Try a new search

Format these results:

Searched for:

in-biosketch:true

person:raetze01

Total Results:

279


Dasatinib Maintains Outstanding 5-Year Survival Outcomes in Children with PH plus ALL, but does not Prevent CNS Relapses: Children's Oncology Group (COG) AALL0622 Trial [Meeting Abstract]

Slayton, W; Schultz, K; Kairalla, J; Meenakshi, D; Pulsipher, M; Silverman, L; Borowitz, M; Carroll, A; Heerema, N; Gastier-Foster, J; Mizrahy, S; Wood, B; Merchant, T; Brown, V; Raetz, E; Winick, N; Loh, M; Carroll, W; Hunger, S
ISI:000408978201028
ISSN: 1545-5017
CID: 2767042

Osteonecrosis (ON) is Associated with Improved Event Free Survival (EFS) in High-Risk Acute Lymphoblastic Leukemia (HR-ALL): Results of Children's Oncology Group (COG) Study AALL0232 [Meeting Abstract]

Mattano, LA; Devidas, M; Chen, S; Raetz, E; Loh, M; Winick, N; Hunger, SP; Carroll, WL; Larsen, E
ISI:000408978201029
ISSN: 1545-5017
CID: 2767012

How is the Ph-like signature being incorporated into ALL therapy?

Maese, Luke; Tasian, Sarah K; Raetz, Elizabeth A
Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a recently identified high risk disease subtype characterized by a gene expression profile similar to that observed in Philadelphia chromosome-positive (Ph-positive) ALL, but without an underlying BCR-ABL1 translocation. Adults and children with Ph-like ALL harbor a diversity of alterations that all lead to activated kinase signaling. Outcomes for patients with Ph-like ALL are poor, which has prompted investigation into the role of tyrosine kinase inhibitor (TKI)-based therapies for this disease. Several clinical trials are now ongoing that include screening for the Ph-like signature and treatment of patients with Ph-like ALL with TKI therapy. This review examines how testing for Ph-like ALL is being incorporated into clinical trials.
PMCID:6053910
PMID: 29050695
ISSN: 1532-1924
CID: 2927202

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

Hardy, Kristina K; Embry, Leanne; Kairalla, John A; Helian, Shanjun; Devidas, Meenakshi; Armstrong, Daniel; Hunger, Stephen; Carroll, William L; Larsen, Eric; Raetz, Elizabeth A; Loh, Mignon L; Yang, Wenjian; Relling, Mary V; Noll, Robert B; Winick, Naomi
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.
PMCID:5549456
PMID: 28671857
ISSN: 1527-7755
CID: 2617182

Impact of Initial CSF Findings on Outcome Among Patients With National Cancer Institute Standard- and High-Risk B-Cell Acute Lymphoblastic Leukemia: A Report From the Children's Oncology Group

Winick, Naomi; Devidas, Meenakshi; Chen, Si; Maloney, Kelly; Larsen, Eric; Mattano, Leonard; Borowitz, Michael J; Carroll, Andrew; Gastier-Foster, Julie M; Heerema, Nyla A; Willman, Cheryl; Wood, Brent; Loh, Mignon L; Raetz, Elizabeth; Hunger, Stephen P; Carroll, William L
Purpose To determine the prognostic significance of blasts, and of white and red blood cells, in CSF samples at diagnosis of acute lymphoblastic leukemia (ALL), a uniform CSF and risk group classification schema was incorporated into Children's Oncology Group B-cell ALL (B-ALL) clinical trials. Methods CSF status was designated as follows: CNS1, no blasts; CNS2a to 2c, < 5 WBCs/muL and blasts with/without >/= 10 RBCs/muL or >/= 5 WBCs/muL plus blasts, with WBCs >/= 5 times the number of RBCs; CNS3a to 3c, >/= 5 WBCs/muL plus blasts with/without >/= 10 RBCs/muL or clinical signs of CNS disease. CNS2 status did not affect therapy; patients with CNS3 status received two extra intrathecal treatments during induction and augmented postinduction therapy with 18 Gy of cranial radiation. Results Among 8,379 evaluable patients enrolled from 2004 to 2010, 7,395 (88.3%) had CNS1 status; 857 (10.2%), CNS2; and 127 (1.5%), CNS3. The 5-year event-free and overall survival rates were, respectively, 85% and 92.7% for CNS1, 76% and 86.8% for CNS2, and 76% and 82.1% for CNS3 ( P < .001). In multivariable analysis that included age, race/ethnicity, initial WBC, and day-29 minimal residual disease < 0.1%, CSF blast, regardless of cell count, was an independent adverse predictor of outcome for patients with standard- or high-risk disease according to National Cancer Institute criteria. The EFS difference reflected a significant difference in the incidence of CNS, not marrow, relapse in patients with CNS1 versus CNS2 and/or CNS3 status. Conclusion Low levels of CNS leukemia, regardless of RBCs, predict inferior outcome and higher rates of CNS relapse. These data suggest that additional augmentation of CNS-directed therapy is warranted for CNS2 disease.
PMCID:5536164
PMID: 28535084
ISSN: 1527-7755
CID: 2574752

The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia

Liu, Yu; Easton, John; Shao, Ying; Maciaszek, Jamie; Wang, Zhaoming; Wilkinson, Mark R; McCastlain, Kelly; Edmonson, Michael; Pounds, Stanley B; Shi, Lei; Zhou, Xin; Ma, Xiaotu; Sioson, Edgar; Li, Yongjin; Rusch, Michael; Gupta, Pankaj; Pei, Deqing; Cheng, Cheng; Smith, Malcolm A; Auvil, Jaime Guidry; Gerhard, Daniela S; Relling, Mary V; Winick, Naomi J; Carroll, Andrew J; Heerema, Nyla A; Raetz, Elizabeth; Devidas, Meenakshi; Willman, Cheryl L; Harvey, Richard C; Carroll, William L; Dunsmore, Kimberly P; Winter, Stuart S; Wood, Brent L; Sorrentino, Brian P; Downing, James R; Loh, Mignon L; Hunger, Stephen P; Zhang, Jinghui; Mullighan, Charles G
Genetic alterations that activate NOTCH1 signaling and T cell transcription factors, coupled with inactivation of the INK4/ARF tumor suppressors, are hallmarks of T-lineage acute lymphoblastic leukemia (T-ALL), but detailed genome-wide sequencing of large T-ALL cohorts has not been carried out. Using integrated genomic analysis of 264 T-ALL cases, we identified 106 putative driver genes, half of which had not previously been described in childhood T-ALL (for example, CCND3, CTCF, MYB, SMARCA4, ZFP36L2 and MYCN). We describe new mechanisms of coding and noncoding alteration and identify ten recurrently altered pathways, with associations between mutated genes and pathways, and stage or subtype of T-ALL. For example, NRAS/FLT3 mutations were associated with immature T-ALL, JAK3/STAT5B mutations in HOXA1 deregulated ALL, PTPN2 mutations in TLX1 deregulated T-ALL, and PIK3R1/PTEN mutations in TAL1 deregulated ALL, which suggests that different signaling pathways have distinct roles according to maturational stage. This genomic landscape provides a logical framework for the development of faithful genetic models and new therapeutic approaches.
PMCID:5535770
PMID: 28671688
ISSN: 1546-1718
CID: 2617152

Genome-wide study links PNPLA3 variant with elevated hepatic transaminase after acute lymphoblastic leukemia therapy

Liu, Yiwei; Fernandez, Christian A; Smith, Colton; Yang, Wenjian; Cheng, Cheng; Panetta, John C; Kornegay, Nancy; Liu, Chengcheng; Ramsey, Laura B; Karol, Seth E; Janke, Laura J; Larsen, Eric C; Winick, Naomi; Carroll, William L; Loh, Mignon L; Raetz, Elizabeth A; Hunger, Stephen P; Devidas, Meenakshi; Yang, Jun J; Mullighan, Charles G; Zhang, Jinghui; Evans, William E; Jeha, Sima; Pui, Ching-Hon; Relling, Mary V
Remission induction therapy for acute lymphoblastic leukemia (ALL) includes medications that may cause hepatotoxicity, including asparaginase. We used a genome-wide association study (GWAS) to identify loci associated with elevated alanine transaminase (ALT) levels after induction therapy in children with ALL enrolled on St. Jude Children's Research Hospital (SJCRH) protocols. Germline DNA was genotyped using arrays and exome sequencing. Adjusting for age, body mass index, ancestry, asparaginase preparation and dosage, the PNPLA3 rs738409 (C>G) I148M variant, previously associated with fatty liver disease risk, had the strongest genetic association with ALT (P = 2.5x10-8 ). The PNPLA3 rs738409 variant explained 3.8% of the variability in ALT, and partly explained race-related differences in ALT. The PNPLA3 rs738409 association was replicated in an independent cohort of 2,285 patients treated on Children's Oncology Group protocol AALL0232 (P = 0.024). This is an example of a pharmacogenetic variant overlapping with a disease risk variant
PMCID:5511775
PMID: 28090653
ISSN: 1532-6535
CID: 2413722

Targetable kinase gene fusions in high risk B-ALL: a study from the Children's Oncology Group

Reshmi, Shalini C; Harvey, Richard C; Roberts, Kathryn G; Stonerock, Eileen; Smith, Amy; Jenkins, Heather; Chen, I-Ming; Valentine, Marc; Liu, Yu; Li, Yongjin; Shao, Ying; Easton, John; Payne-Turner, Debbie; Gu, Zhaohui; Tran, Thai Hoa; Nguyen, Jonathan V; Devidas, Meenakshi; Dai, Yunfeng; Heerema, Nyla A; Carroll, Andrew J 3rd; Raetz, Elizabeth A; Borowitz, Michael J; Wood, Brent L; Angiolillo, Anne L; Burke, Michael J; Salzer, Wanda L; Zweidler-McKay, Patrick A; Rabin, Karen R; Carroll, William L; Zhang, Jinghui; Loh, Mignon L; Mullighan, Charles G; Willman, Cheryl L; Gastier-Foster, Julie M; Hunger, Stephen P
Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a high-risk subtype characterized by genomic alterations that activate cytokine receptor and kinase signaling. We examined the frequency and spectrum of targetable genetic lesions in a retrospective cohort of 1389 consecutively diagnosed childhood B-ALL patients with high-risk clinical features and/or elevated minimal residual disease at the end of remission induction therapy. The Ph-like gene expression profile was identified in 341 of 1389 patients, 57 of which were excluded from additional analysis because of the presence of BCR-ABL1 (n=46) or ETV6-RUNX1 (n=11). Among the remaining 284 (20.4%) patients, overexpression and rearrangement of CRLF2 (IGH-CRLF2 or P2RY8-CRLF2) were identified in 124 (43.7%), with concomitant genomic alterations activating the JAK-STAT pathway (JAK1, JAK2, IL7R) identified in 63 patients (50.8% of CRLF2-rearranged cases). Of the remaining patients, using RT-PCR or transcriptome sequencing, we identified targetable ABL-class fusions (ABL1, ABL2, CSF1R and PDGFRB) in 14.1% of Ph-like ALL cases, EPOR rearrangements or JAK2 fusions (8.8%), alterations activating other JAK-STAT signaling genes (IL7R, SH2B3, JAK1; 6.3%) and other kinases (FLT3, NTRK3, LYN; 4.6%), and mutations involving the Ras pathway (KRAS, NRAS, NF1, PTPN11; 6%). We identified eight new rearrangement partners for four kinase genes previously reported to be rearranged in Ph-like ALL. The current findings provide support for the precision-medicine testing and treatment approach for Ph-like ALL that has been implemented in Children's Oncology Group ALL trials.
PMCID:5482101
PMID: 28408464
ISSN: 1528-0020
CID: 2528372

Genetics of ancestry-specific risk for relapse in acute lymphoblastic leukemia

Karol, S E; Larsen, E; Cheng, C; Cao, X; Yang, W; Ramsey, L B; Fernandez, C A; McCorkle, J R; Paugh, S W; Autry, R J; Lopez-Lopez, E; Diouf, B; Jeha, S; Pui, C-H; Raetz, E A; Winick, N J; Carroll, W L; Hunger, S P; Loh, M L; Devidas, M; Evans, W E; Yang, J J; Relling, M V
The causes of individual relapses in children with acute lymphoblastic leukemia (ALL) remain incompletely understood. We evaluated the contribution of germline genetic factors to relapse in 2225 children treated on Children's Oncology Group trial AALL0232. We identified 302 germline single nucleotide polymorphisms (SNPs) associated with relapse after adjusting for treatment and ancestry and 715 additional SNPs associated with relapse in an ancestry-specific manner. We tested for replication of these relapse-associated SNPs in external data sets of antileukemic drug pharmacokinetics and pharmacodynamics and an independent clinical cohort. 224 SNPs were associated with rapid drug clearance or drug resistance, and 32 were replicated in the independent cohort. The adverse risk associated with black and Hispanic ancestries was attenuated by addition of the 4 SNPs most strongly associated with relapse in these populations [for blacks: model without SNPs hazard ratio (HR)=2.32, P=2.27 x 10-4, model with SNPs HR=1.07, P=0.79; for Hispanics: model without SNPs HR=1.7, P=8.23 x 10-5, model with SNPs HR=1.31, P=0.065]. Relapse SNPs associated with asparaginase resistance or allergy were overrepresented among SNPs associated with relapse in the more asparaginase intensive treatment arm (20/54 in Capizzi-methorexate arm vs 8/54 in high-dose methotrexate arm, P=0.015). Inherited genetic variation contributes to race-specific and treatment-specific relapse risk.Leukemia accepted article preview online, 18 January 2017. doi:10.1038/leu.2017.24.
PMCID:5462853
PMID: 28096535
ISSN: 1476-5551
CID: 2413872

Beating the Clock in T-Cell Acute Lymphoblastic Leukemia

Carroll, William L; Aifantis, Iannis; Raetz, Elizabeth A
CDK4/6 inhibition was synergistic with dexmethasome and everolimus but antagonistic with conventional chemotherapy in T-cell acute lymphoblastic leukemia (T-ALL) pre-clinical models. Cyclin dependent kinase inhibition in combination with glucocorticoids and mTOR inhibition offers a unique therapeutic opportunity in T-ALL.
PMID: 28007775
ISSN: 1078-0432
CID: 2374552