Searched for: in-biosketch:true
person:raetze01
The addition of sirolimus to tacrolimus/methotrexate GVHD prophylaxis in children with ALL: a phase 3 Children's Oncology Group/Pediatric Blood and Marrow Transplant Consortium trial
Pulsipher, Michael A; Langholz, Bryan; Wall, Donna A; Schultz, Kirk R; Bunin, Nancy; Carroll, William L; Raetz, Elizabeth; Gardner, Sharon; Gastier-Foster, Julie M; Howrie, Denise; Goyal, Rakesh K; Douglas, James G; Borowitz, Michael; Barnes, Yvonne; Teachey, David T; Taylor, Candace; Grupp, Stephan A
Sirolimus has activity against acute lymphoblastic leukemia (ALL) in xenograft models and efficacy in preventing acute graft-versus-host disease (aGVHD). We tested whether addition of sirolimus to GVHD prophylaxis of children with ALL would decrease aGVHD and relapse. Patients were randomized to tacrolimus/methotrexate (standard) or tacrolimus/methotrexate/sirolimus (experimental). The study met futility rules for survival after enrolling 146 of 259 patients. Rate of Grade 2-4 aGVHD was 31% vs 18% (standard vs experimental, P = .04), however, grade 3-4 aGVHD was not different (13% vs 10%, P = .28). Rates of veno-occlusive disease (VOD) and thrombotic microangiopathy (TMA) were lower in the nonsirolimus arm (9% vs 21% VOD, P = .05; 1% vs 10% TMA, P = .06). At 2 years, event free survival (EFS) and overall survival (OS) were 56% vs 46%, and 65% vs 55% (standard vs experimental), respectively (P = .28 and .23). Multivariate analysis showed increased relapse risk in children with >/=0.1% minimal residual disease (MRD) pretransplant, and decreased risk in patients with grades 1-3 aGVHD (P = .04). Grades 1-3 aGVHD were associated with improved EFS (P = .02), whereas grade 4 aGVHD and extramedullary disease at diagnosis led to inferior OS. Although addition of sirolimus decreased aGVHD, survival was not improved. This study is registered with ClinicalTrials.gov as #NCT00382109.
PMCID:3968388
PMID: 24497539
ISSN: 0006-4971
CID: 1029002
Refining prognosis in BCR-ABL1-positive ALL [Comment]
Raetz, Elizabeth A; Carroll, William L
PMID: 24627518
ISSN: 0006-4971
CID: 945782
Inherited GATA3 variants are associated with Ph-like childhood acute lymphoblastic leukemia and risk of relapse
Perez-Andreu, Virginia; Roberts, Kathryn G; Harvey, Richard C; Yang, Wenjian; Cheng, Cheng; Pei, Deqing; Xu, Heng; Gastier-Foster, Julie; E, Shuyu; Lim, Joshua Yew-Suang; Chen, I-Ming; Fan, Yiping; Devidas, Meenakshi; Borowitz, Michael J; Smith, Colton; Neale, Geoffrey; Burchard, Esteban G; Torgerson, Dara G; Klussmann, Federico Antillon; Villagran, Cesar Rolando Najera; Winick, Naomi J; Camitta, Bruce M; Raetz, Elizabeth; Wood, Brent; Yue, Feng; Carroll, William L; Larsen, Eric; Bowman, W Paul; Loh, Mignon L; Dean, Michael; Bhojwani, Deepa; Pui, Ching-Hon; Evans, William E; Relling, Mary V; Hunger, Stephen P; Willman, Cheryl L; Mullighan, Charles G; Yang, Jun J
Recent genomic profiling of childhood acute lymphoblastic leukemia (ALL) identified a high-risk subtype with an expression signature resembling that of Philadelphia chromosome-positive ALL and poor prognosis (Ph-like ALL). However, the role of inherited genetic variation in Ph-like ALL pathogenesis remains unknown. In a genome-wide association study (GWAS) of 511 ALL cases and 6,661 non-ALL controls, we identified a susceptibility locus for Ph-like ALL (GATA3, rs3824662; P = 2.17 x 10(-14), odds ratio (OR) = 3.85 for Ph-like ALL versus non-ALL; P = 1.05 x 10(-8), OR = 3.25 for Ph-like ALL versus non-Ph-like ALL), with independent validation. The rs3824662 risk allele was associated with somatic lesions underlying Ph-like ALL (CRLF2 rearrangement, JAK gene mutation and IKZF1 deletion) and with variation in GATA3 expression. Finally, genotype at the GATA3 SNP was also associated with early treatment response and risk of ALL relapse. Our results provide insights into interactions between inherited and somatic variants and their role in ALL pathogenesis and prognosis.
PMCID:4039076
PMID: 24141364
ISSN: 1061-4036
CID: 703672
Intrachromosomal Amplification of Chromosome 21 Is Associated With Inferior Outcomes in Children With Acute Lymphoblastic Leukemia Treated in Contemporary Standard-Risk Children's Oncology Group Studies: A Report From the Children's Oncology Group
Heerema, Nyla A; Carroll, Andrew J; Devidas, Meenakshi; Loh, Mignon L; Borowitz, Michael J; Gastier-Foster, Julie M; Larsen, Eric C; Mattano, Leonard A Jr; Maloney, Kelly W; Willman, Cheryl L; Wood, Brent L; Winick, Naomi J; Carroll, William L; Hunger, Stephen P; Raetz, Elizabeth A
PURPOSE: Five-year overall survival (OS) for children with B-cell precursor acute lymphoblastic leukemia (B-ALL) exceeds 90% with risk-adapted therapy. Age, initial WBC count, genetic aberrations, and minimal residual disease (MRD) are used for risk stratification. Intrachromosomal amplification of a region of chromosome 21 (iAMP21; three or more extra copies of RUNX1 on an abnormal chromosome 21) is a recently identified recurrent genomic lesion associated with inferior outcome in some studies. We investigated the impact of iAMP21 in a large cohort treated in contemporary Children's Oncology Group (COG) ALL trials. PATIENTS AND METHODS: Fluorescent in situ hybridization for specific genetic aberrations was required at diagnosis. MRD was measured by flow cytometry at end induction. Outcome was measured as event-free survival (EFS) and OS. RESULTS: iAMP21 was found in 158 (2%) of 7,793 patients with B-ALL age >/= 1 year; 74 (1.5%) of 5,057 standard-risk (SR) patients, and 84 (3.1%) of 2,736 high-risk (HR) patients. iAMP21 was associated with age >/= 10 years, WBC less than 50,000/muL, female sex, and detectable MRD at day 29. Four-year EFS and OS were significantly worse for patients with iAMP21 and SR B-ALL, but iAMP21 was not a statistically significant prognostic factor in HR patients. There was no interaction between MRD and iAMP21. Among SR patients, day 29 MRD >/= 0.01% and iAMP21 were associated with the poorest EFS and OS; absence of both was associated with the best outcome. CONCLUSION: iAMP21 is associated with inferior outcome in pediatric B-ALL, particularly SR patients who require more intensive therapy and are now treated on HR COG ALL protocols.
PMCID:3770866
PMID: 23940221
ISSN: 0732-183x
CID: 540832
OUTCOME FOR CHILDREN WITH HYPODIPLOID ACUTE LYMPHOBLASTIC LEUKEMIA (ALL) ON CONTEMPORARY CHILDREN'S ONCOLOGY GROUP (COG) CLINICAL TRIALS [Meeting Abstract]
Devidas, M.; Raetz, E. A.; Loh, M. L.; Carroll, A. J.; Hereema, N. A.; Gastier-Foster, J.; Borowitz, M. J.; Wood, B.; Larsen, E.; Maloney, K. W.; Mattano, L.; Winick, N. J.; Hunger, S. P.; Carroll, W. L.
ISI:000324735500035
ISSN: 1545-5009
CID: 627532
IMPROVEMENTS IN OVERALL SURVIVAL (OS) IN CHILDREN WITH NEWLY DIAGNOSED ACUTE LYMPHOBLASTIC LEUKEMIA (ALL) ENROLLED IN CHILDREN'S ONCOLOGY GROUP (COG) TRIALS IN 2000-05 VS. 2006-09 [Meeting Abstract]
Hunger, S.; Devidas, M.; Raetz, E.; Loh, M.; Winick, N.; Carroll, W.; Lu, X.
ISI:000324735500033
ISSN: 1545-5009
CID: 627542
Erwinia asparaginase achieves therapeutic activity after pegaspargase allergy: a report from the Children's Oncology Group
Salzer, Wanda L; Asselin, Barbara; Supko, Jeffrey G; Devidas, Meenakshi; Kaiser, Nicole A; Plourde, Paul; Winick, Naomi J; Reaman, Gregory H; Raetz, Elizabeth; Carroll, William L; Hunger, Stephen P
Children's Oncology Group AALL07P2 tested whether substitution of Erwinia asparaginase 25,000-IU/m2 for 6 doses intramuscularly (IM) given Monday/Wednesday/Friday to children and young adults with acute lymphoblastic leukemia (ALL) and clinical allergy to pegaspargase would provide a 48-hour nadir serum asparaginase activity (NSAA) >/=0.10-IU/mL in at least 70% of patients. AALL07P2 enrolled 55 eligible/evaluable patients. NSAA >/=0.1-IU/mL was achieved in 38/41 patients (92.7%) with samples meeting acceptability criteria 48-hours after dosing and in 38/43 patients (88.4%) 72-hours after dosing during course 1. Among acceptable samples obtained during all therapy courses, 95.8% (252/263) of 48-hour samples and 84.5% (125/148) of 72-hour samples had NSAA >/=0.10-IU/mL. Pharmacokinetic parameters were estimated by fitting the serum asparaginase activity-time course for all 6 doses given during course 1 to a one compartment open model with first order absorption. Erwinia asparaginase administered with this schedule achieved therapeutic NSAA at both 48- and 72-hours and was well tolerated with no reports of hemorrhage, thrombosis, or death, and few cases of grade 2-3 allergic reaction (n=6), grade 1-3 hyperglycemia (n=6), and grade 1 pancreatitis (n=1). Following allergy to pegaspargase, Erwinia asparaginase 25,000-IU/m2 x 6 doses IM Monday/Wednesday/Friday for 2 weeks can be substituted for a single dose of pegaspargase.
PMCID:3724190
PMID: 23741010
ISSN: 0006-4971
CID: 453372
Phase I/II trial of clofarabine and cytarabine in children with relapsed/refractory acute lymphoblastic leukemia (AAML0523): A report from the Children's Oncology Group
Cooper, Todd M; Razzouk, Bassem I; Gerbing, Robert; Alonzo, Todd A; Adlard, Kathleen; Raetz, Elizabeth; Gamis, Alan S; Perentesis, John; Whitlock, James A
BACKGROUND: The discovery of effective re-induction regimens for children with more than one relapse of acute lymphoblastic leukemia (ALL) remains elusive. The novel nucleoside analog clofarabine exhibits modest single agent efficacy in relapsed ALL, though optimal combinations of this agent with other active chemotherapy drugs have not yet been defined. Herein we report the response rates of relapsed ALL patients treated on Children's Oncology Group study AAML0523, a Phase I/II study of the combination of clofarabine and cytarabine. PROCEDURE: AAML0523 enrolled 21 children with ALL in second or third relapse, or those refractory to re-induction therapy. The study consisted of two phases: a dose finding phase and an efficacy phase. The dose finding portion consisted of a single dose escalation/de-escalation of clofarabine for 5 days in combination with a fixed dose of cytarabine (1 g/m(2) /day for 5 days). Eight patients received clofarabine at 40 mg/m(2) /day and 13 patients at 52 mg/m(2) /day. RESULTS: Toxicities observed at all doses of clofarabine were typical of intensive chemotherapy regimens for leukemia, with infection being the most common. We did not observe significant hepatotoxicity as reported in other clofarabine combination regimens. The recommended pediatric Phase II dose of clofarabine in combination with cytarabine for the efficacy portion of AAML0523 was 52 mg/m(2) . Of 21 patients with ALL, 3 (14%) achieved a complete response (CR). Based on the two-stage design definition of first-stage inactivity, the therapy was deemed ineffective. CONCLUSION: The combination of clofarabine and cytarabine in relapsed/refractory childhood ALL does not warrant further clinical investigation. Pediatr Blood Cancer (c) 2013 Wiley Periodicals, Inc.
PMCID:4605828
PMID: 23335239
ISSN: 1545-5009
CID: 222682
Children's Oncology Group's 2013 blueprint for research: acute lymphoblastic leukemia
Hunger, Stephen P; Loh, Mignon L; Whitlock, James A; Winick, Naomi J; Carroll, William L; Devidas, Meenakshi; Raetz, Elizabeth A
Approximately 90% of the 2,000 children, adolescents, and young adults enrolled each year in Children's Oncology Group acute lymphoblastic leukemia (ALL) trials will be cured. However, high-risk subsets with significantly inferior survival remain, including infants, newly diagnosed patients with age >/=10 years, white blood count >/=50,000/microl, poor early response or T-cell ALL, and relapsed ALL patients. Effective strategies to improve survival include better risk stratification, optimizing standard chemotherapy and combining targeted therapies with cytotoxic chemotherapy, the latter of which is dependent upon identification of key driver mutations present in ALL. Pediatr Blood Cancer (c) 2012 Wiley Periodicals, Inc.
PMCID:4045498
PMID: 23255467
ISSN: 1545-5009
CID: 222702
Lymphoproliferative disorder that resembles heptosplenic lymphoma during maintenance treatment for T-cell acute lymphoblastic leukemia
Hu, Youjun; Ahmed, Tahmeena; Zaslav, Ann-Leslie; Golightly, Marc; Spitzer, Silvia G; Raetz, Elizabeth; Chan, Edward L
A 6-year-old male presented with a testicular mass, hepatosplenomegaly, and a pleural effusion while undergoing maintenance chemotherapy for treatment of T-cell acute lymphoblastic leukemia (T-ALL). He was subsequently diagnosed with a lymphoproliferative disorder that resembled hepatosplenic lymphoma (HSL). While the extranodal presentation and the protracted yet aggressive clinical course are consistent with HSL, the findings of monosomy 8 and polymorphic cell populations are unique and have not been previously described in this type of lymphoma. Pediatr Blood Cancer (c) 2013 Wiley Periodicals, Inc.
PMID: 23303724
ISSN: 1545-5009
CID: 222692