Network


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

Hotspot


Dive into the research topics where Joseph R. Sherbotie is active.

Publication


Featured researches published by Joseph R. Sherbotie.


Pediatric Nephrology | 2001

The use of mycophenolate mofetil suspension in pediatric renal allograft recipients.

Timothy E. Bunchman; Mercedes Navarro; Michel Broyer; Joseph R. Sherbotie; Blanche M. Chavers; Burkhard Tönshoff; Patricia E. Birk; Gary Lerner; David S. Lirenman; Laurence A. Greenbaum; Rowan G. Walker; Lothar B. Zimmerhackl; Douglas L. Blowey; Godfrey Clark; Robert B. Ettenger; Sarah Arterburn; Karen Klamerus; Alice Fong; Helen Tang; Susan E. Thomas; Eleanor Ramos

Abstract. Mycophenolate mofetil (MMF) is widely used to prevent acute rejection in adults after renal, cardiac, and liver transplantation. This study investigated the safety, tolerability, and pharmacokinetics of MMF suspension in pediatric renal allograft recipients. One hundred renal allograft recipients were enrolled into three age groups (33 patients, 3 months to <6 years; 34 patients, 6 to <12 years; 33 patients, 12 to 18 years). Patients received MMF 600 mg/m2 b.i.d. concomitantly with cyclosporine and corticosteroids with or without antilymphocyte antibody induction. One year after transplantation, patient and graft survival (including death) were 98% and 93%, respectively. Twenty-five patients (25%) experienced a biopsy-proven (Banff grade borderline or higher) or presumptive acute rejection within the first 6 months post-transplantation. Analysis of pharmacokinetic parameters for mycophenolic acid (MPA) and mycophenolic acid glucuronide showed no clinically significant differences among the age groups. The dosing regimen of MMF 600 mg/m2 b.i.d. achieved the targeted early post-transplantation MPA 12-h area under concentration-time curve (AUC0–12) of 27.2 µg h per ml. Adverse events had similar frequencies among the age groups (with the exception of diarrhea, leukopenia, sepsis, and anemia, which were more frequent in the <6 years age group) and led to withdrawal of MMF in about 10% of patients. Administration of MMF 600 mg/m2 b.i.d. is effective in prevention of acute rejection, provides predictable pharmacokinetics, and is associated with an acceptable safety profile in pediatric renal transplant recipients.


Journal of Clinical Hypertension | 2008

Efficacy, Safety, and Pharmacokinetics of Candesartan Cilexetil in Hypertensive Children Aged 6 to 17 Years

Howard Trachtman; James W. Hainer; Jennifer Sugg; Renli Teng; Jonathan M. Sorof; Jerilynn Radcliffe; Johan Vande Walle; Laszio Szabo; Tivadar Tulassay; Sándor Túri; Eva Marova; Alexander Jurko; Maria Horakova; Robert Achtel; John Barcia; Donald L. Batisky; Patrick D. Brophy; Bonita Falkner; Joseph T. Flynn; Randall Jenkins; Vijay Kusnoor; Kenneth A. Miller; Ana Paredes; Irene Restaino; Joseph R. Sherbotie; Gaston Zilleruelo; Myra Chiang; Farahnak Assadi; Shashi Nagaraj; Janice E. Sullivan

This 4‐week randomized, double blind, placebo‐controlled study (N=240), 1‐year open label trial (N=233), and single‐dose pharmacokinetic study (N=22) evaluated candesartan cilexetil (3 doses) in hypertensive children aged 6 to 17 years. Seventy‐one percent were 12 years of age or older, 71% were male, and 47% were black. Systolic (SBP)/diastolic (DBP) blood pressure declined 8.6/4.8–11.2/8.0 mm Hg with candesartan and 3.7/1.8 mm Hg with placebo (P<.01 compared to placebo for SBP and for the mid and high doses for DBP; placebo‐corrected 4.9/3.0–7.5/6.2 mm Hg). The slopes for dose were not, however, different from zero (P>.05). The response rate (SBP and DBP <95th percentile) after 1 year was 53%. The pharmacokinetic profiles in 6‐ to 12‐ and 12‐ to 17‐year‐olds were similar and were comparable to adults. Eight candesartan patients discontinued treatment because of an adverse event. Candesartan is an effective, well‐tolerated antihypertensive agent for children aged 6 to 17 years and has a pharmacokinetic profile that is similar to that in adults.


The Journal of Clinical Pharmacology | 2011

Inosine monophosphate dehydrogenase (IMPDH) activity as a pharmacodynamic biomarker of mycophenolic acid effects in pediatric kidney transplant recipients.

Tsuyoshi Fukuda; Jens Goebel; Håvard Thøgersen; Denise Maseck; Shareen Cox; Barbara Logan; Joseph R. Sherbotie; Mouin G. Seikaly; Alexander A. Vinks

Monitoring inosine monophosphate dehydrogenase (IMPDH) activity as a biomarker of mycophenolic acid (MPA)–induced immunosuppression may serve as a novel approach in pharmacokinetics (PK)/pharmacodynamics (PD)–guided therapy. The authors prospectively studied MPA pharmacokinetics and IMPDH inhibition in 28 pediatric de novo kidney transplant recipients. Pretransplant IMPDH activity and full PK/PD profiles were obtained at 3 different occasions: 1 to 3 days, 4 to 9 days, and approximately 6 months after transplant. Large intra‐ and interpatient variability was noted in MPA pharmacokinetics and exposure and IMPDH inhibition. MPA exposure (AUC0–12 h) was low early posttransplant and increased over time and stabilized at months 3 to 6. Mean pretransplant IMPDH activity (6.4 ± 4.6 nmol/h/mg protein) was lower than previously reported in adults. In most of the patients, IMPDH enzyme activity decreased with increasing MPA plasma concentration, with maximum inhibition coinciding with maximum MPA concentration. The overall relationship between MPA concentration and IMPDH activity was described by a direct inhibitory Emax model (EC50 = 0.97 mg/L). This study suggests the importance of early PK/PD monitoring to improve drug exposure. Because IMPDH inhibition is well correlated to MPA concentration, pretransplant IMPDH activity may serve as an early marker to guide the initial level of MPA exposure required in a pediatric population.


Therapeutic Drug Monitoring | 2012

UGT1A9, UGT2B7, and MRP2 genotypes can predict mycophenolic acid pharmacokinetic variability in pediatric kidney transplant recipients.

Tsuyoshi Fukuda; Jens Goebel; Shareen Cox; Denise Maseck; Kejian Zhang; Joseph R. Sherbotie; Eileen N. Ellis; Laura P. James; Robert M. Ward; Alexander A. Vinks

Background: Mycophenolic acid (MPA) exposure in pediatric patients with kidney transplant receiving body surface area (BSA)–based dosing exhibits large variability. Several genetic variants in glucuronosyltransferases (UGTs) and of multidrug resistance–associated protein 2 (MRP2) have independently been suggested to predict MPA exposure in adult patients with varying results. Here, the combined contribution of these genetic variants to MPA pharmacokinetic variability was investigated in pediatric renal transplant recipients who were on mycophenolic mofetil maintenance therapy. Methods: MPA and MPA-glucuronide concentrations from 32 patients were quantified by high-performance liquid chromatography. MPA exposure (AUC) was estimated using a 4-point abbreviated sampling strategy (predose/trough and 20 minutes, 1 hour, and 3 hours after dose) using a validated pediatric Bayesian estimator. Genotyping was performed for all of the following single nucleotide polymorphisms (SNPs): UGT1A8 830G>A(*3), UGT1A9 98T>C(*3), UGT1A9-440C>T, UGT1A9-2152C>T, UGT1A9-275T>A, UGT2B7-900A>G, and MRP2-24T>C. Results: Recipients heterozygous for MRP2-24T>C who also had UGT1A9-440C>T or UGT2B7-900A>G (n = 4), and MRP2-24T>C-negative recipients having both UGT1A9-440C>T and UGT2B7-900A>G (n = 5) showed a 2.2 and 1.7 times higher dose-dependent and BSA-normalized MPA-AUC compared with carriers of no or only 1 UGT-SNP (P < 0.001 and P = 0.01, respectively) (n = 7). Dose-dependent and BSA-normalized predose MPA concentrations were 3.0 and 2.4 times higher, respectively (P < 0.001). Interindividual variability in peak concentrations could be explained by the presence of the UGT1A9-440C>T genotype (P < 0.05). Conclusion: Our preliminary study demonstrates that combined UGT1A9-440C>T, UGT2B7-900A>G, and MRP2-24T>C polymorphisms can be important predictors of interindividual variability in MPA exposure in the pediatric population.


Pediatric Transplantation | 2008

Collecting duct carcinoma arising in association with bk nephropathy post-transplantation in a pediatric patient. a case report with immunohistochemical and in situ hybridization study

Lyska Emerson; Heather M. Carney; Lester J. Layfield; Joseph R. Sherbotie

Abstract:  The development of malignancy in a renal transplant graft is an uncommon phenomenon. A renal neoplasm developing in the adult donor kidney of a pediatric transplant recipient has only rarely been reported. We report a case of collecting duct carcinoma arising in association with BK virus nephropathy in an adult living‐related donor renal allograft to a pediatric recipient. Our case is the second report of neoplasia occurring in association with BK virus nephropathy post‐transplantation, suggesting that BK virus may play a role in oncogenesis. It has been proposed that the T‐Ag protein encoded by the polyomavirus family of viruses disrupts chromosomal integrity, creating oncogenes, and inactivating tumor suppressor genes. In our study, immunohistochemical staining with antibody directed against BK virus large T antigen showed nuclear staining within urothelium, tubular epithelium, tubular intraepithelial neoplasia, and invasive carcinoma. In situ hybridization did not identify BK virus DNA within neoplastic cells. T‐Ag protein expression has been shown to be tumor‐specific in bladder, gastric, and colorectal cancers. The finding of T‐Ag protein expression in both intraepithelial and invasive neoplastic tissues in our case raises the possibility of BK virus as a causative agent in oncogenesis.


Clinical Pharmacology & Therapeutics | 2009

UGT Genotype May Contribute to Adverse Events Following Medication With Mycophenolate Mofetil in Pediatric Kidney Transplant Recipients

Se Prausa; Tsuyoshi Fukuda; D Maseck; Kl Curtsinger; C Liu; Kejian Zhang; Tg Nick; Joseph R. Sherbotie; Eileen N. Ellis; Jens Goebel; Alexander A. Vinks

Leukopenia and diarrhea are the predominant adverse events associated with mycophenolate mofetil (MMF), leading to dose reduction or discontinuation in children. Polymorphisms of the drugs main metabolizing enzyme, uridine diphosphate–glucuronosyl transferase (UGT), confer alteration in drug exposure. We studied the incidence of these polymorphisms in pediatric kidney transplant recipients experiencing MMF‐associated leukopenia and diarrhea. UGT genotypes of 16 affected children who recovered after MMF dose reduction or discontinuation were compared with those of 22 children who tolerated the drug at standard doses. DNA was extracted and sequenced using standard procedures to detect polymorphisms associated with increased (e.g., UGT1A9 −331T>C) or decreased drug exposure. All three patients who were homozygous for UGT1A9 −331T>C developed leukopenia, and heterozygotes also had significantly more toxicity (P = 0.04). A weaker association (P = 0.08) existed in UGT2B7 −900G>A carriers. Our data implicate UGT polymorphisms associated with altered drug exposure as potential predictors of MMF adverse events.


Pediatric Transplantation | 2001

Indications, results, and complications of tacrolimus conversion in pediatric renal transplantation.

Joseph T. Flynn; Timothy E. Bunchman; Joseph R. Sherbotie

Abstract: It is the practice of many pediatric renal transplant programs to ‘convert’ children taking cyclosporin A (CsA) to tacrolimus, although the indications for, outcome, and complications of this practice remain obscure. To better understand these aspects of tacrolimus ‘conversion’, a fax survey was sent to 119 North American pediatric renal transplant centers. Analyzable responses were received from 52 centers (44%), and included data from ≈ 1,815 pediatric renal transplants performed between 1991 and 98. Strong indications for tacrolimus conversion were: antibody‐resistant rejection, CsA‐resistant rejection, and CsA intolerance (strong indication in 72%, 65%, and 52% of centers, respectively). Steroid‐resistant rejection and cosmetic side‐effects were considered strong indications less often. Initial anti‐rejection therapy was usually increased corticosteroid dose (47/52 centers). Antibody therapy was most commonly used for steroid‐resistant rejection (44 centers). For steroid‐ and antibody‐resistant rejection, tacrolimus conversion was most common (33 centers). Tacrolimus conversion for antibody‐resistant rejection led to improvement of serum creatinine (SCr) in 27% of patients, stabilization of SCr in 46%, worsening of SCr in 11%, and graft loss in 16%. Reported complications after tacrolimus conversion included hyperglycemia, hyperkalemia, lymphoproliferative disorder, infection, and neurologic problems. We conclude that the major indication for tacrolimus conversion in pediatric transplant programs appears to be rejection. Outcome after tacrolimus conversion appears good, with the majority of patients experiencing stable or improved allograft function. These data provide direction for further study, including timing of tacrolimus conversion and interaction with other therapies.


Therapeutic Drug Monitoring | 2014

Effects of unbound mycophenolic acid on inosine monophosphate dehydrogenase inhibition in pediatric kidney transplant patients.

Thomas A. Smits; Shareen Cox; Tsuyoshi Fukuda; Joseph R. Sherbotie; Robert M. Ward; Jens Goebel; Alexander A. Vinks

Background: Mycophenolic acid (MPA) is a key immunosuppressive drug that acts through inhibition of inosine monophosphate dehydrogenase (IMPDH). MPA is commonly measured, as part of therapeutic drug monitoring, as the total concentration in plasma. However, it has been postulated that the free (unbound) fraction of MPA (fMPA) is responsible for the immunosuppressive effects. In this study, a sensitive low volume high-performance liquid chromatography (HPLC) assay was developed to measure fMPA concentrations to explore the relationship between fMPA and IMPDH activity. Methods: To obtain fMPA concentrations, plasma samples were filtrated using Centrifree ultrafiltration devices. The ultrafiltrate was analyzed by HPLC using a Kinetex C18 column (2.6 &mgr;m, 3.0 × 75 mm). fMPA concentrations were compared with the total MPA concentrations available in 28 pediatric kidney transplant patients at 3 consecutive occasions after transplantation. The relationship between fMPA and IMPDH activity was analyzed using an Emax model. Results: The HPLC assay, using 25 &mgr;L of the ultrafiltrates, was validated over a range from 2.5 to 1000 &mgr;L with good accuracy, precision, and reproducibility. Total and free MPA concentrations were well correlated (R2 = 0.85, P < 0.0001), although large intraindividual and interindividual variability in the bound MPA fractions was observed. The overall relationship between fMPA concentrations and IMPDH inhibition using the Emax model was comparable with that of total MPA, as previously reported. The model estimated EC50 value (164.5 &mgr;L) is in good agreement with reported in vitro EC50 values. Conclusions: This study provides a simple HPLC method for the measurement of fMPA and a pharmacologically reasonable EC50 estimate. The good correlation between the total and free MPA concentrations suggests that routine measurement of fMPA to characterize mycophenolate pharmacokinetic and pharmacodynamic does not seem warranted, although the large variability in the bound fractions of MPA warrants further study.


Pediatric Transplantation | 2017

Bortezomib in the treatment of antibody-mediated rejection in pediatric kidney transplant recipients: A multicenter Midwest Pediatric Nephrology Consortium study

Sarah Kizilbash; Donna J. Claes; Isa F. Ashoor; Ashton Chen; Sara Jandeska; Raed Bou Matar; Jason M. Misurac; Joseph R. Sherbotie; Katherine Twombley; Priya S. Verghese

Antibody‐mediated rejection leads to allograft loss after kidney transplantation. Bortezomib has been used in adults for the reversal of antibody‐mediated rejection; however, pediatric data are limited. This retrospective study was conducted in collaboration with the Midwest Pediatric Nephrology Consortium. Pediatric kidney transplant recipients who received bortezomib for biopsy‐proven antibody‐mediated rejection between 2008 and 2015 were included. The objective was to characterize the use of bortezomib in pediatric kidney transplant recipients. Thirty‐three patients received bortezomib for antibody‐mediated rejection at nine pediatric kidney transplant centers. Ninety percent of patients received intravenous immunoglobulin, 78% received plasmapheresis, and 78% received rituximab. After a median follow‐up of 15 months, 65% of patients had a functioning graft. The estimated glomerular filtration rate improved or stabilized in 61% and 36% of patients at 3 and 12 months post‐bortezomib, respectively. The estimated glomerular filtration rate at diagnosis significantly predicted estimated glomerular filtration rate at 12 months after adjusting for chronic histologic changes (P .001). Fifty‐six percent of patients showed an at least 25% reduction in the mean fluorescence intensity of the immune‐dominant donor‐specific antibody, 1‐3 months after the first dose of bortezomib. Non‐life‐threatening side effects were documented in 21 of 33 patients. Pediatric kidney transplant recipients tolerated bortezomib without life‐threatening side effects. Bortezomib may stabilize estimated glomerular filtration rate for 3‐6 months in pediatric kidney transplant recipients with antibody‐mediated rejection.


Pediatric Drugs | 2012

Pharmacokinetics of Olmesartan Medoxomil in Pediatric Patients with Hypertension

Thomas G. Wells; Douglas L. Blowey; Janice E. Sullivan; Jeffrey L. Blumer; Joseph R. Sherbotie; SaeHeum Song; Shashank Rohatagi; Reinilde Heyrman; Daniel E. Salazar

AbstractBackground: The prevalence and importance of hypertension in younger patients is becoming increasingly recognized; however, only a limited number of clinical trials have been conducted in the pediatric population. Objective: The aim of this study was to characterize the pharmacokinetics and short-term safety of olmesartan medoxomil in children and adolescents with hypertension. Methods: An open-label, multicenter, single-dose study was conducted in children and adolescents aged 12 months–16 years who were receiving treatment for hypertension or, if not currently treated for hypertension, had either a systolic blood pressure (SBP) or diastolic blood pressure (DBP) ≤95th percentile, or SBP or DBP ≤90th percentile if diabetic or with a family history of hypertension. Patients were stratified by age: 12–23 months (Group 1; none enrolled), 2–5 years (Group 2; n = 4), 6–12 years (Group 3; n = 10), and 13–16 years (Group 4; n = 10). All patients received a single oral dose of olmesartan medoxomil based on the individual’s age and bodyweight. Patients aged <6 years received an oral suspension of olmesartan medoxomil at a dose of 0.3 mg/kg of bodyweight (not to exceed 20 mg), those aged ≤6 years who weighed ≤35 kg received olmesartan medoxomil 40 mg tablets, and those who weighed <35 kg received olmesartan medoxomil 20 mg tablets. Results: In Groups 2, 3, and 4, the weight-adjusted apparent total body clearance (CL/F) of olmesartan medoxomil was 0.100 ± 0.034, 0.062 ± 0.020, and 0.072± 0.022L/h/kg, respectively, and the weight-adjusted apparent volume of distribution (Vd/F) was 0.32± 0.16, 0.33 ±0.14, and 0.49±0.23 L/kg, respectively. CL/F and Vd/F in Groups 3 and 4 were not significantly different. Statistical comparisons between Groups 3 or 4 and Group 2 were not performed due to the small sample size of Group 2 (n =4). Plasma elimination half-life and time to maximum plasma concentration were similar across the three groups. In Groups 3 and 4, considerable interindividual variability was seen in maximum plasma concentration (Cmax), area under the curve (AUC) from time zero to the last measurable concentration, and apparent clearance, with AUC and Cmax approximately 30% greater in Group 3. Four of 24 (16.7%) patients experienced treatment-emergent adverse events that were all mild in severity and considered not drug-related. No deaths, serious adverse events, or discontinuations due to adverse events occurred in the study. Conclusions: Olmesartan medoxomil was well tolerated and demonstrated a pharmacokinetic profile in pediatric patients similar to that of adults when adjusted for body size. Trial Registration: ClinicalTrials.gov identifier: NCT00151814

Collaboration


Dive into the Joseph R. Sherbotie's collaboration.

Top Co-Authors

Avatar

Alexander A. Vinks

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jens Goebel

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tsuyoshi Fukuda

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shareen Cox

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Denise Maseck

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Douglas L. Blowey

University of Missouri–Kansas City

View shared research outputs
Top Co-Authors

Avatar

Eileen N. Ellis

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kejian Zhang

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge