Try a new search

Format these results:

Searched for:

person:raetze01

in-biosketch:true

Total Results:

267


Racial and ethnic disparities in childhood and young adult acute lymphocytic leukaemia: secondary analyses of eight Children's Oncology Group cohort trials

Gupta, Sumit; Dai, Yunfeng; Chen, Zhiguo; Winestone, Lena E; Teachey, David T; Bona, Kira; Aplenc, Richard; Rabin, Karen R; Zweidler-McKay, Patrick; Carroll, Andrew J; Heerema, Nyla A; Gastier-Foster, Julie; Borowitz, Michael J; Wood, Brent L; Maloney, Kelly W; Mattano, Leonard A; Larsen, Eric C; Angiolillo, Anne L; Burke, Michael J; Salzer, Wanda L; Winter, Stuart S; Brown, Patrick A; Guest, Erin M; Dunsmore, Kimberley P; Kairalla, John A; Winick, Naomi J; Carroll, William L; Raetz, Elizabeth A; Hunger, Stephen P; Loh, Mignon L; Devidas, Meenakshi
BACKGROUND:Previous studies have identified racial and ethnic disparities in childhood acute lymphocytic leukaemia survival. We aimed to establish whether disparities persist in contemporaneous cohorts and, if present, are attributable to differences in leukaemia biology or insurance status. METHODS:Patients with newly diagnosed acute lymphocytic leukaemia in inpatient and outpatient centres in the USA, Canada, Australia, and New Zealand, aged 0-30 years, who had race or ethnicity data available, enrolled on eight completed Children's Oncology Group trials (NCT00103285, NCT00075725, NCT00408005, NCT01190930, NCT02883049, NCT02112916, NCT02828358, and NCT00557193) were included in this secondary analysis. Race and ethnicity were categorised as non-Hispanic White, Hispanic, non-Hispanic Black, non-Hispanic Asian, and non-Hispanic other. Event-free survival and overall survival were compared across race and ethnicity groups. The relative contribution of clinical and biological disease prognosticators and insurance status was examined through multivariable regression models, both among the entire cohort and among those with B-cell lineage versus T-cell lineage disease. FINDINGS/RESULTS:Between Jan 1, 2004, and Dec 31, 2019, 24 979 eligible children, adolescents, and young adults with acute lymphocytic leukaemia were enrolled, of which 21 152 had race or ethnicity data available. 11 849 (56·0%) were male and 9303 (44·0%) were female. Non-Hispanic White patients comprised the largest racial or ethnic group (13 872 [65·6%]), followed by Hispanic patients (4354 [20·6%]), non-Hispanic Black patients (1517 [7·2%]), non-Hispanic Asian (n=1071 [5·1%]), and non-Hispanic other (n=338 [1·6%]). 5-year event-free survival was 87·4% (95% CI 86·7-88·0%) among non-Hispanic White patients compared with 82·8% (81·4-84·1%; hazard ratio [HR] 1·37, 95% CI 1·26-1·49; p<0·0001) among Hispanic patients and 81·8% (79·3-84·0; HR 1·45, 1·28-1·65; p<0·0001) among non-Hispanic Black patients. Non-hispanic Asian patients had a 5-year event-free survival of 88·1% (95% CI 85·5-90·3%) and non-Hispanic other patients had a survival of 82·8% (76·4-87·6%). Inferior event-free survival among Hispanic patients was substantially attenuated by disease prognosticators and insurance status (HR decreased from 1·37 [1·26-1·49; p<0·0001] to 1·11 [1·00-1·22; p=0·045]). The increased risk among non-Hispanic Black patients was minimally attenuated (HR 1·45 [1·28-1·65; p<0·0001] to 1·32 [1·14-1·52; p<0·0001]). 5-year overall survival was 93·6% (91·5-95·1%) in non-Hispanic Asian patients, 93·3% (92·8-93·7%) in non-Hispanic White patients, 89·9% (88·7-90·9%) in Hispanic, 89·7% (87·6-91·4%) in non-Hispanic Black patients, 88·9% (83·2-92·7%) in non-Hispanic other patients. Disparities in overall survival were wider than event-free survival (eg, among non-Hispanic other patients, the HR for event-free survival was 1·43 [1·10-1·85] compared with 1·74 [1·27-2·40] for overall survival). Disparities were restricted to patients with B-cell acute lymphocytic leukaemia, no differences in event-free survival or overall survival were seen in the T-cell acute lymphocytic leukaemia group. INTERPRETATION/CONCLUSIONS:Substantial disparities in outcome for B-cell acute lymphocytic leukaemia persist by race and ethnicity, but are not observed in T-cell acute lymphocytic leukaemia. Future studies of relapsed patients, access to and quality of care, and other potential aspects of structural racism are warranted to inform interventions aimed at dismantling racial and ethnic disparities. FUNDING/BACKGROUND:National Cancer Institute and St Baldrick's Foundation.
PMID: 36725118
ISSN: 2352-3026
CID: 5420142

Association of Inherited Genetic Factors With Drug-Induced Hepatic Damage Among Children With Acute Lymphoblastic Leukemia

Yang, Wenjian; Karol, Seth E; Hoshitsuki, Keito; Lee, Shawn; 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; Relling, Mary V; Yang, Jun J
IMPORTANCE:Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Hepatotoxic effects, including hyperbilirubinemia and elevated alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, are common during all phases of therapy and are linked to several chemotherapeutic agents, including asparaginase, mercaptopurine, and methotrexate. OBJECTIVE:To determine which genetic variants were associated with hyperbilirubinemia and elevated ALT and AST levels in children, adolescents, and young adults treated for ALL. DESIGN, SETTING, AND PARTICIPANTS:This retrospective analysis of a multiethnic genome-wide association study was conducted between January 1, 2019, and April 15, 2022, including patients treated as part of Children's Oncology Group (COG) trials with centers in the United States, Canada, and Australia, which accrued data from December 29, 2003, to January 21, 2011 (AALL0232), and from January 22, 2007, to July 24, 2014 (AALL0434). Germline genotypes were interrogated using genome-wide arrays and imputed using a National Institutes of Health TOPMed Imputation server. Mixed-effects logistic regressions were used to account for multiple episodes for an individual patient. Genotype × treatment phase interaction was tested to uncover phase-specific genetic risk factors. EXPOSURES:Total duration of multiagent protocol chemotherapy ranging from 2.5 to 3.5 years. MAIN OUTCOMES AND MEASURES:The primary outcomes were National Cancer Institute Common Terminology Criteria for Adverse Events (version 4) hyperbilirubinemia of grade 3 or higher and elevated liver ALT and AST levels. RESULTS:A total of 3557 participants were included in the analysis (2179 [61.3%] male; median age, 11.1 [range, 1-30] years). Among 576 known variants associated with these liver function test results in the general population, UGT1A1 variant rs887829 and PNPLA3 variant rs738409 were associated with increased risk of hyperbilirubinemia (odds ratio [OR], 2.18 [95% CI, 1.89-2.53]; P = 6.7 × 10-27) and ALT and AST levels (OR, 1.27 [95% CI, 1.15-1.40]; P = 3.7 × 10-7), respectively, during treatment for ALL. Corresponding polygenic risk scores were associated with hepatotoxic effects across all therapy phases and were largely driven by UGT1A1 and PNPLA3 variants. Genome-wide association analysis revealed an age-specific variant near the CPT1A gene that was only associated with elevated ALT and AST levels among patients younger than 10 years (OR, 1.28 [95% CI, 1.18-1.39]; P = 8.7 × 10-10). CONCLUSIONS AND RELEVANCE:These results suggest a strong genetic basis for interpatient variability in hyperbilirubinemia and aminotransferase level elevations during leukemia chemotherapy.
PMCID:9857512
PMID: 36580335
ISSN: 2574-3805
CID: 5409692

Improving infectious adverse event reporting for children and adolescents enrolled in clinical trials for acute lymphoblastic leukemia: A report from the Children's Oncology Group

Elgarten, Caitlin W; Thompson, Joel C; Angiolillo, Anne; Chen, Zhiguo; Conway, Susan; Devidas, Meenakshi; Gupta, Sumit; Kairalla, John A; McNeer, Jennifer L; O'Brien, Maureen M; Rabin, Karen R; Rau, Rachel E; Rheingold, Susan R; Wang, Cindy; Wood, Charlotte; Raetz, Elizabeth A; Loh, Mignon L; Alexander, Sarah; Miller, Tamara P
Infections cause substantial morbidity for children with acute lymphoblastic leukemia (ALL). Therefore, accurate characterization of infectious adverse events (AEs) reported on clinical trials is imperative to defining, comparing, and managing safety and toxicity. Here, we describe key processes implemented to improve reporting of infectious AEs on two active phase III Children's Oncology Group (COG) ALL trials. Processes include: (a) identifying infections as a targeted toxicity, (b) incorporation of infection-specific case report form questions, and (c) physician review of AEs with real-time data cleaning. Preliminary assessment of these processes suggests improved reporting, as well as opportunities for further improvement.
PMID: 36083863
ISSN: 1545-5017
CID: 5333852

How do mTOR inhibitors fit in the relapsed acute lymphoblastic leukemia treatment landscape?

Pinchinat, Ashley; Raetz, Elizabeth
Not available.
PMID: 35112555
ISSN: 1592-8721
CID: 5153742

Molecular mechanisms of ARID5B-mediated genetic susceptibility to acute lymphoblastic leukemia

Zhao, Xujie; Qian, Maoxiang; Goodings, Charnise; Zhang, Yang; Yang, Wenjian; Wang, Ping; Xu, Beisi; Tian, Cheng; Pui, Ching-Hon; Hunger, Stephen P; Raetz, Elizabeth A; Devidas, Meenakshi; Relling, Mary V; Loh, Mignon L; Savic, Daniel; Li, Chunliang; Yang, Jun J
BACKGROUND:There is growing evidence for the inherited basis of susceptibility to childhood acute lymphoblastic leukemia (ALL). Genome-wide association studies have identified non-coding ALL risk variants at the ARID5B gene locus, but their exact functional effects and the molecular mechanism linking ARID5B to B-ALL leukemogenesis remain largely unknown. METHODS:We performed targeted sequencing of ARID5B in germline DNA of 5,008 children with ALL. Variants were evaluated for association with ALL susceptibility using 3,644 subjects from the UK10K cohort as non-ALL controls, under an additive model. Cis-regulatory elements in ARID5B were systematically identified using CRISPRi enhancer screen in ALL cells. Disruption of transcription factor binding by ARID5B variant was predicted informatically and then confirmed using chromatin immunoprecipitation and co-immunoprecipitation. ARID5B variant association with hematological traits was examined using UK Biobank dataset. All statistical tests are two-sided. RESULTS:We identified 54 common variants in ARID5B significantly associated with leukemia risk, all of which were non-coding. Six cis-regulatory elements at the ARID5B locus were discovered using CRISPR-based high-throughput enhancer screening. Strikingly, the top ALL risk variant (rs7090445, P=5.57 × 10-45) is located precisely within the strongest enhancer element, which is also distally tethered to the ARID5B promoter. The variant allele disrupts the MEF2C binding motif sequence, resulting in reduced MEF2C affinity and decreased local chromosome accessibility. MEF2C influences ARID5B expression in ALL, likely via a transcription factor complex with RUNX1. Using the UK Biobank dataset (n = 349,861), we showed that rs7090445 was also associated with lymphocyte percentage and count in the general population (P=8.6 × 10-22 and 2.1 × 10-18, respectively). CONCLUSION/CONCLUSIONS:Our results indicate that ALL risk variants in ARID5B function by modulating cis-regulatory elements at this locus.
PMID: 35575404
ISSN: 1460-2105
CID: 5249172

The genomic landscape of pediatric acute lymphoblastic leukemia

Brady, Samuel W; Roberts, Kathryn G; Gu, Zhaohui; Shi, Lei; Pounds, Stanley; Pei, Deqing; Cheng, Cheng; Dai, Yunfeng; Devidas, Meenakshi; Qu, Chunxu; Hill, Ashley N; Payne-Turner, Debbie; Ma, Xiaotu; Iacobucci, Ilaria; Baviskar, Pradyuamna; Wei, Lei; Arunachalam, Sasi; Hagiwara, Kohei; Liu, Yanling; Flasch, Diane A; Liu, Yu; Parker, Matthew; Chen, Xiaolong; Elsayed, Abdelrahman H; Pathak, Omkar; Li, Yongjin; Fan, Yiping; Michael, J Robert; Rusch, Michael; Wilkinson, Mark R; Foy, Scott; Hedges, Dale J; Newman, Scott; Zhou, Xin; Wang, Jian; Reilly, Colleen; Sioson, Edgar; Rice, Stephen V; Pastor Loyola, Victor; Wu, Gang; Rampersaud, Evadnie; Reshmi, Shalini C; Gastier-Foster, Julie; Guidry Auvil, Jaime M; Gesuwan, Patee; Smith, Malcolm A; Winick, Naomi; Carroll, Andrew J; Heerema, Nyla A; Harvey, Richard C; Willman, Cheryl L; Larsen, Eric; Raetz, Elizabeth A; Borowitz, Michael J; Wood, Brent L; Carroll, William L; Zweidler-McKay, Patrick A; Rabin, Karen R; Mattano, Leonard A; Maloney, Kelly W; Winter, Stuart S; Burke, Michael J; Salzer, Wanda; Dunsmore, Kimberly P; Angiolillo, Anne L; Crews, Kristine R; Downing, James R; Jeha, Sima; Pui, Ching-Hon; Evans, William E; Yang, Jun J; Relling, Mary V; Gerhard, Daniela S; Loh, Mignon L; Hunger, Stephen P; Zhang, Jinghui; Mullighan, Charles G
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Here, using whole-genome, exome and transcriptome sequencing of 2,754 childhood patients with ALL, we find that, despite a generally low mutation burden, ALL cases harbor a median of four putative somatic driver alterations per sample, with 376 putative driver genes identified varying in prevalence across ALL subtypes. Most samples harbor at least one rare gene alteration, including 70 putative cancer driver genes associated with ubiquitination, SUMOylation, noncoding transcripts and other functions. In hyperdiploid B-ALL, chromosomal gains are acquired early and synchronously before ultraviolet-induced mutation. By contrast, ultraviolet-induced mutations precede chromosomal gains in B-ALL cases with intrachromosomal amplification of chromosome 21. We also demonstrate the prognostic significance of genetic alterations within subtypes. Intriguingly, DUX4- and KMT2A-rearranged subtypes separate into CEBPA/FLT3- or NFATC4-expressing subgroups with potential clinical implications. Together, these results deepen understanding of the ALL genomic landscape and associated outcomes.
PMID: 36050548
ISSN: 1546-1718
CID: 5332172

Persistence of chemotherapy-induced peripheral neuropathy despite vincristine reduction in childhood b-acute lymphoblastic leukemia

Rodwin, Rozalyn L; Kairalla, John A; Hibbitts, Emily; Devidas, Meenakshi; Whitley, Moira K; Mohrmann, Caroline E; Schore, Reuven J; Raetz, Elizabeth; Winick, Naomi J; Hunger, Stephen P; Loh, Mignon L; Hockenberry, Marilyn J; Angiolillo, Anne L; Ness, Kirsten K; Kadan-Lottick, Nina S
BACKGROUND:Children with B-acute lymphoblastic leukemia (B-ALL) are at risk for chemotherapy-induced peripheral neuropathy (CIPN). Children's Oncology Group AALL0932 randomized reduction in vincristine/dexamethasone (every 4-week [VCR/DEX4] vs. 12-week [VCR/DEX12]) during maintenance in the average-risk subset of NCI standard-B-ALL (SR AR B-ALL). We longitudinally measured CIPN, overall, and by treatment group. METHODS:AALL0932 SR AR B-ALL patients ≥3 years old were evaluated at T1-T4 (end-consolidation, maintenance month-1, maintenance month-18, 12-months post-therapy). Physical/occupational therapists (PT/OT) measured motor CIPN (hand/ankle strength, dorsiflexion/plantarflexion range of motion [ROM]), sensory CIPN (finger/toe vibration and touch), and function (dexterity [Purdue Pegboard], walking efficiency [Six Minute Walk]). Proxy-reported function (Pediatric Outcome Data Collection Instrument) and quality of life (Pediatric Quality of Life Inventory) were assessed. Age/sex matched Z-scores and proportion impaired were measured longitudinally and compared between groups. RESULTS:Consent and data were obtained from 150 participants (mean age 5.1 years [SD = 1.7], 48.7% female). Among participants with completed evaluations, 81.8% had CIPN at T1 (74.5% motor, 34.1% sensory). When examining severity of PT/OT outcomes, only handgrip strength (p<.001) and walking efficiency (p=.02) improved from T1-T4 and only dorsiflexion ROM (46.7% vs. 14.7%, p=.008) and handgrip strength (22.2% vs. 37.1%, p=.03) differed in VCR/DEX4 vs. VCR/DEX12 at T4. Proxy-reported outcomes improved from T1 to T4 (P<.001) and most did not differ between groups. CONCLUSIONS:CIPN is prevalent early in B-ALL therapy and persists at least 12-months post-therapy. Most outcomes did not differ between treatment groups despite reduction in vincristine frequency. Children with B-ALL should be monitored for CIPN, even with reduced vincristine frequency.
PMID: 35552709
ISSN: 1460-2105
CID: 5214842

Rational drug combinations with CDK4/6 inhibitors in acute lymphoblastic Leukemia

Bride, Karen L; Hu, Hai; Tikhonova, Anastasia; Fuller, Tori J; Vincent, Tiffaney L; Shraim, Rawan; Li, Marilyn M; Carroll, William L; Raetz, Elizabeth A; Aifantis, Iannis; Teachey, David T
Despite improvements in outcomes for children with B and T-cell acute lymphoblastic leukemia (B-ALL and T-ALL), patients with resistant or relapsed disease fare poorly. Previous studies have demonstrated the essential role of cyclin D3 in T-ALL disease initiation and progression and that targeting of the CDK4/6-cyclin D complex can suppress T-ALL proliferation, leading to efficient cell death in animal models. Studies in leukemia and other malignancies, suggest that schedule is important when combining CDK4/6 inhibitors (CDKis) with cytotoxic agents. Based on these observations, we broadened evaluation of two CDKis, palbociclib (PD-0332991, Pfizer) and ribociclib (LEE011, Novartis) in B and T-ALL as single agent and in combination with conventional cytotoxic chemotherapy, using different schedules in preclinical models. As monotherapy, CDKis caused cell cycle arrest with a significant decrease in S phase entry and were active in vivo across a broad number of patient-derived xenograft samples. Prolonged monotherapy induces resistance, for which we identified a potential novel mechanism using transcriptome profiling. Importantly, simultaneous but not sequential treatment of CDKis with conventional chemotherapy (dexamethasone, L-asparaginase and vincristine) led to improved efficacy compared to monotherapy in vivo. We provide novel evidence that combining CDKis and conventional chemotherapy can be safe and effective. These results led to the rational design of a clinical trial.
PMID: 34937317
ISSN: 1592-8721
CID: 5108942

Do Not Forget About the Ticks: An Unusual Cause of Fever, GI Distress, and Cytopenias in a Child With ALL

Ungar, Stephanie P; Varkey, Joyce; Pierro, Joanna; Raetz, Elizabeth; Ratner, Adam J
We report the case of a 5-year-old male with B-cell acute lymphoblastic leukemia in remission, receiving maintenance chemotherapy, who presented with fever, emesis, diarrhea, headache, and lethargy. He developed rapidly progressive cytopenias and was found to have acute human granulocytic anaplasmosis as well as evidence of past infection with Babesia microti. The case highlights the need to maintain a broad differential for infection in children undergoing chemotherapy or other immunosuppressive therapies with possible or known tick exposure.
PMID: 34935737
ISSN: 1536-3678
CID: 5108892

Activation of the mitogen-activated protein kinase-extracellular signal-regulated kinase pathway in childhood B-cell acute lymphoblastic leukemia

Pillai, Pallavi M; Mallory, Nicole; Pierro, Joanna; Saliba, Jason; Newman, Daniel; Hu, Jiyuan; Bhatla, Teena; Raetz, Elizabeth; Carroll, William L; Evensen, Nikki A
RAS mutations are frequently observed in childhood B-cell acute lymphoblastic leukemia (B-ALL) and previous studies have yielded conflicting results as to whether they are associated with a poor outcome. We and others have demonstrated that the mitogen-activated protein kinase-extracellular signal-regulated kinase (MAPK) pathway can be activated through epigenetic mechanisms in the absence of RAS pathway mutations. Herein, we examined whether MAPK activation, as determined by measuring phosphorylated extracellular signal-regulated kinase (pERK) levels in 80 diagnostic patient samples using phosphoflow cytometry, could be used as a prognostic biomarker for pediatric B-ALL. The mean fluorescence intensity of pERK (MFI) was measured at baseline and after exogenous stimulation with or without pretreatment with the mitogen-activated protein kinase kinase (MEK) inhibitor trametinib. Activation levels (MFI stimulated/MFI baseline) ranged from 0.76 to 4.40 (median = 1.26), and inhibition indexes (MFI stimulated/MFI trametinib stimulated) ranged from 0.439 to 5.640 (median = 1.30), with no significant difference between patients with wildtype versus mutant RAS for either. Logistic regression demonstrated that neither MAPK activation levels nor RAS mutation status at diagnosis alone or in combination was prognostic of outcome. However, 35% of RAS wildtype samples showed MAPK inhibition indexes greater than the median, thus raising the possibility that therapeutic strategies to inhibit MAPK activation may not be restricted to patients whose blasts display Ras pathway defects.
PMID: 35593589
ISSN: 1545-5017
CID: 5247702