Searched for: in-biosketch:true
person:raetze01
Class II HLA Variants Associate with Risk of Pegaspargase Hypersensitivity
Liu, Yiwei; Yang, Wenjian; Smith, Colton; Cheng, Cheng; Karol, Seth E; Larsen, Eric C; Winick, Naomi; Carroll, William L; Loh, Mignon L; Raetz, Elizabeth A; Hunger, Stephen P; Winter, Stuart S; Dunsmore, Kimberly P; Devidas, Meenakshi; Yang, Jun J; Evans, William E; Jeha, Sima; Pui, Ching-Hon; Inaba, Hiroto; Relling, Mary V
We conducted the first HLA allele and genome wide association study to identify loci associated with hypersensitivity reactions exclusively to the PEGylated preparation of asparaginase (pegaspargase) in racially diverse cohorts of pediatric leukemia patients: St. Jude Children's Research Hospital's Total XVI (TXVI, n = 598), Children's Oncology Group AALL0232 (n = 2472) and AALL0434 (n = 1189). Germline DNA was genotyped using arrays. Genetic variants not genotyped directly were imputed. HLA alleles were imputed using SNP2HLA or inferred using BWAkit. Analyses between genetic variants and hypersensitivity were performed in each cohort first using cohort-specific covariates and then combined using meta-analyses. Nongenetic risk factors included fewer intrathecal injections (P = 2.7x10-5 in TXVI) and male sex (P = 0.025 in AALL0232). HLA alleles DQB1*02:02, DRB1*07:01, and DQA1*02:01 had the strongest associations with pegaspargase hypersensitivity (P < 5.0x10-5 ) in patients with primarily European ancestry (EA), with the three alleles associating in a single haplotype. The top allele HLA-DQB1*02:02 was tagged by HLA-DQB1 rs1694129 in EAs (r2 = 0.96) and less so in non-EAs. All single nucleotide polymorphisms associated with pegaspargase hypersensitivity reaching genome-wide significance in EAs were in class II HLA loci, and were partially replicated in non-EAs, as is true for other HLA associations. The rs9958628 variant, in ARHGAP28 (previously linked to immune response in children) had the strongest genetic association (P = 8.9x10-9 ) in non-EAs. The HLA-DQB1*02:02-DRB1*07:01-DQA1*02:01 associated with hypersensitivity reactions to pegaspargase is the same haplotype associated with reactions to non-PEGylated asparaginase, even though the antigens differ between the two preparations.
PMID: 33768542
ISSN: 1532-6535
CID: 4822982
Genome-Wide Association Study of Susceptibility Loci for TCF3-PBX1 Acute Lymphoblastic Leukemia in Children
Lee, Shawn H R; Qian, Maoxiang; Yang, Wentao; Diedrich, Jonathan D; Raetz, Elizabeth; Yang, Wenjian; Dong, Qian; Devidas, Meenakshi; Pei, Deqing; Yeoh, Allen; Cheng, Cheng; Pui, Ching-Hon; Evans, William E; Mullighan, Charles G; Hunger, Stephen P; Savic, Daniel; Relling, Mary V; Loh, Mignon L; Yang, Jun J
Acute lymphoblastic leukemia (ALL) is the most common cancer in children. TCF3-PBX1 fusion defines a common molecular subtype of ALL with unique clinical features, but the molecular basis of its inherited susceptibility is unknown. In a genome-wide association study of 1494 ALL cases and 2057 non-ALL controls, we identified a germline risk locus located in an intergenic region between BCL11A and PAPOLG: rs2665658, P = 1.88 × 10-8 for TCF3-PBX1 ALL vs non-ALL, and P = 1.70 × 10-8 for TCF3-PBX1 ALL vs other-ALL. The lead variant was validated in a replication cohort, and conditional analyses pointed to a single causal variant with subtype-specific effect. The risk variant is located in a regulatory DNA element uniquely activated in ALL cells with the TCF3-PBX1 fusion and may distally modulate the transcription of the adjacent gene REL. Our results expand the understanding of subtype-specific ALL susceptibility and highlight plausible interplay between germline variants and somatic genomic abnormalities in ALL pathogenesis.
PMCID:8487647
PMID: 32882024
ISSN: 1460-2105
CID: 5171722
Severe toxicity free survival: physician-derived definitions of unacceptable long-term toxicities following acute lymphocytic leukaemia
Andrés-Jensen, Liv; Attarbaschi, Andishe; Bardi, Edit; Barzilai-Birenboim, Shlomit; Bhojwani, Deepa; Hagleitner, Melanie M; Halsey, Christina; Harila-Saari, Arja; van Litsenburg, Raphaele R L; Hudson, Melissa M; Jeha, Sima; Kato, Motohiro; Kremer, Leontien; Mlynarski, Wojciech; Möricke, Anja; Pieters, Rob; Piette, Caroline; Raetz, Elizabeth; Ronceray, Leila; Toro, Claudia; Grazia Valsecchi, Maria; Vrooman, Lynda M; Weinreb, Sigal; Winick, Naomi; Schmiegelow, Kjeld
5-year overall survival rates have surpassed 90% for childhood acute lymphocytic leukaemia, but survivors are at risk for permanent health sequelae. Although event-free survival appropriately represents the outcome for cancers with poor overall survival, this metric is inadequate when cure rates are high but challenged by serious, persistent complications. Accordingly, a group of experts in paediatric haematology-oncology, representative of 17 international acute lymphocytic leukaemia study groups, launched an initiative to construct a measure, designated severe toxicity-free survival (STFS), to quantify the occurrence of physician-prioritised toxicities to be integrated with standard cancer outcome reporting. Five generic inclusion criteria (not present before cancer diagnosis, symptomatic, objectifiable, of unacceptable severity, permanent, or requiring unacceptable treatments) were used to assess 855 health conditions, which resulted in inclusion of 21 severe toxicities. Consensus definitions were reached through a modified Delphi process supplemented by two additional plenary meetings. The 21 severe toxicities include severe adverse health conditions that substantially affect activities of daily living and are refractory to therapy (eg, refractory seizures), are without therapeutic options (eg, blindness), or require substantially invasive treatment (eg, cardiac transplantation). Incorporation of STFS assessment into clinical trials has the potential to improve and diversify treatment strategies, focusing not only on traditional outcome events and overall survival but also the frequencies of the most severe toxicities. The two major aims of this Review were to: prioritise and define unacceptable long-term toxicity for patients with childhood acute lymphocytic leukaemia, and define how these toxicities should be combined into a composite quantity to be integrated with other reported outcomes. Although STFS quantifies the clinically unacceptable health tradeoff for cure using childhood acute lymphocytic leukaemia as a model disease, the prioritised severe toxicities are based on generic considerations of relevance to any other cancer diagnosis and age group.
PMID: 34171282
ISSN: 2352-3026
CID: 4925792
Germline RUNX1 variation and predisposition to childhood acute lymphoblastic leukemia
Li, Yizhen; Yang, Wentao; Devidas, Meenakshi; Winter, Stuart S; Kesserwan, Chimene; Yang, Wenjian; Dunsmore, Kimberly P; Smith, Colton; Qian, Maoxiang; Zhao, Xujie; Zhang, Ranran; Gastier-Foster, Julie M; Raetz, Elizabeth A; Carroll, William L; Li, Chunliang; Liu, Paul P; Rabin, Karen R; Sanda, Takaomi; Mullighan, Charles G; Nichols, Kim E; Evans, William E; Pui, Ching-Hon; Hunger, Stephen P; Teachey, David T; Relling, Mary V; Loh, Mignon L; Yang, Jun J
Genetic alterations in the RUNX1 gene are associated with benign and malignant blood disorders, particularly of megakaryocyte and myeloid lineages. The role of RUNX1 in acute lymphoblastic leukemia (ALL) is less clear, particularly how germline genetic variation influences the predisposition to this type of leukemia. Sequencing 4,836 children with B-ALL and 1,354 cases of T-ALL, we identified 31 and 18 germline RUNX1 variants, respectively. RUNX1 variants in B-ALL consistently showed minimal damaging effects. By contrast, 6 T-ALL-related variants result in drastic loss of RUNX1 activity as a transcription activator in vitro. Ectopic expression of dominant-negative RUNX1 variants in human CD34+ cells repressed differentiation into erythroid, megakaryocytes, and T cells, while promoting myeloid cell development. Chromatin immunoprecipitation sequencing of T-ALL models showed distinctive patterns of RUNX1 binding by variant proteins. Further whole genome sequencing identified JAK3 mutation as the most frequent somatic genomic abnormality in T-ALL with germline RUNX1 variants. Co-introduction of RUNX1 variant and JAK3 mutation in hematopoietic stem and progenitor cells in mice gave rise to T-ALL with early T-cell precursor phenotype. Taken together, these results indicated that RUNX1 is an important predisposition gene for T-ALL and pointed to novel biology of RUNX1-mediated leukemogenesis in the lymphoid lineages.
PMID: 34166225
ISSN: 1558-8238
CID: 4916812
Correction to: FLT3 inhibitor lestaurtinib plus chemotherapy for newly diagnosed KMT2A-rearranged infant acute lymphoblastic leukemia: Children's Oncology Group trial AALL0631
Brown, Patrick A; Kairalla, John A; Hilden, Joanne M; Dreyer, ZoAnn E; Carroll, Andrew J; Heerema, Nyla A; Wang, Cindy; Devidas, Meenakshi; Gore, Lia; Salzer, Wanda L; Winick, Naomi J; Carroll, William L; Raetz, Elizabeth A; Borowitz, Michael J; Small, Donald; Loh, Mignon L; Hunger, Stephen P
PMID: 33846544
ISSN: 1476-5551
CID: 4845822
FLT3 inhibitor lestaurtinib plus chemotherapy for newly diagnosed KMT2A-rearranged infant acute lymphoblastic leukemia: Children's Oncology Group trial AALL0631
Brown, Patrick A; Kairalla, John A; Hilden, Joanne M; Dreyer, ZoAnn E; Carroll, Andrew J; Heerema, Nyla A; Wang, Cindy; Devidas, Meenakshi; Gore, Lia; Salzer, Wanda L; Winick, Naomi J; Carroll, William L; Raetz, Elizabeth A; Borowitz, Michael J; Small, Donald; Loh, Mignon L; Hunger, Stephen P
Infants with KMT2A-rearranged acute lymphoblastic leukemia (KMT2A-r ALL) have a poor prognosis. KMT2A-r ALL overexpresses FLT3, and the FLT3 inhibitor (FLT3i) lestaurtinib potentiates chemotherapy-induced cytotoxicity in preclinical models. Children's Oncology Group (COG) AALL0631 tested whether adding lestaurtinib to post-induction chemotherapy improved event-free survival (EFS). After chemotherapy induction, KMT2A-r infants received either chemotherapy only or chemotherapy plus lestaurtinib. Correlative assays included FLT3i plasma pharmacodynamics (PD), which categorized patients as inhibited or uninhibited, and FLT3i ex vivo sensitivity (EVS), which categorized leukemic blasts as sensitive or resistant. There was no difference in 3-year EFS between patients treated with chemotherapy plus lestaurtinib (n = 67, 36 ± 6%) vs. chemotherapy only (n = 54, 39 ± 7%, p = 0.67). However, for the lestaurtinib-treated patients, FLT3i PD and FLT3i EVS significantly correlated with EFS. For FLT3i PD, EFS for inhibited/uninhibited was 59 ± 10%/28 ± 7% (p = 0.009) and for FLTi EVS, EFS for sensitive/resistant was 52 ± 8%/5 ± 5% (p < 0.001). Seventeen patients were both inhibited and sensitive, with an EFS of 88 ± 8%. Adding lestaurtinib did not improve EFS overall, but patients achieving potent FLT3 inhibition and those whose leukemia blasts were sensitive FLT3-inhibition ex vivo did benefit from the addition of lestaurtinib. Patient selection and PD-guided dose escalation may enhance the efficacy of FLT3 inhibition for KMT2A-r infant ALL.
PMID: 33623141
ISSN: 1476-5551
CID: 4794562
Minimal Residual Disease in Acute Lymphoblastic Leukemia: Current Practice and Future Directions
Contreras Yametti, Gloria Paz; Ostrow, Talia H; Jasinski, Sylwia; Raetz, Elizabeth A; Carroll, William L; Evensen, Nikki A
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer and advances in its clinical and laboratory biology have grown exponentially over the last few decades. Treatment outcome has improved steadily with over 90% of patients surviving 5 years from initial diagnosis. This success can be attributed in part to the development of a risk stratification approach to identify those subsets of patients with an outstanding outcome that might qualify for a reduction in therapy associated with fewer short and long term side effects. Likewise, recognition of patients with an inferior prognosis allows for augmentation of therapy, which has been shown to improve outcome. Among the clinical and biological variables known to impact prognosis, the kinetics of the reduction in tumor burden during initial therapy has emerged as the most important prognostic variable. Specifically, various methods have been used to detect minimal residual disease (MRD) with flow cytometric and molecular detection of antigen receptor gene rearrangements being the most common. However, many questions remain as to the optimal timing of these assays, their sensitivity, integration with other variables and role in treatment allocation of various ALL subgroups. Importantly, the emergence of next generation sequencing assays is likely to broaden the use of these assays to track disease evolution. This review will discuss the biological basis for utilizing MRD in risk assessment, the technical approaches and limitations of MRD detection and its emerging applications.
PMCID:8069391
PMID: 33924381
ISSN: 2072-6694
CID: 4853632
Favorable Trisomies and ETV6-RUNX1 Predict Cure in Low-Risk B-Cell Acute Lymphoblastic Leukemia: Results From Children's Oncology Group Trial AALL0331
Mattano, Leonard A; Devidas, Meenakshi; Maloney, Kelly W; Wang, Cindy; Friedmann, Alison M; Buckley, Patrick; Borowitz, Michael J; Carroll, Andrew J; Gastier-Foster, Julie M; Heerema, Nyla A; Kadan-Lottick, Nina S; Matloub, Yousif H; Marshall, David T; Stork, Linda C; Loh, Mignon L; Raetz, Elizabeth A; Wood, Brent L; Hunger, Stephen P; Carroll, William L; Winick, Naomi J
PURPOSE/OBJECTIVE:Children's Oncology Group (COG) AALL0331 tested whether pegaspargase intensification on a low-intensity chemotherapy backbone would improve the continuous complete remission (CCR) rate in a low-risk subset of children with standard-risk B-acute lymphoblastic leukemia (ALL). METHODS:fusion or simultaneous trisomies of chromosomes 4, 10, and 17). Random assignment was to standard COG low-intensity therapy (including two pegaspargase doses, one each during induction and delayed intensification) with or without four additional pegaspargase doses at 3-week intervals during consolidation and interim maintenance. The study was powered to detect a 4% improvement in 6-year CCR rate from 92% to 96%. RESULTS:< .0001). CONCLUSION/CONCLUSIONS:Standard COG therapy without intensified pegaspargase, which can easily be given as an outpatient with limited toxicity, cures nearly all children with B-ALL identified as low-risk by clinical, early response, and favorable cytogenetic criteria.
PMID: 33739852
ISSN: 1527-7755
CID: 4818142
Genetics of osteonecrosis in pediatric acute lymphoblastic leukemia and general populations
Yang, Wenjian; Devidas, Meenakshi; Liu, Yiwei; Smith, Colton; Dai, Yunfeng; Winick, Naomi; Hunger, Stephen P; Loh, Mignon L; Raetz, Elizabeth A; Larsen, Eric; Carroll, William L; Winter, Stuart S; Dunsmore, Kimberly Panter; Mattano, Leonard A; Relling, Mary V; Karol, Seth E
PMID: 33106839
ISSN: 1528-0020
CID: 4646452
Effect of Postreinduction Therapy Consolidation With Blinatumomab vs Chemotherapy on Disease-Free Survival in Children, Adolescents, and Young Adults With First Relapse of B-Cell Acute Lymphoblastic Leukemia: A Randomized Clinical Trial
Brown, Patrick A; Ji, Lingyun; Xu, Xinxin; Devidas, Meenakshi; Hogan, Laura E; Borowitz, Michael J; Raetz, Elizabeth A; Zugmaier, Gerhard; Sharon, Elad; Bernhardt, Melanie B; Terezakis, Stephanie A; Gore, Lia; Whitlock, James A; Pulsipher, Michael A; Hunger, Stephen P; Loh, Mignon L
Importance:Standard chemotherapy for first relapse of B-cell acute lymphoblastic leukemia (B-ALL) in children, adolescents, and young adults is associated with high rates of severe toxicities, subsequent relapse, and death, especially for patients with early relapse (high risk) or late relapse with residual disease after reinduction chemotherapy (intermediate risk). Blinatumomab, a bispecific CD3 to CD19 T cell-engaging antibody construct, is efficacious in relapsed/refractory B-ALL and has a favorable toxicity profile. Objective:To determine whether substituting blinatumomab for intensive chemotherapy in consolidation therapy would improve survival in children, adolescents, and young adults with high- and intermediate-risk first relapse of B-ALL. Design, Setting, and Participants:This trial was a randomized phase 3 clinical trial conducted by the Children's Oncology Group at 155 hospitals in the US, Canada, Australia, and New Zealand with enrollment from December 2014 to September 2019 and follow-up until September 30, 2020. Eligible patients included those aged 1 to 30 years with B-ALL first relapse, excluding those with Down syndrome, Philadelphia chromosome-positive ALL, prior hematopoietic stem cell transplant, or prior blinatumomab treatment (n = 669). Interventions:All patients received a 4-week reinduction chemotherapy course, followed by randomized assignment to receive 2 cycles of blinatumomab (n = 105) or 2 cycles of multiagent chemotherapy (n = 103), each followed by transplant. Main Outcome and Measures:The primary end point was disease-free survival and the secondary end point was overall survival, both from the time of randomization. The threshold for statistical significance was set at a 1-sided P <.025. Results:Among 208 randomized patients (median age, 9 years; 97 [47%] females), 118 (57%) completed the randomized therapy. Randomization was terminated at the recommendation of the data and safety monitoring committee without meeting stopping rules for efficacy or futility; at that point, 80 of 131 planned events occurred. With 2.9 years of median follow-up, 2-year disease-free survival was 54.4% for the blinatumomab group vs 39.0% for the chemotherapy group (hazard ratio for disease progression or mortality, 0.70 [95% CI, 0.47-1.03]); 1-sided P = .03). Two-year overall survival was 71.3% for the blinatumomab group vs 58.4% for the chemotherapy group (hazard ratio for mortality, 0.62 [95% CI, 0.39-0.98]; 1-sided P = .02). Rates of notable serious adverse events included infection (15%), febrile neutropenia (5%), sepsis (2%), and mucositis (1%) for the blinatumomab group and infection (65%), febrile neutropenia (58%), sepsis (27%), and mucositis (28%) for the chemotherapy group. Conclusions and Relevance:Among children, adolescents, and young adults with high- and intermediate-risk first relapse of B-ALL, postreinduction treatment with blinatumomab compared with chemotherapy, followed by transplant, did not result in a statistically significant difference in disease-free survival. However, study interpretation is limited by early termination with possible underpowering for the primary end point. Trial Registration:ClinicalTrials.gov Identifier: NCT02101853.
PMID: 33651090
ISSN: 1538-3598
CID: 4801342