Try a new search

Format these results:

Searched for:

person:raetze01

in-biosketch:true

Total Results:

276


Determinants of survival after first relapse of acute lymphoblastic leukemia: a Children's Oncology Group study

Rheingold, Susan R; Bhojwani, Deepa; Ji, Lingyun; Xu, Xinxin; Devidas, Meenakshi; Kairalla, John A; Shago, Mary; Heerema, Nyla A; Carroll, Andrew J; Breidenbach, Heather; Borowitz, Michael; Wood, Brent L; Angiolillo, Anne L; Asselin, Barbara L; Bowman, W Paul; Brown, Patrick; Dreyer, ZoAnn E; Dunsmore, Kimberly P; Hilden, Joanne M; Larsen, Eric; Maloney, Kelly; Matloub, Yousif; Mattano, Leonard A; Winter, Stuart S; Gore, Lia; Winick, Naomi J; Carroll, William L; Hunger, Stephen P; Raetz, Elizabeth A; Loh, Mignon L
Limited prognostic factors have been associated with overall survival (OS) post-relapse in childhood Acute Lymphoblastic Leukemia (ALL). Patients enrolled on 12 Children's Oncology Group frontline ALL trials (1996-2014) were analyzed to assess for additional prognostic factors associated with OS post-relapse. Among 16,115 patients, 2053 (12.7%) relapsed. Relapse rates were similar for B-ALL (12.5%) and T-ALL (11.2%) while higher for infants (34.2%). Approximately 50% of B-ALL relapses occurred late (≥36 months) and 72.5% involved the marrow. Conversely, 64.8% of T-ALL relapses occurred early (<18 months) and 47.1% involved the central nervous system. The 5-year OS post-relapse for the entire cohort was 48.9 ± 1.2%; B-ALL:52.5 ± 1.3%, T-ALL:35.5 ± 3.3%, and infant ALL:21.5 ± 3.9%. OS varied by early, intermediate and late time-to-relapse; 25.8 ± 2.4%, 49.5 ± 2.2%, and 66.4 ± 1.8% respectively for B-ALL and 29.8 ± 3.9%, 33.3 ± 7.6%, 58 ± 9.8% for T-ALL. Patients with ETV6::RUNX1 or Trisomy 4 + 10 had median time-to-relapse of 43 months and higher OS post-relapse 74.4 ± 3.1% and 70.2 ± 3.6%, respectively. Patients with hypodiploidy, KMT2A-rearrangement, and TCF3::PBX1 had short median time-to-relapse (12.5-18 months) and poor OS post-relapse (14.2 ± 6.1%, 31.9 ± 7.7%, 36.8 ± 6.6%). Site-of-relapse varied by cytogenetic subtype. This large dataset provided the opportunity to identify risk factors for OS post-relapse to inform trial design and highlight populations with dismal outcomes post-relapse.
PMID: 39261601
ISSN: 1476-5551
CID: 5690472

Clonal evolution of the 3D chromatin landscape in patients with relapsed pediatric B-cell acute lymphoblastic leukemia

Narang, Sonali; Ghebrechristos, Yohana; Evensen, Nikki A; Murrell, Nina; Jasinski, Sylwia; Ostrow, Talia H; Teachey, David T; Raetz, Elizabeth A; Lionnet, Timothee; Witkowski, Matthew; Aifantis, Iannis; Tsirigos, Aristotelis; Carroll, William L
Relapsed pediatric B-cell acute lymphoblastic leukemia (B-ALL) remains one of the leading causes of cancer mortality in children. We performed Hi-C, ATAC-seq, and RNA-seq on 12 matched diagnosis/relapse pediatric leukemia specimens to uncover dynamic structural variants (SVs) and 3D chromatin rewiring that may contribute to relapse. While translocations are assumed to occur early in leukemogenesis and be maintained throughout progression, we discovered novel, dynamic translocations and confirmed several fusion transcripts, suggesting functional and therapeutic relevance. Genome-wide chromatin remodeling was observed at all organizational levels: A/B compartments, TAD interactivity, and chromatin loops, including some loci shared by 25% of patients. Shared changes were found to drive the expression of genes/pathways previously implicated in resistance as well as novel therapeutic candidates, two of which (ATXN1 and MN1) we functionally validated. Overall, these results demonstrate chromatin reorganization under the selective pressure of therapy and offer the potential for discovery of novel therapeutic interventions.
PMCID:11358475
PMID: 39198446
ISSN: 2041-1723
CID: 5701942

Daratumumab in Pediatric Relapsed/Refractory Acute Lymphoblastic Leukemia or Lymphoblastic Lymphoma: DELPHINUS Study

Bhatla, Teena; Hogan, Laura; Teachey, David Trent; Bautista, Franciso; Moppett, John P; Velasco, Pablo; Micalizzi, Concetta; Rossig, Claudia; Shukla, Neerav Narendra; Gilad, Gil; Locatelli, Franco; Baruchel, André; Zwaan, Michel; Bezler, Natalie S; Rubio-San-Simón, Alba; Taussig, David; Raetz, Elizabeth A; Mao, Zhengwei J; Wood, Brent; Alvarez Arias, Diana; Krevvata, Maria; Nnane, Ivo; Bandyopadhyay, Nibedita; Lopez Solano, Lorena; Dennis, Robyn M; Carson, Robin; Vora, Ajay
Patients with relapsed/refractory acute lymphoblastic leukemia (ALL) or lymphoblastic lymphoma (LL) have poor outcomes compared with newly diagnosed, treatment-naïve patients. The phase 2, open-label DELPHINUS study evaluated daratumumab (16 mg/kg intravenously) plus backbone chemotherapy in children with relapsed/refractory B-cell ALL (n=7) after ≥2 relapses and children and young adults with T-cell ALL (children, n=24; young adults, n=5) or LL (n=10) after first relapse. The primary endpoint was complete response (CR) in the B-cell ALL (end of Cycle 2) and T-cell ALL (end of Cycle 1) cohorts, after which patients could proceed off study to allogeneic hematopoietic stem cell transplant (HSCT). Seven patients with advanced B-cell ALL received daratumumab with no CRs achieved; this cohort was closed due to futility. For the childhood T-cell ALL, young adult T-cell ALL, and T-cell LL cohorts, the CR (end of Cycle 1) rates were 41.7%, 60.0%, and 30.0%, respectively; overall response rates (any time point) were 83.3% (CR+CR with incomplete count recovery [CRi]), 80.0% (CR+CRi), and 50.0% (CR+partial response); minimal residual disease-negativity (<0.01%) rates were 45.8%, 20.0%, and 50.0%; observed 24-month event-free survival rates were 36.1%, 20.0%, and 20.0%; observed 24-month overall survival rates were 41.3%, 25.0%, and 20.0%; and allogeneic HSCT rates were 75.0%, 60.0%, and 30.0%. No new safety concerns with daratumumab were observed. In conclusion, daratumumab was safely combined with backbone chemotherapy in children and young adults with T-cell ALL/LL and contributed to successful bridging to HSCT. This trial was registered at www.ClinicalTrials.gov as NCT03384654.
PMID: 39158071
ISSN: 1528-0020
CID: 5680452

Genomic Determinants of Outcome in Acute Lymphoblastic Leukemia

Chang, Ti-Cheng; Chen, Wenan; Qu, Chunxu; Cheng, Zhongshan; Hedges, Dale; Elsayed, Abdelrahman; Pounds, Stanley B; Shago, Mary; Rabin, Karen R; Raetz, Elizabeth A; Devidas, Meenakshi; Cheng, Cheng; Angiolillo, Anne; Baviskar, Pradyuamna; Borowitz, Michael; Burke, Michael J; Carroll, Andrew; Carroll, William L; Chen, I-Ming; Harvey, Richard; Heerema, Nyla; Iacobucci, Ilaria; Wang, Jeremy R; Jeha, Sima; Larsen, Eric; Mattano, Leonard; Maloney, Kelly; Pui, Ching-Hon; Ramirez, Nilsa C; Salzer, Wanda; Willman, Cheryl; Winick, Naomi; Wood, Brent; Hunger, Stephen P; Wu, Gang; Mullighan, Charles G; Loh, Mignon L
PURPOSE/OBJECTIVE:Although cure rates for childhood acute lymphoblastic leukemia (ALL) exceed 90%, ALL remains a leading cause of cancer death in children. Half of relapses arise in children initially classified with standard-risk (SR) disease. MATERIALS AND METHODS/METHODS:To identify genomic determinants of relapse in children with SR ALL, we performed genome and transcriptome sequencing of diagnostic and remission samples of children with SR (n = 1,381) or high-risk B-ALL with favorable cytogenetic features (n = 115) enrolled on Children's Oncology Group trials. We used a case-control study design analyzing 439 patients who relapsed and 1,057 who remained in complete remission for at least 5 years. RESULTS:in high-hyperdiploid ALL. CONCLUSION/CONCLUSIONS:Genetic subtype, patterns of aneuploidy, and secondary genomic alterations determine risk of relapse in childhood ALL. Comprehensive genomic analysis is required for optimal risk stratification.
PMID: 39121442
ISSN: 1527-7755
CID: 5696962

Impact of Propofol Exposure on Neurocognitive Outcomes in Children With High-Risk B ALL: A Children's Oncology Group Study

Alexander, Sarah; Kairalla, John A; Gupta, Sumit; Hibbitts, Emily; Weisman, Hannah; Anghelescu, Doralina; Winick, Naomi J; Krull, Kevin R; Salzer, Wanda L; Burke, Michael J; Gore, Lia; Devidas, Meenakshi; Embry, Leanne; Raetz, Elizabeth A; Hunger, Stephen P; Loh, Mignon L; Hardy, Kristina K
PURPOSE/OBJECTIVE:Many children treated for ALL develop long-term neurocognitive impairments. Increased risk of these impairments is associated with treatment and demographic factors. Exposure to anesthesia is an additional possible risk factor. This study evaluated the impact of cumulative exposure to anesthesia on neurocognitive outcomes among a multicenter cohort of children with ALL. METHODS:This study was embedded in AALL1131, a Children's Oncology Group phase III trial for patients with high-risk B-ALL. In consenting patients age 6-12 years, prospective uniform assessments of neurocognitive function were performed during and at 1 year after completion of therapy. Exposure to all episodes of anesthetic agents was abstracted. Multivariable linear regression models determined associations of cumulative anesthetic agents with the primary neurocognitive outcome reaction time/processing speed (age-normed) at 1 year off therapy, adjusting for baseline neurocognitive score, age, sex, race/ethnicity, insurance status (as a proxy for socioeconomic status), and leukemia risk group. RESULTS:= .03). CONCLUSION/CONCLUSIONS:In a multicenter and uniformly treated cohort of children with B-ALL, cumulative exposure to propofol was an independent risk factor for impairment in reaction time/processing speed 1 year after therapy. Anesthesia exposure is a modifiable risk, and opportunities to minimize propofol use should be considered.
PMID: 38603641
ISSN: 1527-7755
CID: 5695592

The genomic basis of childhood T-lineage acute lymphoblastic leukaemia

Pölönen, Petri; Di Giacomo, Danika; Seffernick, Anna Eames; Elsayed, Abdelrahman; Kimura, Shunsuke; Benini, Francesca; Montefiori, Lindsey E; Wood, Brent L; Xu, Jason; Chen, Changya; Cheng, Zhongshan; Newman, Haley; Myers, Jason; Iacobucci, Ilaria; Li, Elizabeth; Sussman, Jonathan; Hedges, Dale; Hui, Yawei; Diorio, Caroline; Uppuluri, Lahari; Frank, David; Fan, Yiping; Chang, Yunchao; Meshinchi, Soheil; Ries, Rhonda; Shraim, Rawan; Li, Alexander; Bernt, Kathrin M; Devidas, Meenakshi; Winter, Stuart S; Dunsmore, Kimberly P; Inaba, Hiroto; Carroll, William L; Ramirez, Nilsa C; Phillips, Aaron H; Kriwacki, Richard W; Yang, Jun J; Vincent, Tiffaney L; Zhao, Yaqi; Ghate, Pankaj S; Wang, Jian; Reilly, Colleen; Zhou, Xin; Sanders, Mathijs A; Takita, Junko; Kato, Motohiro; Takasugi, Nao; Chang, Bill H; Press, Richard D; Loh, Mignon; Rampersaud, Evadnie; Raetz, Elizabeth; Hunger, Stephen P; Tan, Kai; Chang, Ti-Cheng; Wu, Gang; Pounds, Stanley B; Mullighan, Charles G; Teachey, David T
T-lineage acute lymphoblastic leukaemia (T-ALL) is a high-risk tumour1 that has eluded comprehensive genomic characterization, which is partly due to the high frequency of noncoding genomic alterations that result in oncogene deregulation2,3. Here we report an integrated analysis of genome and transcriptome sequencing of tumour and remission samples from more than 1,300 uniformly treated children with T-ALL, coupled with epigenomic and single-cell analyses of malignant and normal T cell precursors. This approach identified 15 subtypes with distinct genomic drivers, gene expression patterns, developmental states and outcomes. Analyses of chromatin topology revealed multiple mechanisms of enhancer deregulation that involve enhancers and genes in a subtype-specific manner, thereby demonstrating widespread involvement of the noncoding genome. We show that the immunophenotypically described, high-risk entity of early T cell precursor ALL is superseded by a broader category of 'early T cell precursor-like' leukaemia. This category has a variable immunophenotype and diverse genomic alterations of a core set of genes that encode regulators of hematopoietic stem cell development. Using multivariable outcome models, we show that genetic subtypes, driver and concomitant genetic alterations independently predict treatment failure and survival. These findings provide a roadmap for the classification, risk stratification and mechanistic understanding of this disease.
PMID: 39143224
ISSN: 1476-4687
CID: 5701902

Azacitidine as epigenetic priming for chemotherapy is safe and well-tolerated in infants with newly diagnosed KMT2A-rearranged acute lymphoblastic leukemia: Children's Oncology Group trial AALL15P1

Guest, Erin M; Kairalla, John A; Devidas, Meenakshi; Hibbitts, Emily; Carroll, Andrew J; Heerema, Nyla A; Kubaney, Holly R; August, Margaret A; Ramesh, Sidharth; Yoo, Byunggil; Farooqi, Midhat S; Pauly, Melinda G; Wechsler, Daniel S; Miles, Rodney R; Reid, Joel M; Kihei, Cynthia D; Gore, Lia; Raetz, Elizabeth A; Hunger, Stephen P; Loh, Mignon L; Brown, Patrick A
Infants less than 1 year old diagnosed with KMT2A-rearranged (KMT2A-r) acute lymphoblastic leukemia (ALL) are at high risk of remission failure, relapse, and death due to leukemia, despite intensive therapies. Infant KMT2A-r ALL blasts are characterized by DNA hypermethylation. Epigenetic priming with DNA methyltransferase inhibitors increases the cytotoxicity of chemotherapy in preclinical studies. The Children's Oncology Group trial AALL15P1 tested the safety and tolerability of five days of azacitidine immediately prior to the start of chemotherapy on day six, in four post-induction chemotherapy courses for infants with newly diagnosed KMT2A-r ALL. The treatment was welltolerated, with only two of 31 evaluable patients (6.5%) experiencing dose-limiting toxicity. Whole genome bisulfite sequencing of peripheral blood mononuclear cells (PBMCs) demonstrated decreased DNA methylation in 87% of samples tested following five days of azacitidine. Event-free survival was similar to prior studies of newly diagnosed infant ALL. Azacitidine is safe and results in decreased DNA methylation of PBMCs in infants with KMT2A-r ALL, but the incorporation of azacitidine to enhance cytotoxicity did not impact survival. Clinicaltrials.gov identifier: NCT02828358.
PMID: 38867582
ISSN: 1592-8721
CID: 5669242

Germ line genetic NBN variation and predisposition to B-cell acute lymphoblastic leukemia in children

Escherich, Carolin S; Chen, Wenan; Li, Yizhen; Yang, Wenjian; Nishii, Rina; Li, Zhenhua; Raetz, Elizabeth A; Devidas, Meenakshi; Wu, Gang; Nichols, Kim E; Inaba, Hiroto; Pui, Ching-Hon; Jeha, Sima; Camitta, Bruce M; Larsen, Eric; Hunger, Stephen P; Loh, Mignon L; Yang, Jun J
Biallelic mutation in the DNA-damage repair gene NBN is the genetic cause of Nijmegen breakage syndrome, which is associated with predisposition to lymphoid malignancies. Heterozygous carriers of germ line NBN variants may also be at risk for leukemia development, although this is much less characterized. By sequencing 4325 pediatric patients with B-cell acute lymphoblastic leukemia (B-ALL), we systematically examined the frequency of germ line NBN variants and identified 25 unique, putatively damaging NBN coding variants in 50 patients. Compared with the frequency of NBN variants in gnomAD noncancer controls (189 unique, putatively damaging NBN coding variants in 472 of 118 479 individuals), we found significant overrepresentation in pediatric B-ALL (P = .004; odds ratio, 1.8). Most B-ALL-risk variants were missense and cluster within the NBN N-terminal domains. Using 2 functional assays, we verified 14 of 25 variants with severe loss-of-function phenotypes and thus classified these as nonfunctional or partially functional. Finally, we found that germ line NBN variant carriers, all of whom were identified as heterozygous genotypes, showed similar survival outcomes relative to those with wild type status. Taken together, our findings provide novel insights into the genetic predisposition to B-ALL, and the impact of NBN variants on protein function and suggest that heterozygous NBN variant carriers may safely receive B-ALL therapy. These trials were registered at www.clinicaltrials.gov as #NCT01225874, NCT00075725, NCT00103285, NCI-T93-0101D, and NCT00137111.
PMID: 38446568
ISSN: 1528-0020
CID: 5662942

MRD at the end of induction and EFS in T-cell lymphoblastic lymphoma: Children's Oncology Group trial AALL1231

Hayashi, Robert J; Hermiston, Michelle L; Wood, Brent L; Teachey, David T; Devidas, Meenakshi; Chen, Zhiguo; Annett, Robert D; Asselin, Barbara L; August, Keith; Cho, Steve; Dunsmore, Kimberly P; Freedman, Jason Lawrence; Galardy, Paul J; Harker-Murray, Paul; Horton, Terzah M; Jaju, Alok; Lam, Allison; Messinger, Yoav H; Miles, Rodney R; Okada, Maki; Patel, Samir; Schafer, Eric S; Schechter, Tal; Shimano, Kristin A; Singh, Neelam; Steele, Amii; Sulis, Maria L; Vargas, Sarah L; Winter, Stuart S; Wood, Charlotte; Zweidler-McKay, Patrick A; Loh, Mignon L; Hunger, Stephen P; Raetz, Elizabeth A; Bollard, Catherine M; Allen, Carl E
Defining prognostic variables in T-lymphoblastic lymphoma (T-LL) remains a challenge. AALL1231 was a Children's Oncology Group phase 3 clinical trial for newly diagnosed patients with T acute lymphoblastic leukemia or T-LL, randomizing children and young adults to a modified augmented Berlin-Frankfurt-Münster backbone to receive standard therapy (arm A) or with addition of bortezomib (arm B). Optional bone marrow samples to assess minimal residual disease (MRD) at the end of induction (EOI) were collected in T-LL analyzed to assess the correlation of MRD at the EOI to event-free survival (EFS). Eighty-six (41%) of the 209 patients with T-LL accrued to this trial submitted samples for MRD assessment. Patients with MRD <0.1% (n = 75) at EOI had a superior 4-year EFS vs those with MRD ≥0.1% (n = 11) (89.0% ± 4.4% vs 63.6% ± 17.2%; P = .025). Overall survival did not significantly differ between the 2 groups. Cox regression for EFS using arm A as a reference demonstrated that MRD EOI ≥0.1% was associated with a greater risk of inferior outcome (hazard ratio, 3.73; 95% confidence interval, 1.12-12.40; P = .032), which was independent of treatment arm assignment. Consideration to incorporate MRD at EOI into future trials will help establish its value in defining risk groups. CT# NCT02112916.
PMID: 38457359
ISSN: 1528-0020
CID: 5655592

Enhanced Risk Stratification for Children and Young Adults with B-Cell Acute Lymphoblastic Leukemia: A Children's Oncology Group Report

DelRocco, N J; Loh, M L; Borowitz, M J; Gupta, S; Rabin, K R; Zweidler-McKay, P; Maloney, K W; Mattano, L A; Larsen, E; Angiolillo, A; Schore, R J; Burke, M J; Salzer, W L; Wood, B L; Carroll, A J; Heerema, N A; Reshmi, S C; Gastier-Foster, J M; Harvey, R; Chen, I M; Roberts, K G; Mullighan, C G; Willman, C; Winick, N; Carroll, W L; Rau, R E; Teachey, D T; Hunger, S P; Raetz, E A; Devidas, M; Kairalla, J A
Current strategies to treat pediatric acute lymphoblastic leukemia rely on risk stratification algorithms using categorical data. We investigated whether using continuous variables assigned different weights would improve risk stratification. We developed and validated a multivariable Cox model for relapse-free survival (RFS) using information from 21199 patients. We constructed risk groups by identifying cutoffs of the COG Prognostic Index (PICOG) that maximized discrimination of the predictive model. Patients with higher PICOG have higher predicted relapse risk. The PICOG reliably discriminates patients with low vs. high relapse risk. For those with moderate relapse risk using current COG risk classification, the PICOG identifies subgroups with varying 5-year RFS. Among current COG standard-risk average patients, PICOG identifies low and intermediate risk groups with 96% and 90% RFS, respectively. Similarly, amongst current COG high-risk patients, PICOG identifies four groups ranging from 96% to 66% RFS, providing additional discrimination for future treatment stratification. When coupled with traditional algorithms, the novel PICOG can more accurately risk stratify patients, identifying groups with better outcomes who may benefit from less intensive therapy, and those who have high relapse risk needing innovative approaches for cure.
PMID: 38360863
ISSN: 1476-5551
CID: 5635932