Network


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

Hotspot


Dive into the research topics where Eric Larsen is active.

Publication


Featured researches published by Eric Larsen.


Blood | 2013

Tyrosine kinome sequencing of pediatric acute lymphoblastic leukemia: a report from the Children's Oncology Group TARGET Project

Mignon L. Loh; Jinghui Zhang; Richard C. Harvey; Kathryn G. Roberts; Debbie Payne-Turner; Huining Kang; Gang Wu; Xiang Chen; Jared Becksfort; Michael Edmonson; Kenneth H. Buetow; William L. Carroll; I. Ming Chen; Brent L. Wood; Michael J. Borowitz; Meenakshi Devidas; Daniela S. Gerhard; Paul Bowman; Eric Larsen; Naomi J. Winick; Elizabeth A. Raetz; Malcolm A. Smith; James R. Downing; Cheryl L. Willman; Charles G. Mullighan; Stephen P. Hunger

One recently identified subtype of pediatric B-precursor acute lymphoblastic leukemia (ALL) has been termed BCR-ABL1-like or Ph-like because of similarity of the gene expression profile to BCR-ABL1 positive ALL suggesting the presence of lesions activating tyrosine kinases, frequent alteration of IKZF1, and poor outcome. Prior studies demonstrated that approximately half of these patients had genomic lesions leading to CRLF2 overexpression, with half of such cases harboring somatic mutations in the Janus kinases JAK1 and JAK2. To determine whether mutations in other tyrosine kinases might also occur in ALL, we sequenced the tyrosine kinome and downstream signaling genes in 45 high-risk pediatric ALL cases with either a Ph-like gene expression profile or other alterations suggestive of activated kinase signaling. Aside from JAK mutations and 1 FLT3 mutation, no somatic mutations were found in any other tyrosine kinases, suggesting that alternative mechanisms are responsible for activated kinase signaling in high-risk ALL.


Pediatrics | 2012

Continuous Versus Bolus Infusion of Doxorubicin in Children With ALL: Long-term Cardiac Outcomes

Steven E. Lipshultz; Tracie L. Miller; Stuart R. Lipsitz; Donna Neuberg; Suzanne E. Dahlberg; Steven D. Colan; Lewis B. Silverman; Jacqueline M. Henkel; Vivian L. Franco; Laura L. Cushman; Barbara L. Asselin; Luis A. Clavell; Uma H. Athale; Bruno Michon; Caroline Laverdière; Marshall A. Schorin; Eric Larsen; G. Naheed Usmani; Stephen E. Sallan

BACKGROUND AND OBJECTIVES: Doxorubicin, effective against many malignancies, is limited by cardiotoxicity. Continuous-infusion doxorubicin, compared with bolus-infusion, reduces early cardiotoxicity in adults. Its effectiveness in reducing late cardiotoxicity in children remains uncertain. We determined continuous-infusion doxorubicin cardioprotective efficacy in long-term survivors of childhood acute lymphoblastic leukemia (ALL). METHODS: The Dana-Farber Cancer Institute ALL Consortium Protocol 91-01 enrolled pediatric patients between 1991 and 1995. Newly diagnosed high-risk patients were randomly assigned to receive a total of 360 mg/m2 of doxorubicin in 30 mg/m2 doses every 3 weeks, by either continuous (over 48 hours) or bolus-infusion (within 15 minutes). Echocardiograms at baseline, during, and after doxorubicin therapy were blindly remeasured centrally. Primary outcomes were late left ventricular (LV) structure and function. RESULTS: A total of 102 children were randomized to each treatment group. We analyzed 484 serial echocardiograms from 92 patients (n = 49 continuous; n = 43 bolus) with ≥1 echocardiogram ≥3 years after assignment. Both groups had similar demographics and normal baseline LV characteristics. Cardiac follow-up after randomization (median, 8 years) showed changes from baseline within the randomized groups (depressed systolic function, systolic dilation, reduced wall thickness, and reduced mass) at 3, 6, and 8 years; there were no statistically significant differences between randomized groups. Ten-year ALL event-free survival rates did not differ between the 2 groups (continuous-infusion, 83% versus bolus-infusion, 78%; P = .24). CONCLUSIONS: In survivors of childhood high-risk ALL, continuous-infusion doxorubicin, compared with bolus-infusion, provided no long-term cardioprotection or improvement in ALL event-free survival, hence provided no benefit over bolus-infusion.


Blood | 2015

Prognostic significance of minimal residual disease in high risk B-ALL: a report from Children’s Oncology Group study AALL0232

Michael J. Borowitz; Brent L. Wood; Meenakshi Devidas; Mignon L. Loh; Elizabeth A. Raetz; Wanda L. Salzer; James Nachman; Andrew J. Carroll; Nyla A. Heerema; Julie M. Gastier-Foster; Cheryl L. Willman; Yunfeng Dai; Naomi J. Winick; Stephen P. Hunger; William L. Carroll; Eric Larsen

Minimal residual disease (MRD) is highly prognostic in pediatric B-precursor acute lymphoblastic leukemia (B-ALL). In Childrens Oncology Group high-risk B-ALL study AALL0232, we investigated MRD in subjects randomized in a 2 × 2 factorial design to receive either high-dose methotrexate (HD-MTX) or Capizzi methotrexate (C-MTX) during interim maintenance (IM) or prednisone or dexamethasone during induction. Subjects with end-induction MRD ≥0.1% or those with morphologic slow early response were nonrandomly assigned to receive a second IM and delayed intensification phase. MRD was measured by 6-color flow cytometry in 1 of 2 reference labs, with excellent agreement between the two. Subjects with end-induction MRD <0.01% had a 5-year event-free survival (EFS) of 87% ± 1% vs 74% ± 4% for those with MRD 0.01% to 0.1%; increasing MRD amounts was associated with progressively worse outcome. Subjects converting from MRD positive to negative by end consolidation had a relatively favorable 79% ± 5% 5-year disease-free survival vs 39% ± 7% for those with MRD ≥0.01%. Although HD-MTX was superior to C-MTX, MRD retained prognostic significance in both groups (86% ± 2% vs 58% ± 4% for MRD-negative vs positive C-MTX subjects; 88% ± 2% vs 68% ± 4% for HD-MTX subjects). Intensified therapy given to subjects with MRD >0.1% did not improve either 5-year EFS or overall survival (OS). However, these subjects showed an early relapse rate similar to that seen in MRD-negative ones, with EFS/OS curves for patients with 0.1% to 1% MRD crossing those with 0.01% to 0.1% MRD at 3 and 4 years, thus suggesting that the intensified therapy altered the disease course of MRD-positive subjects. Additional interventions targeted at the MRD-positive group may further improve outcome. This trial was registered at www.clinicaltrials.gov as #NCT00075725.


Blood | 2015

Genetics of glucocorticoid-associated osteonecrosis in children with acute lymphoblastic leukemia.

Seth E. Karol; Wenjian Yang; Sara L. Van Driest; Tamara Chang; Sue C. Kaste; Erica Bowton; Melissa A. Basford; Dan M. Roden; Joshua C. Denny; Eric Larsen; Naomi J. Winick; William L. Carroll; Cheng Cheng; Deqing Pei; Christian A. Fernandez; Chengcheng Liu; Colton Smith; Mignon L. Loh; Elizabeth A. Raetz; Stephen P. Hunger; Paul Scheet; Sima Jeha; Ching-Hon Pui; William E. Evans; Meenakshi Devidas; Leonard A. Mattano; Mary V. Relling

Glucocorticoids are important therapy for acute lymphoblastic leukemia (ALL) and their major adverse effect is osteonecrosis. Our goal was to identify genetic and nongenetic risk factors for osteonecrosis. We performed a genome-wide association study of single nucleotide polymorphisms (SNPs) in a discovery cohort comprising 2285 children with ALL, treated on the Childrens Oncology Group AALL0232 protocol (NCT00075725), adjusting for covariates. The minor allele at SNP rs10989692 (near the glutamate receptor GRIN3A locus) was associated with osteonecrosis (hazard ratio = 2.03; P = 3.59 × 10(-7)). The association was supported by 2 replication cohorts, including 361 children with ALL on St. Judes Total XV protocol (NCT00137111) and 309 non-ALL patients from Vanderbilt Universitys BioVU repository treated with glucocorticoids (odds ratio [OR] = 1.87 and 2.26; P = .063 and .0074, respectively). In a meta-analysis, rs10989692 was also highest ranked (P = 2.68 × 10(-8)), and the glutamate pathway was the top ranked pathway (P = 9.8 × 10(-4)). Osteonecrosis-associated glutamate receptor variants were also associated with other vascular phenotypes including cerebral ischemia (OR = 1.64; P = 2.5 × 10(-3)), and arterial embolism and thrombosis (OR = 1.88; P = 4.2 × 10(-3)). In conclusion, osteonecrosis was associated with inherited variations near glutamate receptor genes. Further understanding this association may allow interventions to decrease osteonecrosis. These trials are registered at www.clinicaltrials.gov as #NCT00075725 and #NCT00137111.


Pediatric Blood & Cancer | 2011

Augmented Therapy Improves Outcome For Pediatric High Risk Acute Lymphocytic Leukemia: Results Of Children's Oncology Group Trial P9906

W. Paul Bowman; Eric Larsen; Meenakshi Devidas; Stephen B. Linda; Laurie Blach; Andrew J. Carroll; William L. Carroll; D. Jeanette Pullen; Jonathan J. Shuster; Cheryl L. Willman; Naomi J. Winick; Bruce M. Camitta; Stephen P. Hunger; Michael J. Borowitz

The augmented BFM regimen improves outcome for children with NCI high acute lymphoblastic leukemia (ALL). Patient age, sex, and presenting white blood cell count (WBC) can be used to identify a subset of approximately 12% of children with B‐precursor ALL that had a 5‐year continuous complete remission (CCR) rate of only about 50% on earlier Pediatric Oncology Group (POG) trials.


Blood | 2015

Genome-wide analysis links NFATC2 with asparaginase hypersensitivity

Christian A. Fernandez; Colton Smith; Wenjian Yang; Charles G. Mullighan; Chunxu Qu; Eric Larsen; W. Paul Bowman; Chengcheng Liu; Laura B. Ramsey; Tamara Chang; Seth E. Karol; Mignon L. Loh; Elizabeth A. Raetz; Naomi J. Winick; Stephen P. Hunger; William L. Carroll; Sima Jeha; Ching-Hon Pui; William E. Evans; Meenakshi Devidas; Mary V. Relling

Asparaginase is used to treat acute lymphoblastic leukemia (ALL); however, hypersensitivity reactions can lead to suboptimal asparaginase exposure. Our objective was to use a genome-wide approach to identify loci associated with asparaginase hypersensitivity in children with ALL enrolled on St. Jude Childrens Research Hospital (SJCRH) protocols Total XIIIA (n = 154), Total XV (n = 498), and Total XVI (n = 271), or Childrens Oncology Group protocols POG 9906 (n = 222) and AALL0232 (n = 2163). Germline DNA was genotyped using the Affymetrix 500K, Affymetrix 6.0, or the Illumina Exome BeadChip array. In multivariate logistic regression, the intronic rs6021191 variant in nuclear factor of activated T cells 2 (NFATC2) had the strongest association with hypersensitivity (P = 4.1 × 10(-8); odds ratio [OR] = 3.11). RNA-seq data available from 65 SJCRH ALL tumor samples and 52 Yoruba HapMap samples showed that samples carrying the rs6021191 variant had higher NFATC2 expression compared with noncarriers (P = 1.1 × 10(-3) and 0.03, respectively). The top ranked nonsynonymous polymorphism was rs17885382 in HLA-DRB1 (P = 3.2 × 10(-6); OR = 1.63), which is in near complete linkage disequilibrium with the HLA-DRB1*07:01 allele we previously observed in a candidate gene study. The strongest risk factors for asparaginase allergy are variants within genes regulating the immune response.


Journal of Clinical Oncology | 2018

TP53 germline variations influence the predisposition and prognosis of b-cell acute lymphoblastic leukemia in children

Maoxiang Qian; Xueyuan Cao; Meenakshi Devidas; Wenjian Yang; Cheng Cheng; Yunfeng Dai; Andrew J. Carroll; Nyla A. Heerema; Hui Zhang; Takaya Moriyama; Julie M. Gastier-Foster; Heng Xu; Elizabeth A. Raetz; Eric Larsen; Naomi J. Winick; W. Paul Bowman; Paul L. Martin; Elaine R. Mardis; Robert S. Fulton; Gerard P. Zambetti; Michael J. Borowitz; Brent L. Wood; Kim E. Nichols; William L. Carroll; Ching-Hon Pui; Charles G. Mullighan; William E. Evans; Stephen P. Hunger; Mary V. Relling; Mignon L. Loh

Purpose Germline TP53 variation is the genetic basis of Li-Fraumeni syndrome, a highly penetrant cancer predisposition condition. Recent reports of germline TP53 variants in childhood hypodiploid acute lymphoblastic leukemia (ALL) suggest that this type of leukemia is another manifestation of Li-Fraumeni syndrome; however, the pattern, prevalence, and clinical relevance of TP53 variants in childhood ALL remain unknown. Patients and Methods Targeted sequencing of TP53 coding regions was performed in 3,801 children from the Childrens Oncology Group frontline ALL clinical trials, AALL0232 and P9900. TP53 variant pathogenicity was evaluated according to experimentally determined transcriptional activity, in silico prediction of damaging effects, and prevalence in non-ALL control populations. TP53 variants were analyzed for their association with ALL presenting features and treatment outcomes. Results We identified 49 unique nonsilent rare TP53 coding variants in 77 (2.0%) of 3,801 patients sequenced, of which 22 variants were classified as pathogenic. TP53 pathogenic variants were significantly over-represented in ALL compared with non-ALL controls (odds ratio, 5.2; P < .001). Children with TP53 pathogenic variants were significantly older at ALL diagnosis (median age, 15.5 years v 7.3 years; P < .001) and were more likely to have hypodiploid ALL (65.4% v 1.2%; P < .001). Carrying germline TP53 pathogenic variants was associated with inferior event-free survival and overall survival (hazard ratio, 4.2 and 3.9; P < .001 and .001, respectively). In particular, children with TP53 pathogenic variants were at a dramatically higher risk of second cancers than those without pathogenic variants, with 5-year cumulative incidence of 25.1% and 0.7% ( P < .001), respectively. Conclusion Loss-of-function germline TP53 variants predispose children to ALL and to adverse treatment outcomes with ALL therapy, particularly the risk of second malignant neoplasms.


Journal of Clinical Oncology | 2017

Neurocognitive Functioning of Children Treated for High-Risk B-Acute Lymphoblastic Leukemia Randomly Assigned to Different Methotrexate and Corticosteroid Treatment Strategies: A Report From the Children’s Oncology Group

Kristina K. Hardy; Leanne Embry; John A. Kairalla; Shanjun Helian; Meenakshi Devidas; Daniel Armstrong; Stephen P. Hunger; William L. Carroll; Eric Larsen; Elizabeth A. Raetz; Mignon L. Loh; Wenjian Yang; Mary V. Relling; Robert B. Noll; Naomi J. Winick

Purpose Survivors of childhood acute lymphoblastic leukemia (ALL) are at risk for neurocognitive deficits that are associated with treatment, individual, and environmental factors. This study examined the impact of different methotrexate (MTX) and corticosteroid treatment strategies on neurocognitive functioning in children with high-risk B-lineage ALL. Methods Participants were randomly assigned to receive high-dose MTX with leucovorin rescue or escalating dose MTX with PEG asparaginase without leucovorin rescue. Patients were also randomly assigned to corticosteroid therapy that included either dexamethasone or prednisone. A neurocognitive evaluation of intellectual functioning (IQ), working memory, and processing speed (PS) was conducted 8 to 24 months after treatment completion (n = 192). Results The method of MTX delivery and corticosteroid assignment were unrelated to differences in neurocognitive outcomes after controlling for ethnicity, race, age, gender, insurance status, and time off treatment; however, survivors who were age < 10 years at diagnosis (n = 89) had significantly lower estimated IQ ( P < .001) and PS scores ( P = .02) compared with participants age ≥ 10 years. In addition, participants who were covered by US public health insurance had estimated IQs that were significantly lower ( P < .001) than those with US private or military insurance. Conclusion Children with high-risk B-lineage ALL who were age < 10 years at diagnosis are at risk for deficits in IQ and PS in the absence of cranial radiation, regardless of MTX delivery or corticosteroid type. These data may serve as a basis for developing screening protocols to identify children who are at high risk for deficits so that early intervention can be initiated to mitigate the impact of therapy on neurocognitive outcomes.


Cancer | 1990

Surgery only for the treatment of patients with stage I (Cassady) Wilms' tumor

Eric Larsen; Antonio R. Perez-Atayde; Daniel M. Green; Alan B. Retik; Luis A. Clavell; Stephen E. Sallan


Journal of Clinical Oncology | 2017

Reply to I.J. Cohen

Kristina K. Hardy; Leanne Embry; John A. Kairalla; Shanjun Helian; Meenakshi Devidas; F. Daniel Armstrong; Stephen P. Hunger; William L. Carroll; Eric Larsen; Elizabeth A. Raetz; Mignon L. Loh; Wenjian Yang; Mary V. Relling; Robert B. Noll; Naomi J. Winick

Collaboration


Dive into the Eric Larsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mignon L. Loh

Children's Oncology Group

View shared research outputs
Top Co-Authors

Avatar

Naomi J. Winick

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenjian Yang

University Of Tennessee System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William E. Evans

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge