Network


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

Hotspot


Dive into the research topics where Kathleen Neville is active.

Publication


Featured researches published by Kathleen Neville.


Pain | 2009

Three new datasets supporting use of the Numerical Rating Scale (NRS-11) for children's self-reports of pain intensity.

Carl L. von Baeyer; Lara J. Spagrud; Julia C. McCormick; Eugene Choo; Kathleen Neville; Mark Connelly

ABSTRACT Despite wide usage of the Numerical Rating Scale (NRS) for self‐report of pain intensity in clinical practice with children and adolescents, validation data are lacking. We present here three datasets from studies in which the NRS was used together with another self‐report scale. Study A compared post‐operative pain ratings on the NRS with scores on the Faces Pain Scale‐Revised (FPS‐R) in 69 children age 7–17 years who had undergone a variety of surgical procedures. Study B compared post‐operative pain ratings on the NRS with scores on the Visual Analogue Scale (VAS) in 29 children age 9–17 years who had undergone pectus excavatum repair. Study C compared ratings of remembered immunization pain in 236 children who comprised an NRS group and a sex‐ and age‐matched VAS group. Correlations of the NRS with the FPS‐R and VAS were r = 0.87 and 0.89 in Studies A and B, respectively. In Study C, the distributions of scores on the NRS and VAS were very similar except that scores closest to the no pain anchor were more likely to be selected on the VAS than the NRS. The NRS can be considered functionally equivalent to the VAS and FPS‐R except for very mild pain (<1/10). We conclude that use of the NRS is tentatively supported for clinical practice with children of 8 years and older, and we recommend further research on the lower age limit and on standardized age‐appropriate anchors and instructions for this scale.


Journal of Clinical Oncology | 2004

Phase I Study of the Proteasome Inhibitor Bortezomib in Pediatric Patients With Refractory Solid Tumors: A Children's Oncology Group Study (ADVL0015)

Susan M. Blaney; Mark Bernstein; Kathleen Neville; Jill P. Ginsberg; Brenda J. Kitchen; Terzah M. Horton; Stacey L. Berg; Mark Krailo; Peter C. Adamson

PURPOSE To determine the maximum-tolerated dose, dose-limiting toxicity (DLT), and pharmacodynamics of the proteasome inhibitor bortezomib (formerly PS-341) in children with recurrent or refractory solid tumors. PATIENTS AND METHODS An intravenous bolus of bortezomib was administered twice weekly for 2 consecutive weeks at either 1.2 or 1.6 mg/m2/dose followed by a 1-week rest. The pharmacodynamics of bortezomib were evaluated by measurement of whole blood 20S proteasome activity. RESULTS Fifteen patients, 11 assessable, were enrolled between November 2001 and February 2003. Dose-limiting thrombocytopenia, which prevented administration of a complete course (four doses in 2 weeks) of therapy, occurred in two of five assessable children enrolled at the 1.6 mg/m2 dose level. There were no other DLTs. Inhibition of 20S proteasome activity seemed to be dose dependent. The average inhibition 1 hour after drug administration on day 1 was 67.2% +/- 7.6% at the 1.2 mg/m2/dose and 76.5% +/- 3.3% at the 1.6 mg/m2/dose. There were no objective antitumor responses. CONCLUSION Bortezomib is well tolerated in children with recurrent or refractory solid tumors. The recommended phase II dose of bortezomib for children with solid tumors is 1.2 mg/m2/dose, administered as an intravenous bolus twice weekly for 2 weeks followed by a 1-week break.


Clinical Cancer Research | 2004

Plasma and Cerebrospinal Fluid Pharmacokinetics of Imatinib after Administration to Nonhuman Primates

Kathleen Neville; Robert A. Parise; Patrick A. Thompson; Alexander Aleksic; Merrill J. Egorin; Frank M. Balis; Leticia McGuffey; Cynthia McCully; Stacey L. Berg; Susan M. Blaney

Purpose: Imatinib mesylate (Gleevec, Glivec, STI571, imatinib) is a potent tyrosine kinase inhibitor approved for the treatment of chronic myelogenous leukemia and gastrointestinal stromal tumors. The role of imatinib in the treatment of malignant gliomas and other solid tumors is being evaluated. We used a nonhuman primate model that is highly predictive of the cerebrospinal fluid penetration of drugs in humans to study the pharmacokinetics of imatinib in plasma and cerebrospinal fluid (CSF) after i.v. and p.o. administration. Experimental Design: Imatinib, 15 mg/kg i.v. over 30 min (n = 3) or 30 mg/kg p.o. (n = 3), was administered to nonhuman primates. Imatinib was measured in serial samples of plasma and CSF using high-pressure liquid chromatography with UV absorbance or mass spectroscopic detection. Pharmacokinetic parameters were estimated using model-independent methods. Results: Peak plasma imatinib concentrations ranged from 6.4 to 9.5 μm after i.v. dosing and 0.8 to 2.8 μm after p.o. dosing. The mean ±SD area under the plasma concentration versus time curve was 2480 ±1340 μm·min and 1191 ±146 μm·min after i.v. and p.o. dosing, respectively. The terminal half-life was 529 ±167 min after i.v. dosing and 266 ±88 min after p.o. dosing. After i.v. dosing the steady state volume of distribution was 5.9 ±2.8 liter/kg, and the total body clearance was 12 ±5 ml/min/kg. The mean peak CSF concentration was 0.25 ±0.07 μm after i.v. dosing and 0.07 ±0.04 μm after p.o. dosing. The mean CSF:plasma area under the plasma concentration versus time curve ratio for all of the animals was 5% ±2%. Conclusions: There is limited penetration of imatinib into the CSF of nonhuman primates after i.v. and p.o. administration.


PLOS ONE | 2015

A Phase I Trial of DFMO Targeting Polyamine Addiction in Patients with Relapsed/Refractory Neuroblastoma

Giselle Saulnier Sholler; Eugene W. Gerner; Genevieve Bergendahl; Robert B. MacArthur; Alyssa VanderWerff; Takamaru Ashikaga; Jeffrey P. Bond; William Ferguson; William Roberts; Randal K. Wada; Don Eslin; Jacqueline M. Kraveka; Joel Kaplan; Deanna Mitchell; Nehal Parikh; Kathleen Neville; Leonard S. Sender; Timothy Higgins; Masao Kawakita; Kyoko Hiramatsu; Shun-suke Moriya; André S. Bachmann

Background Neuroblastoma (NB) is the most common cancer in infancy and most frequent cause of death from extracranial solid tumors in children. Ornithine decarboxylase (ODC) expression is an independent indicator of poor prognosis in NB patients. This study investigated safety, response, pharmacokinetics, genetic and metabolic factors associated with ODC in a clinical trial of the ODC inhibitor difluoromethylornithine (DFMO) ± etoposide for patients with relapsed or refractory NB. Methods and Findings Twenty-one patients participated in a phase I study of daily oral DFMO alone for three weeks, followed by additional three-week cycles of DFMO plus daily oral etoposide. No dose limiting toxicities (DLTs) were identified in patients taking doses of DFMO between 500-1500 mg/m2 orally twice a day. DFMO pharmacokinetics, single nucleotide polymorphisms (SNPs) in the ODC gene and urinary levels of substrates for the tissue polyamine exporter were measured. Urinary polyamine levels varied among patients at baseline. Patients with the minor T-allele at rs2302616 of the ODC gene had higher baseline levels (p=0.02) of, and larger decreases in, total urinary polyamines during the first cycle of DFMO therapy (p=0.003) and had median progression free survival (PFS) that was over three times longer, compared to patients with the major G allele at this locus although this last result was not statistically significant (p=0.07). Six of 18 evaluable patients were progression free during the trial period with three patients continuing progression free at 663, 1559 and 1573 days after initiating treatment. Median progression-free survival was less among patients having increased urinary polyamines, especially diacetylspermine, although this result was not statistically significant (p=0.056). Conclusions DFMO doses of 500-1500mg/m2/day are safe and well tolerated in children with relapsed NB. Children with the minor T allele at rs2302616 of the ODC gene with relapsed or refractory NB had higher levels of urinary polyamine markers and responded better to therapy containing DFMO, compared to those with the major G allele at this locus. These findings suggest that this patient subset may display dependence on polyamines and be uniquely susceptible to therapies targeting this pathway. Trial Registration Clinicaltrials.gov NCT#01059071


American Journal of Hematology | 2015

Health-related quality of life in children with sickle cell anemia: impact of blood transfusion therapy

Lauren M. Beverung; John J. Strouse; Monica L. Hulbert; Kathleen Neville; Robert I. Liem; Baba Inusa; Beng Fuh; Allison King; Emily Riehm Meier; James F. Casella; Michael R. DeBaun; Julie A. Panepinto

The completion of the Multicenter Silent Infarct Transfusion Trial demonstrated that children with pre‐existing silent cerebral infarct and sickle cell anemia (SCA) who received regular blood transfusion therapy had a 58% relative risk reduction of infarct recurrence when compared to observation. However, the total benefit of blood transfusion therapy, as assessed by the parents, was not measured against the burden of monthly blood transfusion therapy. In this planned ancillary study, we tested the hypothesis that a patient centered outcome, health‐related quality of life (HRQL), would be greater in participants randomly assigned to the blood transfusion therapy group than the observation group. A total of 89% (175 of 196) of the randomly allocated participants had evaluable entry and exit HRQL evaluations. The increase in Change in Health, measured as the childs health being better, was significantly greater for the transfusion group than the observation group (difference estimate = −0.54, P ≤ 0.001). This study provides the first evidence that children with SCA who received regular blood transfusion therapy felt better and had better overall HRQL than those who did not receive transfusion therapy. Am. J. Hematol. 90:139–143, 2015.


Pediatric Blood & Cancer | 2014

VKORC1 and CYP2C9 genotypes are predictors of warfarin‐related outcomes in children

Kaitlyn Shaw; Ursula Amstutz; Claudette Hildebrand; S. Rod Rassekh; Martin Hosking; Kathleen Neville; J. Steven Leeder; Michael R. Hayden; Colin Ross; Bruce Carleton

Despite substantial evidence supporting a pharmacogenetic approach to warfarin therapy in adults, evidence on the importance of genetics in warfarin therapy in children is limited, particularly for clinical outcomes. We assessed the contribution of CYP2C9/VKORC1/CYP4F2 genotypes and variation in other genes involved in vitamin K and coagulation pathways to warfarin dose and related clinical outcomes in children.


Genomics | 2010

Mutation in erythroid specific transcription factor KLF1 causes Hereditary Spherocytosis in the Nan hemolytic anemia mouse model

Daniel P. Heruth; Troy Hawkins; Derek P. Logsdon; Margaret Gibson; Inna Sokolovsky; Ndona N. Nsumu; Stephanie Major; Barbara Fegley; Gerald M. Woods; Karen Lewing; Kathleen Neville; Kenneth Cornetta; Kenneth R. Peterson; Robert A. White

KLF1 regulates definitive erythropoiesis of red blood cells by facilitating transcription through high affinity binding to CACCC elements within its erythroid specific target genes including those encoding erythrocyte membrane skeleton (EMS) proteins. Deficiencies of EMS proteins in humans lead to the hemolytic anemia Hereditary Spherocytosis (HS) which includes a subpopulation with no known genetic defect. Here we report that a mutation, E339D, in the second zinc finger domain of KLF1 is responsible for HS in the mouse model Nan. The causative nature of this mutation was verified with an allelic test cross between Nan/+ and heterozygous Klf1(+/-) knockout mice. Homology modeling predicted Nan KLF1 binds CACCC elements more tightly, suggesting that Nan KLF1 is a competitive inhibitor of wild-type KLF1. This is the first association of a KLF1 mutation with a disease state in adult mammals and also presents the possibility of being another causative gene for HS in humans.


Pediatric Blood & Cancer | 2015

Primary Stroke Prevention in Nigerian Children with Sickle Cell Disease (SPIN): Challenges of Conducting a Feasibility Trial

Najibah A. Galadanci; Shehu Umar Abdullahi; Musa A. Tabari; Shehi Abubakar; Raymond Belonwu; Auwal Salihu; Kathleen Neville; Fenella J. Kirkham; Baba Inusa; Yu Shyr; Sharon Phillips; Adetola A. Kassim; Lori C. Jordan; Muktar H. Aliyu; Brittany Covert; Michael R. DeBaun

The majority of children with sickle cell disease (SCD), approximately 75%, are born in sub‐Saharan Africa. For children with elevated transcranial Doppler (TCD) velocity, regular blood transfusion therapy for primary stroke prevention is standard care in high income countries, but is not feasible in sub‐Saharan Africa.


Pediatric Infectious Disease Journal | 2012

Defining Risk Factors for Red Man Syndrome in Children and Adults

Angela L. Myers; Andrea Gaedigk; Hongying Dai; Laura P. James; Bridgette L. Jones; Kathleen Neville

Background: Red man syndrome (RMS) is a well-known adverse reaction that occurs in pediatric patients receiving vancomycin, yet reported prevalence is varied, and characteristics and risk factors are not well understood. Our objective was to determine the prevalence, characteristics and risk factors for RMS in pediatric patients receiving vancomycin, including contributing genetic factors. Methods: A multicenter retrospective study of 546 subjects (0.5–21 years) who received at least 1 dose of intravenous vancomycin was conducted. Demographic and symptom data were collected through chart review and parent/nurse report. Genotype analysis included 10 single nucleotide polymorphisms in the histamine pathway. Results: RMS was observed in 77 (14%) subjects receiving vancomycin. Forty percent of subjects with RMS symptoms developed rash, pruritis and flushing, without hypotension. Antecedent antihistamine use was identified as a risk factor for RMS (P < 0.001). Multivariate regression analysis identified age > 2 years (P = 0.008), previous RMS (P < 0.001), vancomycin dose (P = 0.02) and vancomycin concentration (P = 0.017) as RMS risk factors, whereas African American race was protective (P = 0.011). We observed an apparent association between RMS and a single nucleotide polymorphism in the diamine oxidasegene (P = 0.044); however, no associations were revealed by multifactor dimensionality reduction analysis. Conclusions: RMS is a common adverse event in children receiving vancomycin. Identified risk factors are Caucasian ethnicity, age≥ 2 years, previous RMS history, vancomycin dose ≥ 10 mg/kg, vancomycin concentration ≥ 5 mg/mL and antecedent antihistamine use. Known genetic variants in histamine metabolism or receptors do not appear to be substantial contributors to risk of RMS.


Archives of Disease in Childhood | 2014

Pupillometry: a non-invasive technique for pain assessment in paediatric patients

Mark Connelly; Jacob T. Brown; Gregory L. Kearns; R. Anderson; Shawn D St Peter; Kathleen Neville

Objective Pupillometry has been used to assess pain intensity and response to analgesic medications in adults. The aim of this observational study was to explore proof of concept for the use of this technique in paediatric patients. Changes in pupil parameters before and after opioid exposure also were evaluated. Design and setting This was a single-centre, prospective study conducted at an academic paediatric medical centre. Patients Children 9–17 years of age undergoing elective surgical correction of pectus excavatum were enrolled into a protocol approved by the human ethical committee (institutional review board). Interventions Pupil size and reactivity were measured using a handheld pupillometer. Pain was assessed using age-appropriate, validated pain self-report scales. Results Thirty patients were enrolled. Each point change on a 10 cm visual analogue pain intensity scale was associated with a statistically significant mean change of 0.11 mm/s in maximum pupil constriction velocity, and of approximately 0.4% in pupil diameter. As expected, there was an association between total opioid dose (expressed as morphine equivalents) and pupil diameter. Age, sex and baseline anxiety scores did not correlate significantly with pupillary response. Conclusions The association of maximum pupillary constriction velocity and diameter with pain scores illustrates the potential for using pupillometry as a non-invasive method to objectively quantitate pain response/intensity in children. The technique holds promise as a pharmacodynamic ‘tool’ to assess opioid response in paediatric patients.

Collaboration


Dive into the Kathleen Neville's collaboration.

Top Co-Authors

Avatar

Gregory L. Kearns

Arkansas Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Julie A. Panepinto

Children's Hospital of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Uttam Garg

Children's Mercy Hospital

View shared research outputs
Top Co-Authors

Avatar

Deanna Mitchell

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael R. DeBaun

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adetola A. Kassim

Vanderbilt University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge