Journal of Medicine | 2014

Controlled Trial of Transfusions for Silent Cerebral Infarcts in Sickle Cell Anemia

Michael R. DeBaun; Mae O. Gordon; Robert C. McKinstry; Michael J. Noetzel; Desirée A. White; Sharada A. Sarnaik; Emily Riehm Meier; Thomas H. Howard; Suvankar Majumdar; Baba Inusa; Paul Telfer; Melanie Kirby-Allen; Timothy L. McCavit; Annie Kamdem; Gladstone Airewele; Gerald M. Woods; Brian Berman; Julie A. Panepinto; Beng Fuh; Janet L. Kwiatkowski; Allison King; Jason Fixler; Melissa Rhodes; Alexis A. Thompson; Mark E. Heiny; Rupa Redding-Lallinger; Fenella J. Kirkham; Natalia Dixon; Corina E. Gonzalez; Karen Kalinyak

BACKGROUND Silent cerebral infarcts are the most common neurologic injury in children with sickle cell anemia and are associated with the recurrence of an infarct (stroke or silent cerebral infarct). We tested the hypothesis that the incidence of the recurrence of an infarct would be lower among children who underwent regular blood-transfusion therapy than among those who received standard care. METHODS In this randomized, single-blind clinical trial, we randomly assigned children with sickle cell anemia to receive regular blood transfusions (transfusion group) or standard care (observation group). Participants were between 5 and 15 years of age, with no history of stroke and with one or more silent cerebral infarcts on magnetic resonance imaging and a neurologic examination showing no abnormalities corresponding to these lesions. The primary end point was the recurrence of an infarct, defined as a stroke or a new or enlarged silent cerebral infarct. RESULTS A total of 196 children (mean age, 10 years) were randomly assigned to the observation or transfusion group and were followed for a median of 3 years. In the transfusion group, 6 of 99 children (6%) had an end-point event (1 had a stroke, and 5 had new or enlarged silent cerebral infarcts). In the observation group, 14 of 97 children (14%) had an end-point event (7 had strokes, and 7 had new or enlarged silent cerebral infarcts). The incidence of the primary end point in the transfusion and observation groups was 2.0 and 4.8 events, respectively, per 100 years at risk, corresponding to an incidence rate ratio of 0.41 (95% confidence interval, 0.12 to 0.99; P=0.04). CONCLUSIONS Regular blood-transfusion therapy significantly reduced the incidence of the recurrence of cerebral infarct in children with sickle cell anemia. (Funded by the National Institute of Neurological Disorders and Stroke and others; Silent Cerebral Infarct Multi-Center Clinical Trial ClinicalTrials.gov number, NCT00072761, and Current Controlled Trials number, ISRCTN52713285.).


Blood | 2011

Silent cerebral infarcts occur despite regular blood transfusion therapy after first strokes in children with sickle cell disease

Monica L. Hulbert; Robert C. McKinstry; JoAnne L. Lacey; Christopher J. Moran; Julie A. Panepinto; Alexis A. Thompson; Sharada A. Sarnaik; Gerald M. Woods; James F. Casella; Baba Inusa; Jo Howard; Fenella J. Kirkham; Kofi A. Anie; Jonathan E. Mullin; Rebecca Ichord; Michael J. Noetzel; Yan Yan; Mark Rodeghier; Michael R. DeBaun

Children with sickle cell disease (SCD) and strokes receive blood transfusion therapy for secondary stroke prevention; despite this, approximately 20% experience second overt strokes. Given this rate of second overt strokes and the clinical significance of silent cerebral infarcts, we tested the hypothesis that silent cerebral infarcts occur among children with SCD being transfused for secondary stroke prevention. A prospective cohort enrolled children with SCD and overt strokes at 7 academic centers. Magnetic resonance imaging and magnetic resonance angiography of the brain were scheduled approximately every 1 to 2 years; studies were reviewed by a panel of neuroradiologists. Eligibility criteria included regularly scheduled blood transfusion therapy. Forty children were included; mean pretransfusion hemoglobin S concentration was 29%. Progressive cerebral infarcts occurred in 45% (18 of 40 children) while receiving chronic blood transfusion therapy; 7 had second overt strokes and 11 had new silent cerebral infarcts. Worsening cerebral vasculopathy was associated with new cerebral infarction (overt or silent; relative risk = 12.7; 95% confidence interval, 2.65-60.5, P = .001). Children with SCD and overt strokes receiving regular blood transfusion therapy experience silent cerebral infarcts at a higher rate than previously recognized. Additional therapies are needed for secondary stroke prevention in children with SCD.


The Lancet | 2016

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

Russell E. Ware; Barry R. Davis; William H. Schultz; R. Clark Brown; Banu Aygun; Sharada A. Sarnaik; Isaac Odame; Beng Fuh; Alex George; William Owen; Lori Luchtman-Jones; Zora R. Rogers; Lee Hilliard; Cynthia Gauger; Connie M. Piccone; Margaret T. Lee; Janet L. Kwiatkowski; Sherron M. Jackson; Scott T. Miller; Carla W. Roberts; Matthew M. Heeney; Theodosia A. Kalfa; Stephen C. Nelson; Hamayun Imran; Kerri Nottage; Ofelia Alvarez; Melissa Rhodes; Alexis A. Thompson; Jennifer A. Rothman; Kathleen J. Helton

Background For children with sickle cell anaemia and elevated transcranial Doppler (TCD) flow velocities, regular blood transfusions effectively prevent primary stroke, but must be continued indefinitely. The efficacy of hydroxyurea in this setting is unknown. Methods TWiTCH was a multicentre Phase III randomised open label, non-inferiority trial comparing standard treatment (transfusions) to alternative treatment (hydroxyurea) in children with abnormal TCD velocities but no severe vasculopathy. Iron overload was managed with chelation (Standard Arm) and serial phlebotomy (Alternative Arm). The primary study endpoint was the 24-month TCD velocity calculated from a general linear mixed model, with non-inferiority margin = 15 cm/sec. Findings Among 121 randomised participants (61 transfusions, 60 hydroxyurea), children on transfusions maintained <30% sickle haemoglobin, while those taking hydroxyurea (mean 27 mg/kg/day) averaged 25% fetal haemoglobin. The first scheduled interim analysis demonstrated non-inferiority, and the sponsor terminated the study. Final model-based TCD velocities (mean ± standard error) on Standard versus Alternative Arm were 143 ± 1.6 and 138 ± 1.6 cm/sec, respectively, with difference (95% CI) = 4.54 (0.10, 8.98), non-inferiority p=8.82 × 10−16 and post-hoc superiority p=0.023. Among 29 new neurological events adjudicated centrally by masked reviewers, no strokes occurred but there were 3 transient ischaemic attacks per arm. Exit brain MRI/MRA revealed no new cerebral infarcts in either arm, but worse vasculopathy in one participant (Standard Arm). Iron burden decreased more in the Alternative Arm, with ferritin difference −1047 ng/mL (−1524, −570), p<0.001 and liver iron difference −4.3 mg Fe/gm dry weight (−6.1, −2.5), p=0.001. Interpretation For high-risk children with sickle cell anaemia and abnormal TCD velocities, after four years of transfusions and without severe MRA vasculopathy, hydroxyurea therapy can substitute for chronic transfusions to maintain TCD velocities and help prevent primary stroke.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 participants 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.


Blood | 2012

Associated risk factors for silent cerebral infarcts in sickle cell anemia: low baseline hemoglobin, sex, and relative high systolic blood pressure

Michael R. DeBaun; Sharada A. Sarnaik; Mark Rodeghier; Caterina P. Minniti; Thomas H. Howard; Rathi V. Iyer; Baba Inusa; Paul Telfer; Melanie Kirby-Allen; Charles T. Quinn; Françoise Bernaudin; Gladstone Airewele; Gerald M. Woods; Julie A. Panepinto; Beng Fuh; Janet K. Kwiatkowski; Allison King; Melissa Rhodes; Alexis A. Thompson; Mark E. Heiny; Rupa Redding-Lallinger; Fenella J. Kirkham; Hernan Sabio; Corina E. Gonzalez; Suzanne Saccente; Karen Kalinyak; John J. Strouse; Jason Fixler; Mae O. Gordon; J. Phillip Miller

The most common form of neurologic injury in sickle cell anemia (SCA) is silent cerebral infarction (SCI). In the Silent Cerebral Infarct Multi-Center Clinical Trial, we sought to identify risk factors associated with SCI. In this cross-sectional study, we evaluated the clinical history and baseline laboratory values and performed magnetic resonance imaging of the brain in participants with SCA (HbSS or HbSβ° thalassemia) between the ages of 5 and 15 years with no history of overt stroke or seizures. Neuroradiology and neurology committees adjudicated the presence of SCI. SCIs were diagnosed in 30.8% (251 of 814) participants who completed all evaluations and had valid data on all prespecified demographic and clinical covariates. The mean age of the participants was 9.1 years, with 413 males (50.7%). In a multivariable logistic regression analysis, lower baseline hemoglobin concentration (P < .001), higher baseline systolic blood pressure (P = .018), and male sex (P = .030) were statistically significantly associated with an increased risk of an SCI. Hemoglobin concentration and systolic blood pressure are risk factors for SCI in children with SCA and may be therapeutic targets for decreasing the risk of SCI. This study is registered at www.clinicaltrials.gov as #NCT00072761.


The Journal of Pediatrics | 2003

Invasive pneumococcal infections in children with sickle cell disease in the era of penicillin prophylaxis, antibiotic resistance, and 23-valent pneumococcal polysaccharide vaccination

Thomas V. Adamkiewicz; Sharada A. Sarnaik; George R. Buchanan; Rathi V. Iyer; Scott T. Miller; Charles H. Pegelow; Zora R. Rogers; Elliott Vichinsky; John A. Elliott; Richard R. Facklam; Katherine L. O'Brien; Benjamin Schwartz; Chris Van Beneden; Michael J. Cannon; James R. Eckman; Harry L. Keyserling; Kevin M. Sullivan; Wing Yen Wong; Winfred C. Wang

Rates and severity of pneumococcal infections in children with sickle cell disease were examined before licensure of pneumococcal-conjugated vaccine (PVC). Rates of peak invasive infection rates in 1-year-old children with hemoglobin SS and mortality in those 0 to 10 years of age were 36.5 to 63.4 and 1.4 to 2.8 per 1000 person-years, respectively (>10 and 100 times as frequent as in the general population). Overall, 71% of serotyped isolates (n=80) were PVC serotypes and 71% of nonvaccine serotype strains were penicillin-sensitive. Clinical presentation in children with hemoglobin SS (n=71; more with hypotension) and hemoglobin SC (n=18; more with acute chest syndrome, otitis media) differed. Penicillin nonsusceptibility (38% of isolates) varied between geographic study sites. Penicillin prophylaxis appeared less effective against intermediate and resistant strains. Of all infected children, meningitis developed in 20% and 15% died (hemoglobin SS, n=15 and 11; hemoglobin SC, n=1 each). Factors associated with death included age >4 years (58%), serotype 19F, and not being followed by a hematologist (42% each). The pneumococcal-polysaccharide vaccine was 80.4% effective within 3 years after vaccination (95% CI, 39.7, 93.6). Children with sickle cell disease of all ages may benefit from PVC boosted with polysaccharide vaccination.


The Journal of Pediatrics | 2010

Renal Function in Infants with Sickle Cell Anemia: Baseline Data from the BABY HUG Trial

Russell E. Ware; Renee C. Rees; Sharada A. Sarnaik; Rathi V. Iyer; Ofelia Alvarez; James F. Casella; Barry L. Shulkin; Eglal Shalaby-Rana; C. Frederic Strife; John H. Miller; Peter A. Lane; Winfred C. Wang; Scott T. Miller

OBJECTIVES To examine the feasibility and accuracy of glomerular filtration rate (GFR) measurements in infants with sickle cell anemia (SCA). STUDY DESIGN The NHLBI/NICHD-sponsored Phase III randomized double-blinded placebo-controlled trial (BABY HUG) tests the hypothesis that hydroxyurea can prevent chronic organ damage in SCA. GFR elevation is a coprimary endpoint, measured quantitatively by technetium 99m-labeled diethylenetriaminepentaacetic acid (DTPA) plasma clearance and estimated by the Schwartz equation with height and creatinine. RESULTS Baseline DTPA GFR measurement was attempted in 191 infants; 176 of 184 completed studies (96%) were interpretable. Average age (mean +/- 1SD) was 13.7 +/- 2.6 months. Average DTPA GFR was 125.2 +/- 34.4 (range 40.2-300.9, normal 91.5 +/- 17.8 mL/min/1.73m(2)), while Schwartz estimates were higher at 184.4 +/- 55.5 mL/min/1.73m(2). DTPA GFR was correlated with Schwartz GFR (r(2) = 0.0658, P = .0012); also with age, weight, height, and kidney volume (all P < .002); but not with hemoglobin, HbF, white blood cell count, reticulocytes, medical events, or splenic function. CONCLUSIONS Quantitative GFR measurement is feasible but variable among infants with SCA. Schwartz GFR estimates are not highly correlated with quantitative DTPA GFR values. Baseline GFR measurements suggest that renal dysfunction in SCA, evidenced by glomerular hyperfiltration, begins during infancy.


Journal of Pediatric Hematology Oncology | 1982

Neurological complications of sickle cell anemia

Sharada A. Sarnaik; Jeanne M. Lusher

A variety of neurological complications occur with sickle cell hemoglobinopathy. The most striking and most common is cerebral infarction. It is also the most devastating. Because of the high recurrence rate in untreated patients, cerebral infarction is the most debilitating neurological complication. The observed frequency varies from 6% to as high as 34% in different reports. The pathogenesis, clinical features, and demography are discussed. Diagnostic procedures should include a spinal tap to rule out potentially treatable lesions. CT scan and cerebral angiography may also be helpful in certain cases. Transfusion therapy remains the mainstay of management in the acute phase of cerebral infarction. There is considerable evidence to indicate that long-term transfusion programs are effective in the prevention of recurrences. Intracranial hemorrhage, spinal cord infarction, isolated neuropathies due to anatomical proximity to infarcted bones, lead neuropathy, auditory problems, and ocular manifestations are other neurological problems which can affect patients with sickle cell disease.


Pediatric Infectious Disease Journal | 1990

Studies on Pneumococcus vaccine alone or mixed with DTP and on Pneumococcus type 6B and Haemophilis influenzae type b capsular polysaccharide-tetanus toxoid conjugates in two- to five-year-old children with sickle cell anemia

Sharada A. Sarnaik; Joseph Kaplan; Gerald Schiffman; Dolores Bryla; John B. Robbins; Rachel Schneerson

Pneumococcus vaccine, injected alone or mixed with diphtheria-tetanus toxoid-pertussis, did not elicit significant concentrations of pneumococcus type 6 antibodies in 2− to 5-year-old sickle cell anemia patients (n = 22). Reinjection 5 months later failed to elicit a booster response to pneumococcus type 6. We then injected conjugates of pneumococcus type 6B and of Haemophilus influenzae type b (Hib), each bound to tetanus toxoid (TT), alternatively at monthly intervals into sickle cell anemia patients of the same age group (n = 25); most received 3 injections of each vaccine. Pneumococcus vaccine was administered to 19 patients and Hib to 1 at approximately 1 year of age. Blood samples were taken before each and approximately 6 months after the last injection. Infrequent and minimal local reactions and only 6 episodes of fever (3%) occurred after injection of the conjugates. Pneumococcus type 6B-TT elicited a rise in the geometric mean concentration of pneumococcus type 6 antibodies (Ab) from 104 ng of antibody nitrogen (AbN)/ml in preimmunization sera to 385 ng of AbN/ml after the first injection (P < 0.01). There were further increases after the 2 subsequent injections; 6 months after the third injection, the mean concentration was 940 ng of AbN/ml and 15 of 16 (94%) had > 300 ng of AbN/ml. Hib-TT elicited a 160-fold increase of Hib antibodies to a geometric mean concentration of 39.0 μg of Ab/ml after the first injection. These levels rose approximately 2-fold following 2 additional injections to 71.7 μg/ml and declined to 10.7 μg/ml at the 6-month sampling. All had > 0.15 μg of Hib Ab/ml (estimated protective level) after all injections. The patients received DTP during infancy and at 18 months of age; all had > 0.01 unit/ml of tetanus antitoxin (protective level) in their preimmunization sera. The mean antitoxin titer rose approximately 8-fold after the first injection (pnemococcus type 6B-TT), remained unchanged after the subsequent injections and declined to about 3-fold over the preimmunization levels at the 6-month blood sampling.


Pediatric Blood & Cancer | 2009

The pediatric hydroxyurea phase III clinical trial (BABY HUG): Challenges of study design

Bruce W. Thompson; Scott T. Miller; Zora R. Rogers; Renee C. Rees; Russell E. Ware; Myron A. Waclawiw; Rathi V. Iyer; James F. Casella; Lori Luchtman-Jones; Sohail Rana; Courtney D. Thornburg; Ram Kalpatthi; Julio C. Barredo; R. Clark Brown; Sharada A. Sarnaik; Thomas H. Howard; Lori Luck; Winfred C. Wang

Evidence of the laboratory benefits of hydroxyurea and its clinical efficacy in reducing acute vaso‐occlusive events in adults and children with sickle cell anemia has accumulated for more than 15 years. A definitive clinical trial showing that hydroxyurea can also prevent organ damage might support widespread use of the drug at an early age. BABY HUG is a randomized, double‐blind placebo‐controlled trial to test whether treating young children ages 9–17 months at entry with a liquid preparation of hydroxyurea (20 mg/kg/day for 2 years) can decrease organ damage in the kidneys and spleen by at least 50%. Creation of BABY HUG entailed unique challenges and opportunities. Although protection of brain function might be considered a more compelling endpoint, preservation of spleen and renal function has clinical relevance, and significant treatment effects might be discernable within the mandated sample size of 200. Concerns about unanticipated severe toxicity and burdensome testing and monitoring requirements were addressed in part by an internal Feasibility and Safety Pilot Study, the successful completion of which was required prior to enrolling a larger number of children on the protocol. Concerns over recruitment of potentially vulnerable subjects were allayed by inclusion of a research subject advocate, or ombudsman. Finally, maintenance of blinding of research personnel was aided by inclusion of an unblinded primary endpoint person, charged with transmitting endpoint data and monitoring blood work locally for toxicity (ClinicalTrials.gov number, NCT00006400). Pediatr Blood Cancer 2010;54:250–255.

Collaboration


Dive into the Sharada A. Sarnaik's collaboration.

Top Co-Authors

Avatar

Russell E. Ware

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Scott T. Miller

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Julie A. Panepinto

Children's Hospital of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Rathi V. Iyer

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar

Zora R. Rogers

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alexis A. Thompson

Children's Memorial Hospital

View shared research outputs
Top Co-Authors

Avatar

Banu Aygun

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

James F. Casella

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Janet L. Kwiatkowski

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

William H. Schultz

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge