Network


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

Hotspot


Dive into the research topics where AeRang Kim is active.

Publication


Featured researches published by AeRang Kim.


Clinical Cancer Research | 2012

Sorafenib is an Inhibitor of UGT1A1 but is Metabolized by UGT1A9: Implications of Genetic Variants on Pharmacokinetics and Hyperbilirubinemia

Cody J. Peer; Tristan M. Sissung; AeRang Kim; Lokesh Jain; Sukyung Woo; Erin R. Gardner; C. Tyler Kirkland; Sarah M. Troutman; Bevin C. English; Emily D. Richardson; Joel Federspiel; David Venzon; William L. Dahut; Elise C. Kohn; Shivaani Kummar; Robert Yarchoan; Giuseppe Giaccone; Brigitte C. Widemann; William D. Figg

Purpose: Several case reports suggest sorafenib exposure and sorafenib-induced hyperbilirubinemia may be related to a (TA)5/6/7 repeat polymorphism in UGT1A1*28 (UGT, uridine glucuronosyl transferase). We hypothesized that sorafenib inhibits UGT1A1 and individuals carrying UGT1A1*28 and/or UGT1A9 variants experience greater sorafenib exposure and greater increase in sorafenib-induced plasma bilirubin concentration. Experimental Design: Inhibition of UGT1A1-mediated bilirubin glucuronidation by sorafenib was assessed in vitro. UGT1A1*28 and UGT1A9*3 genotypes were ascertained with fragment analysis or direct sequencing in 120 cancer patients receiving sorafenib on five different clinical trials. Total bilirubin measurements were collected in prostate cancer patients before receiving sorafenib (n = 41) and 19 to 30 days following treatment and were compared with UGT1A1*28 genotype. Results: Sorafenib exhibited mixed-mode inhibition of UGT1A1-mediated bilirubin glucuronidation (IC50 = 18 μmol/L; Ki = 11.7 μmol/L) in vitro. Five patients carrying UGT1A1*28/*28 (n = 4) or UGT1A9*3/*3 (n = 1) genotypes had first dose, dose-normalized areas under the sorafenib plasma concentration versus time curve (AUC) that were in the 93rd percentile, whereas three patients carrying UGT1A1*28/*28 had AUCs in the bottom quartile of all genotyped patients. The Drug Metabolizing Enzymes and Transporters genotyping platform was applied to DNA obtained from six patients, which revealed the ABCC2-24C>T genotype cosegregated with sorafenib AUC phenotype. Sorafenib exposure was related to plasma bilirubin increases in patients carrying 1 or 2 copies of UGT1A1*28 alleles (n = 12 and n = 5; R2 = 0.38 and R2 = 0.77; P = 0.032 and P = 0.051, respectively). UGT1A1*28 carriers showed two distinct phenotypes that could be explained by ABCC2-24C>T genotype and are more likely to experience plasma bilirubin increases following sorafenib if they had high sorafenib exposure. Conclusions: This pilot study indicates that genotype status of UGT1A1, UGT1A9, and ABCC2 and serum bilirubin concentration increases reflect abnormally high AUC in patients treated with sorafenib. Clin Cancer Res; 18(7); 2099–107. ©2012 AACR.


The New England Journal of Medicine | 2016

Activity of Selumetinib in Neurofibromatosis Type 1–Related Plexiform Neurofibromas

Eva Dombi; Andrea Baldwin; Leigh Marcus; Michael J. Fisher; Brian Weiss; AeRang Kim; Patricia Whitcomb; Staci Martin; Lindsey Aschbacher-Smith; Tilat A. Rizvi; Jianqiang Wu; Rachel Ershler; Pamela L. Wolters; Janet Therrien; John Glod; Jean B. Belasco; Elizabeth K. Schorry; Alessandra Brofferio; Amy J. Starosta; Andrea Gillespie; Austin L. Doyle; Nancy Ratner; Brigitte C. Widemann

BACKGROUND Effective medical therapies are lacking for the treatment of neurofibromatosis type 1-related plexiform neurofibromas, which are characterized by elevated RAS-mitogen-activated protein kinase (MAPK) signaling. METHODS We conducted a phase 1 trial of selumetinib (AZD6244 or ARRY-142886), an oral selective inhibitor of MAPK kinase (MEK) 1 and 2, in children who had neurofibromatosis type 1 and inoperable plexiform neurofibromas to determine the maximum tolerated dose and to evaluate plasma pharmacokinetics. Selumetinib was administered twice daily at a dose of 20 to 30 mg per square meter of body-surface area on a continuous dosing schedule (in 28-day cycles). We also tested selumetinib using a mouse model of neurofibromatosis type 1-related neurofibroma. Response to treatment (i.e., an increase or decrease from baseline in the volume of plexiform neurofibromas) was monitored by using volumetric magnetic resonance imaging analysis to measure the change in size of the plexiform neurofibroma. RESULTS A total of 24 children (median age, 10.9 years; range, 3.0 to 18.5) with a median tumor volume of 1205 ml (range, 29 to 8744) received selumetinib. Patients were able to receive selumetinib on a long-term basis; the median number of cycles was 30 (range, 6 to 56). The maximum tolerated dose was 25 mg per square meter (approximately 60% of the recommended adult dose). The most common toxic effects associated with selumetinib included acneiform rash, gastrointestinal effects, and asymptomatic creatine kinase elevation. The results of pharmacokinetic evaluations of selumetinib among the children in this trial were similar to those published for adults. Treatment with selumetinib resulted in confirmed partial responses (tumor volume decreases from baseline of ≥20%) in 17 of the 24 children (71%) and decreases from baseline in neurofibroma volume in 12 of 18 mice (67%). Disease progression (tumor volume increase from baseline of ≥20%) has not been observed to date. Anecdotal evidence of decreases in tumor-related pain, disfigurement, and functional impairment was observed. CONCLUSIONS Our early-phase data suggested that children with neurofibromatosis type 1 and inoperable plexiform neurofibromas benefited from long-term dose-adjusted treatment with selumetinib without having excess toxic effects. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01362803 .).


Journal of Clinical Oncology | 2010

Glucarpidase, Leucovorin, and Thymidine for High-Dose Methotrexate-Induced Renal Dysfunction: Clinical and Pharmacologic Factors Affecting Outcome

Brigitte C. Widemann; Frank M. Balis; AeRang Kim; Matthew Boron; Nalini Jayaprakash; Aiman Shalabi; Michelle O'Brien; Michelle Eby; Diane E. Cole; Robert F. Murphy; Elizabeth Fox; Percy Ivy; Peter C. Adamson

PURPOSE To assess the role of the recombinant bacterial enzyme, glucarpidase (carboxypeptidase-G(2)), leucovorin, and thymidine in the management and outcome of patients with high-dose methotrexate (HDMTX) -induced nephrotoxicity. METHODS Patients with HDMTX-induced nephrotoxicity received one to three doses of intravenous (IV) glucarpidase and leucovorin rescue. The initial cohort (n = 35) also received thymidine by continuous IV infusion. Subsequently, thymidine was restricted to patients with prolonged exposure (> 96 hours) to methotrexate (MTX) or with substantial MTX toxicity at study entry. Plasma MTX, leucovorin, and 5-methyltetrahydrofolate (5-mTHF) concentrations were measured pre- and postglucarpidase. Toxicities were monitored, and logistic regression analysis was used to assess the relationship of baseline characteristics to the development of severe toxicity and death. RESULTS Glucarpidase was administered at a median of 96 hours (receiving thymidine, n = 44) and 66 hours (not receiving thymidine, n = 56) after the start of the MTX infusion. Plasma MTX concentrations decreased within 15 minutes of glucarpidase by 98.7%. Plasma 5-mTHF concentrations also decreased more than 98% after administration of glucarpidase. Of 12 deaths, six were directly attributed to irreversible MTX toxicity. Presence of grade 4 toxicity before administration of glucarpidase, inadequate initial increase in leucovorin dosing, and administration of glucarpidase more than 96 hours after the start of the MTX infusion were associated with development of grade 4 and 5 toxicity. CONCLUSION Early intervention with the combination of leucovorin and glucarpidase is highly effective in patients who develop HDMTX-induced renal dysfunction. Severe toxicity and mortality occurred in patients in whom glucarpidase rescue was delayed and occurred despite thymidine administration.


Clinical Cancer Research | 2012

A Phase I Trial and Pharmacokinetic Study of the Raf Kinase and Receptor Tyrosine Kinase Inhibitor Sorafenib in Children with Refractory Solid Tumors or Refractory Leukemias

Brigitte C. Widemann; AeRang Kim; Elizabeth Fox; Sylvain Baruchel; Peter C. Adamson; A. Mark Ingle; Julia Glade Bender; Michael J. Burke; Brenda Weigel; Diana Stempak; Frank M. Balis; Susan M. Blaney

Purpose To determine the dose-limiting toxicities (DLT), maximum tolerated dose (MTD), pharmacokinetics and pharmacodynamics of sorafenib in children with refractory extracranial solid tumors and evaluate the tolerability of the solid tumor MTD in children with refractory leukemias. Experimental Design Sorafenib was administered orally q12h for consecutive 28 day cycles. Pharmacokinetics (day 1 and steady state) and pharmacodynamics were performed during cycle 1. Results Of sixty-five patients enrolled, 60 were eligible. In the solid tumor cohort (n=49), 4/6 patients experienced a DLT (hypertension, pain, rash/urticaria, thrombocytopenia, ALT/AST) at the starting dose (150 mg/m2/dose) which resulted in de-escalation to 105 mg/m2/dose. After eligibility criteria modification and dose re-escalation, the MTD was 200 mg/m2/dose for solid tumors and 150 mg/m2/dose for leukemias. Sorafenib exposure was highly variable between patients, but was within the ranges reported in adults. The apparent sorafenib clearance increased with patient age. Diarrhea, rash, fatigue, and increased ALT/AST were the most common sorafenib-related toxicities. Stable disease for ≥ 4 cycles was observed in 14 solid tumor patients, and two patients with acute myeloid leukemia and FLT3 internal tandem duplication experienced a decrease in bone marrow blasts toPurpose: To determine the dose-limiting toxicities (DLT), maximum tolerated dose (MTD), pharmacokinetics, and pharmacodynamics of sorafenib in children with refractory extracranial solid tumors and evaluate the tolerability of the solid tumor MTD in children with refractory leukemias. Experimental Design: Sorafenib was administered orally every 12 hours for consecutive 28-day cycles. Pharmacokinetics (day 1 and steady-state) and pharmacodynamics were conducted during cycle 1. Results: Of 65 patients enrolled, 60 were eligible. In the solid tumor cohort (n = 49), 4 of 6 patients experienced a DLT [hypertension, pain, rash/urticaria, thrombocytopenia, alanine aminotransferase (ALT)/aspartate aminotransferase (AST)] at the starting dose (150 mg/m2/dose) which resulted in de-escalation to 105 mg/m2/dose. After eligibility criteria modification and dose re-escalation, the MTD was 200 mg/m2/dose for solid tumors and 150 mg/m2/dose for leukemias. Sorafenib exposure was highly variable between patients but was within the ranges reported in adults. The apparent sorafenib clearance increased with patient age. Diarrhea, rash, fatigue, and increased ALT/AST were the most common sorafenib-related toxicities. Stable disease for 4 or more cycles was observed in 14 solid tumor patients, and 2 patients with acute myeloid leukemia (AML) and FLT3 internal tandem duplication (FLT3ITD) experienced a decrease in bone marrow blasts to less than 5%. Conclusions: The recommended phase II dose of sorafenib administered every 12 hours continuously for children with solid tumors is 200 mg/m2/dose and 150 mg/m2/dose for children with leukemias. Sorafenib toxicities and distribution in children are similar to adults. The activity of sorafenib in children with AML and FLT3ITD is currently being evaluated, and a phase II study for select solid tumors is ongoing. Clin Cancer Res; 18(21); 6011–22. ©2012 AACR.


JAMA Oncology | 2016

Multicenter Feasibility Study of Tumor Molecular Profiling to Inform Therapeutic Decisions in Advanced Pediatric Solid Tumors: The Individualized Cancer Therapy (iCat) Study

Marian H. Harris; Steven G. DuBois; Julia L. Glade Bender; AeRang Kim; Brian D. Crompton; Erin Parker; Ian P. Dumont; Andrew L. Hong; Dongjing Guo; Alanna Church; Kimberly Stegmaier; Charles W. M. Roberts; Suzanne Shusterman; Wendy B. London; Laura E. MacConaill; Neal I. Lindeman; Lisa Diller; Carlos Rodriguez-Galindo; Katherine A. Janeway

Importance Pediatric cancers represent a unique case with respect to cancer genomics and precision medicine, as the mutation frequency is low, and targeted therapies are less available. Consequently, it is unknown whether clinical sequencing can be of benefit. Objective To assess the feasibility of identifying actionable alterations and making individualized cancer therapy (iCat) recommendations in pediatric patients with extracranial solid tumors. Design, Setting, and Participants Clinical sequencing study at 4 academic medical centers enrolling patients between September 5, 2012, and November 19, 2013, with 1 year of clinical follow-up. Participants were 30 years or younger with high-risk, recurrent, or refractory extracranial solid tumors. The data analysis was performed October 28, 2014. Interventions Tumor profiling performed on archived clinically acquired specimens consisted of mutation detection by a Sequenom assay or targeted next-generation sequencing and copy number assessment by array comparative genomic hybridization. Results were reviewed by a multidisciplinary expert panel, and iCat recommendations were made if an actionable alteration was present, and an appropriate drug was available. Main Outcomes and Measures Feasibility was assessed using a 2-stage design based on the proportion of patients with recommendations. Results Of 100 participants (60 male; median [range] age, 13.4 [0.8-29.8] years), profiling was technically successful in 89 (89% [95% CI, 83%-95%]). Median (range) follow-up was 6.8 (2.0-23.6) months. Overall, 31 (31% [95% CI, 23%-41%]) patients received an iCat recommendation and 3 received matched therapy. The most common actionable alterations leading to an iCat recommendation were cancer-associated signaling pathway gene mutations (n = 10) and copy number alterations in MYC/MYCN (n = 6) and cell cycle genes (n = 11). Additional alterations with implications for clinical care but not resulting in iCat recommendations were identified, including mutations indicating the possible presence of a cancer predisposition syndrome and translocations suggesting a change in diagnosis. In total, 43 (43% [95% CI, 33%-53%]) participants had results with potential clinical significance. Conclusions and Relevance A multi-institution clinical genomics study in pediatric oncology is feasible and a substantial proportion of relapsed or refractory pediatric solid tumors have actionable alterations. Trial Registration clinicaltrials.gov Identifier: NCT01853345.


Pediatric Blood & Cancer | 2013

Phase I trial and pharmacokinetic study of sorafenib in children with neurofibromatosis type I and plexiform neurofibromas

AeRang Kim; Eva Dombi; Kathleen Tepas; Elizabeth Fox; Staci Martin; Pamela L. Wolters; Frank M. Balis; Nalini Jayaprakash; Baris Turkbey; Naira Muradyan; Peter L. Choyke; Alyssa T. Reddy; Bruce R. Korf; Brigitte C. Widemann

Sorafenib targets multiple pathways thought to be crucial in growth of plexiform neurofibroma (PN) in children with neurofibromatosis type 1 (NF1). Sorafenib has been tolerated with manageable toxicities in adults and children with refractory cancer. We conducted a separate study in this population. Monitoring long‐term toxicities such as effects on growth and obtaining additional pharmacokinetic data were of importance due to the young age and long duration of therapy seen in previous phase I trials in children with NF1.


Neurology | 2009

Characteristics of children enrolled in treatment trials for NF1-related plexiform neurofibromas

AeRang Kim; Andrea Gillespie; Eva Dombi; Anne Goodwin; Wendy Goodspeed; Elizabeth Fox; Frank M. Balis; Brigitte C. Widemann

Objective: To describe the characteristics of children enrolled in treatment trials for neurofibromatosis type 1 (NF1)–related plexiform neurofibroma (PN), PN tumor burden, PN-related complications, and treatment outcomes and to highlight the differences between characteristics of children with NF1 vs children with cancers entered on early phase drug trials. Methods: Pre-enrollment characteristics and complications of PN were retrospectively analyzed in a cohort of 59 children with NF1-related PN treated on 1 of 7 clinical trials at the NIH between 1996 and 2007. Outcome was analyzed in a subset of 19 patients enrolled in phase I trials. Comparisons to children with cancer were made from a similar analysis performed recently. Results: The median age at enrollment was 8 years. The median PN volume was 555 mL. Most patients had no prior chemotherapy or radiation, but nearly half had previous surgery for PN. PN-associated complications and NF1 manifestations were common, including pain (53%), other tumors (18%), and hypertension (8%). Investigational drug therapy was well tolerated. A median of 10 treatment cycles was administered. Patients with NF1-related PN were younger, had better performance score, had less prior therapy, and remained on study longer than cancer patients. Conclusions: Children with NF1-related plexiform neurofibroma (PN) enrolled in clinical trials had large tumors with substantial morbidity. Clinical trials in these children provide information about drug tolerance, cumulative toxicity, and pharmacokinetics in a younger population than early phase pediatric cancer trials. This report may aid in the evaluation of the applicability of traditional pediatric cancer trial designs and endpoints for NF1-related PN.


Pediatric Blood & Cancer | 2012

Preclincial testing of sorafenib and RAD001 in the Nf(flox/flox) ;DhhCre mouse model of plexiform neurofibroma using magnetic resonance imaging.

Jianqiang Wu; Eva Dombi; Edwin Jousma; R. Scott Dunn; Diana M. Lindquist; Beverly Schnell; Mi-Ok Kim; AeRang Kim; Brigitte C. Widemann; Timothy P. Cripe; Nancy Ratner

Neurofibromatosis type 1 (NF1) is an inherited disease predisposing affected patients to variable numbers of benign neurofibromas. To date there are no effective chemotherapeutic drugs available for this slow growing tumor. Molecularly targeted agents that aim to slow neurofibroma growth are being tested in clinical trials. So preclinical models for testing potential therapies are urgently needed to prioritize drugs for clinical trials of neurofibromas.


Oncologist | 2009

Sorafenib and Sunitinib

AeRang Kim; Frank M. Balis; Brigitte C. Widemann

Sorafenib (Nexavar®; Bayer Pharmaceuticals, West Haven, CT) (Fig. 1A) and sunitinib (Sutent®; Pfizer Inc., New York, NY) (Fig. 1B) are orally bioavailable, small molecule inhibitors of multiple intracellular and receptor protein kinases that are components of signaling pathways that control tumor growth and angiogenesis. These agents have similar drug profiles and overlapping targets, which are summarized in Table 1. Sorafenib and sunitinib are currently approved by the U.S. Food and Drug Administration for advanced renal cell cancer (RCC) in adults [1]. Sorafenib is also approved for unresect-able hepatocellular carcinoma (HCC) [2] and sunitinib is approved for gastrointestinal stromal tumor (GIST) after disease progression on or intolerance to imatinib mesylate [3]. These differing clinical indications are likely a result of the different development plans of the respective drug companies, rather than the specific mechanism for each drug. There are currently studies underway for GIST patients with sorafenib and for HCC patients with sunitinib. Open in a separate window Figure 1. Chemical structures. (A): Chemical structure of sorafenib. (B): Chemical structure of sunitinib.


Neuro-oncology | 2014

Phase 2 randomized, flexible crossover, double-blinded, placebo-controlled trial of the farnesyltransferase inhibitor tipifarnib in children and young adults with neurofibromatosis type 1 and progressive plexiform neurofibromas

Brigitte C. Widemann; Eva Dombi; Andrea Gillespie; Pamela L. Wolters; Jean B. Belasco; Stewart Goldman; Bruce R. Korf; Jeffrey Solomon; Staci Martin; Wanda L. Salzer; Elizabeth Fox; Nicholas J. Patronas; Mark W. Kieran; John P. Perentesis; Alyssa T. Reddy; John J. Wright; AeRang Kim; Seth M. Steinberg; Frank M. Balis

BACKGROUND RAS is dysregulated in neurofibromatosis type 1 (NF1) related plexiform neurofibromas (PNs). The activity of tipifarnib, which blocks RAS signaling by inhibiting its farnesylation, was tested in children and young adults with NF1 and progressive PNs. METHODS Patients aged 3-25 years with NF1-related PNs and imaging evidence of tumor progression were randomized in a double-blinded fashion to receive tipifarnib (200 mg/m(2) orally every 12 h) or placebo (phase A) and crossed over to the opposite treatment arm at the time of tumor progression (phase B). PN volumes were measured with MRI, and progression was defined as ≥20% volume increase. Time to progression (TTP) in phase A was the primary endpoint, and the trial was powered to detect whether tipifarnib doubled TTP compared with placebo. Toxicity, response, and quality of life were also monitored. RESULTS Sixty-two patients were enrolled. Tipifarnib and placebo were well tolerated. On phase A, the median TTP was 10.6 months on the placebo arm and 19.2 months on the tipifarnib arm (P = .12; 1-sided). Quality of life improved significantly compared with baseline on the tipifarnib arm but not on the placebo arm. Volumetric tumor measurement detected tumor progression earlier than conventional 2-dimensional (WHO) and 1-dimensional (RECIST) methods. CONCLUSIONS Tipifarnib was well tolerated but did not significantly prolong TTP of PNs compared with placebo. The randomized, flexible crossover design and volumetric PN assessment provided a feasible and efficient means of assessing the efficacy of tipifarnib. The placebo arm serves as an historical control group for phase 2 single-arm trials directed at progressive PNs.

Collaboration


Dive into the AeRang Kim's collaboration.

Top Co-Authors

Avatar

Brigitte C. Widemann

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Fox

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Eva Dombi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Frank M. Balis

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Susan M. Blaney

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nalini Jayaprakash

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Pamela L. Wolters

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Staci Martin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrea Baldwin

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge