Try a new search

Format these results:

Searched for:

person:carrow01

Total Results:

370


Minimal residual disease predicts outcomes in KMT2A-rearranged but not KMT2A-germline infant acute lymphoblastic leukemia: Report from Children's Oncology Group study AALL0631

Faulk, Kelly E; Kairalla, John A; Dreyer, ZoAnn E; Carroll, Andrew J; Heerema, Nyla A; Devidas, Meenakshi; Carroll, William L; Raetz, Elizabeth A; Loh, Mignon L; Hunger, Stephen P; Borowitz, Michael; Wang, Cindy; Guest, Erin; Brown, Patrick A
We measured minimal residual disease (MRD) by multiparameter flow cytometry at three time points (TP) in 117 infants with KMT2A (lysine [K]-specific methyltransferase 2A)-rearranged and 58 with KMT2A-germline acute lymphoblastic leukemia (ALL) on Children's Oncology Group AALL0631 study. For KMT2A-rearranged patients, 3-year event-free survival (EFS) by MRD-positive (≥0.01%) versus MRD-negative (<0.01%) was: TP1: 25% (±6%) versus 49% (±7%; p = .0009); TP2: 21% (±8%) versus 47% (±7%; p < .0001); and TP3: 22% (±14%) versus 51% (±6%; p = .0178). For KMT2A-germline patients, 3-year EFS was: TP1: 88% (±12%) versus 87% (±5%; p = .73); TP2: 100% versus 88% (±5%; p = .24); and TP3: 100% versus 87% (±5%; p = .53). MRD was a strong independent outcome predictor in KMT2A-rearranged, but not KMT2A-germline infant ALL.
PMID: 37259259
ISSN: 1545-5017
CID: 5543352

Central Nervous System Status is Prognostic in T-Cell Acute Lymphoblastic Leukemia: A Children's Oncology Group Report

Gossai, Nathan; Devidas, Meenakshi; Chen, Zhiguo; Wood, Brent L; Zweidler-McKay, Patrick A; Rabin, Karen R; Loh, Mignon L; Raetz, Elizabeth A; Winick, Naomi J; Burke, Michael J; Carroll, Andrew J; Esiashvili, Natia; Heerema, Nyla A; Carroll, William L; Hunger, Stephen P; Dunsmore, Kimberly P; Winter, Stuart Sheldon; Teachey, David T
To determine the prognostic significance of central nervous system (CNS) leukemic involvement in newly diagnosed T-cell acute lymphoblastic leukemia (T-ALL), outcomes on consecutive, phase 3 Children's Oncology Group (COG) clinical trials were examined. AALL0434 and AALL1231 tested efficacy of novel agents incorporated into augmented Berlin-Frankfurt-Münster (aBFM) therapy. In addition to testing study-specific chemotherapy through randomization, the AALL0434 regimen delivered cranial radiation (CRT) to the majority of subjects (90.8%), while AALL1231 intensified chemotherapy to eliminate CRT in 88.2% of subjects. In combined analysis of 2,164 T-ALL subjects (AALL0434: 1,550; AALL1231: 614), 1,564 were CNS-1 (72.3%), 441 CNS-2 (20.4%), and 159 CNS-3 (7.3%). The 4-year event-free survival (EFS) was similar for CNS-1 (85.1±1.0%) and CNS-2 (83.2±2.0%), but lower for CNS-3 (71.8±4.0%); p=0.0004. Subjects with CNS-1 and CNS-2 had similar 4-year overall-survival (OS) (90.1±0.8% and 90.5±1.5%), with OS for CNS-3 (82.7±3.4%); p=0.005. Despite therapeutic differences, outcomes for CNS-1 and CNS-2 were similar regardless of CRT, intensified corticosteroids or novel agents. Except for significantly superior outcomes with nelarabine on AALL0434 (4-year disease-free survival 93.1%±5.2%), EFS/OS was inferior with CNS-3 status, all of whom received CRT. Combined analyses of over 2,000 subjects with T-ALL, found that those with CNS-1 and CNS-2 status at diagnosis had similar outcomes. Unlike with B-ALL, CNS-2 status in T-ALL does not impact outcome in the context of aBFM therapy, without additional intrathecal therapy, with or without CRT. Although nelarabine improved outcomes for those with CNS-3 status, novel approaches are needed for further improvements.
PMID: 36603187
ISSN: 1528-0020
CID: 5433512

NSD2 E1099K drives relapse in pediatric acute lymphoblastic leukemia by disrupting 3D chromatin organization

Narang, Sonali; Evensen, Nikki A; Saliba, Jason; Pierro, Joanna; Loh, Mignon L; Brown, Patrick A; Kolekar, Pandurang; Mulder, Heather; Shao, Ying; Easton, John; Ma, Xiaotu; Tsirigos, Aristotelis; Carroll, William L
BACKGROUND:The NSD2 p.E1099K (EK) mutation is shown to be enriched in patients with relapsed acute lymphoblastic leukemia (ALL), indicating a role in clonal evolution and drug resistance. RESULTS:To uncover 3D chromatin architecture-related mechanisms underlying drug resistance, we perform Hi-C on three B-ALL cell lines heterozygous for NSD2 EK. The NSD2 mutation leads to widespread remodeling of the 3D genome, most dramatically in terms of compartment changes with a strong bias towards A compartment shifts. Systematic integration of the Hi-C data with previously published ATAC-seq, RNA-seq, and ChIP-seq data show an expansion in H3K36me2 and a shrinkage in H3K27me3 within A compartments as well as increased gene expression and chromatin accessibility. These results suggest that NSD2 EK plays a prominent role in chromatin decompaction through enrichment of H3K36me2. In contrast, we identify few changes in intra-topologically associating domain activity. While compartment changes vary across cell lines, a common core of decompacting loci are shared, driving the expression of genes/pathways previously implicated in drug resistance. We further perform RNA sequencing on a cohort of matched diagnosis/relapse ALL patients harboring the relapse-specific NSD2 EK mutation. Changes in patient gene expression upon relapse significantly correlate with core compartment changes, further implicating the role of NSD2 EK in genome decompaction. CONCLUSIONS:In spite of cell-context-dependent changes mediated by EK, there appears to be a shared transcriptional program dependent on compartment shifts which could explain phenotypic differences across EK cell lines. This core program is an attractive target for therapeutic intervention.
PMCID:10071675
PMID: 37016431
ISSN: 1474-760x
CID: 5463712

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

An inflammatory state remodels the immune microenvironment and improves risk stratification in acute myeloid leukemia

Lasry, Audrey; Nadorp, Bettina; Fornerod, Maarten; Nicolet, Deedra; Wu, Huiyun; Walker, Christopher J; Sun, Zhengxi; Witkowski, Matthew T; Tikhonova, Anastasia N; Guillamot-Ruano, Maria; Cayanan, Geraldine; Yeaton, Anna; Robbins, Gabriel; Obeng, Esther A; Tsirigos, Aristotelis; Stone, Richard M; Byrd, John C; Pounds, Stanley; Carroll, William L; Gruber, Tanja A; Eisfeld, Ann-Kathrin; Aifantis, Iannis
Acute myeloid leukemia (AML) is a hematopoietic malignancy with poor prognosis and limited treatment options. Here we provide a comprehensive census of the bone marrow immune microenvironment in adult and pediatric patients with AML. We characterize unique inflammation signatures in a subset of AML patients, associated with inferior outcomes. We identify atypical B cells, a dysfunctional B-cell subtype enriched in patients with high-inflammation AML, as well as an increase in CD8+GZMK+ and regulatory T cells, accompanied by a reduction in T-cell clonal expansion. We derive an inflammation-associated gene score (iScore) that associates with poor survival outcomes in patients with AML. Addition of the iScore refines current risk stratifications for patients with AML and may enable identification of patients in need of more aggressive treatment. This work provides a framework for classifying patients with AML based on their immune microenvironment and a rationale for consideration of the inflammatory state in clinical settings.
PMID: 36581735
ISSN: 2662-1347
CID: 5409732

Author Correction: An inflammatory state remodels the immune microenvironment and improves risk stratification in acute myeloid leukemia

Lasry, Audrey; Nadorp, Bettina; Fornerod, Maarten; Nicolet, Deedra; Wu, Huiyun; Walker, Christopher J; Sun, Zhengxi; Witkowski, Matthew T; Tikhonova, Anastasia N; Guillamot-Ruano, Maria; Cayanan, Geraldine; Yeaton, Anna; Robbins, Gabriel; Obeng, Esther A; Tsirigos, Aristotelis; Stone, Richard M; Byrd, John C; Pounds, Stanley; Carroll, William L; Gruber, Tanja A; Eisfeld, Ann-Kathrin; Aifantis, Iannis
PMID: 36658429
ISSN: 2662-1347
CID: 5417042

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

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

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

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