122 results on '"Rabin KR"'
Search Results
2. Integration of cytogenomic data for furthering the characterization of pediatric B-ALL: a multi-institution, multi-platform microarray study
- Author
-
Baughn, LB, Biegel, JA, South, ST, Smolarek, T, Volkert, S, Carroll, A, Heerema, NA, Rabin, KR, Zweidler-McKay, PA, Loh, M, and Hirsch, B
- Subjects
Chromosome Aberrations ,Male ,B-Lymphocytes ,Comparative Genomic Hybridization ,DNA Copy Number Variations ,Loss of Heterozygosity ,Reproducibility of Results ,Prognosis ,Polymorphism, Single Nucleotide ,Sensitivity and Specificity ,Article ,Chromosome Banding ,Karyotyping ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Cytogenetic Analysis ,Humans ,Female ,Child ,In Situ Hybridization, Fluorescence ,Oligonucleotide Array Sequence Analysis - Abstract
It is well documented that among subgroups of B-cell acute lymphoblastic leukemia (B-ALL), the genetic profile of the leukemic blasts has significant impact on prognosis and stratification for therapy. Recent studies have documented the power of microarrays to screen genome-wide for copy number aberrations (CNAs) and regions of copy number-neutral loss of heterozygosity (CNLOH) that are not detectable by G-banding or fluorescence in situ hybridization (FISH). These studies have involved application of a single array platform for the respective cases. The present investigation demonstrates the feasibility and usefulness of integrating array results from multiple laboratories (ARUP, The Children's Hospital of Philadelphia, Cincinnati Children's Hospital Medical Center, and University of Minnesota Medical Center) that utilize different array platforms (Affymetrix, Agilent, or Illumina) in their respective clinical settings. A total of 65 patients enrolled on the Children's Oncology Group (COG) study AALL08B1 were identified for study, as cytogenetic and FISH studies had also been performed on these patients, with a central review of those results available for comparison. Microarray data were first analyzed by the individual laboratories with their respective software systems; raw data files were then centrally validated using NEXUS software. The results demonstrated the added value of integrating multi-platform data with cytogenetic and FISH data and highlight novel findings identified by array including the co-occurrence of low and high risk abnormalities not previously reported to coexist within a clone, novel regions of chromosomal amplification, clones characterized by numerous whole chromosome LOH that do not meet criteria for doubling of a near-haploid, and characterization of array profiles associated with an IKZF1 deletion. Each of these findings raises questions that are clinically relevant to risk stratification.
- Published
- 2014
3. Acute lymphoblastic leukemia in children with Down syndrome: A retrospective analysis from the Ponte di Legno study group
- Author
-
Buitenkamp, T, Izraeli, S, Zimmermann, M, Forestier, E, Heerema, N, van den Heuvel Eibrink, M, Pieters, R, Korbijn, C, Silverman, L, Schmiegelow, K, Liang, D, Horibe, K, Arico, M, Biondi, A, Basso, G, Rabin, K, Schrappe, M, Cario, G, Mann, G, Morak, M, Panzer Grümayer, R, Mondelaers, V, Lammens, T, Cavé, H, Stark, B, Ganmore, I, Moorman, A, Vora, A, Hunger, S, Pui, C, Mullighan, C, Manabe, A, Escherich, G, Kowalczyk, J, Whitlock, J, Zwaan, C, Buitenkamp, TD, Heerema, NA, van den Heuvel Eibrink, MM, Korbijn, CM, Silverman, LB, Liang, DC, Rabin, KR, Moorman, AV, Hunger, SP, Pui, CH, Mullighan, CG, Kowalczyk, JR, Whitlock, JA, Zwaan, CM, BIONDI, ANDREA, Buitenkamp, T, Izraeli, S, Zimmermann, M, Forestier, E, Heerema, N, van den Heuvel Eibrink, M, Pieters, R, Korbijn, C, Silverman, L, Schmiegelow, K, Liang, D, Horibe, K, Arico, M, Biondi, A, Basso, G, Rabin, K, Schrappe, M, Cario, G, Mann, G, Morak, M, Panzer Grümayer, R, Mondelaers, V, Lammens, T, Cavé, H, Stark, B, Ganmore, I, Moorman, A, Vora, A, Hunger, S, Pui, C, Mullighan, C, Manabe, A, Escherich, G, Kowalczyk, J, Whitlock, J, Zwaan, C, Buitenkamp, TD, Heerema, NA, van den Heuvel Eibrink, MM, Korbijn, CM, Silverman, LB, Liang, DC, Rabin, KR, Moorman, AV, Hunger, SP, Pui, CH, Mullighan, CG, Kowalczyk, JR, Whitlock, JA, Zwaan, CM, and BIONDI, ANDREA
- Abstract
Children with Down syndrome (DS) have an increased risk of B-cell precursor (BCP) acute lymphoblastic leukemia (ALL). The prognostic factors and outcome of DS-ALL patients treated in contemporary protocols are uncertain. We studied 653 DS-ALL patients enrolled in 16 international trials from 1995 to 2004. Non-DS BCP-ALL patients from the Dutch Child Oncology Group and Berlin-Frankfurt- Münster were reference cohorts. DS-ALL patients had a higher 8-year cumulative incidence of relapse (26% ± 2% vs 15% ± 1%, P < .001) and 2-year treatment-related mortality (TRM) (7% ± 1% vs 2.0% ± <1%, P < .0001) than non-DS patients, resulting in lower 8-year event-free survival (EFS) (64% ± 2% vs 81% ± 2%, P < .0001) and overall survival (74% ± 2% vs 89% ± 1%, P < .0001). Independent favorable prognostic factors include age <6 years (hazard ratio [HR] = 0.58, P = .002), white blood cell (WBC) count <10 3 109/L (HR = 0.60, P = .005), and ETV6-RUNX1 (HR = 0.14, P = .006) for EFS and age (HR = 0.48, P < .001), ETV6-RUNX1 (HR = 0.1, P = .016) and high hyperdiploidy (HeH) (HR = 0.29, P = .04) for relapse-free survival. TRM was the major cause of death in ETV6-RUNX1 and HeH DSALLs. Thus, while relapse is the main contributor to poorer survival in DS-ALL, infection-associated TRM was increased in all protocol elements, unrelated to treatment phase or regimen. Future strategies to improve outcome in DS-ALL should include improved supportive care throughout therapy and reduction of therapy in newly identified good-prognosis subgroups. (Blood. 2014; 123(1):70-77). © 2014 by The American Society of Hematology.
- Published
- 2014
4. Volatile oil from the leaves ofCallistemon lanceolatus D.C. grown in north-eastern India
- Author
-
Rabin Kr. Sharma, Rumi Kotoky, and Pranab R. Bhattacharyya
- Subjects
Limonene ,biology ,Chemistry ,Myrtaceae ,General Chemistry ,biology.organism_classification ,Eastern india ,law.invention ,chemistry.chemical_compound ,law ,Botany ,Essential oil ,Callistemon lanceolatus ,Food Science - Abstract
The leaf essential oil of Callistemon lanceolatus D.C., obtained by hydrodistillation from matured leaves grown at Jorhat, Assam, in the north-eastern region of India, were analysed by GC and GC–MS. A total of 16 compounds were identified and 1,8 cineole (58.3%), α-pinene (21.2%), α-phellandrene (5.8%), limonene (4.1%) and α-terpineol (3.9%) were the major components representing more than 93% of the oil. Copyright © 2005 John Wiley & Sons, Ltd.
- Published
- 2006
- Full Text
- View/download PDF
5. Volatile oil from the leaves ofCallistemon lanceolatus D.C. grown in north-eastern India
- Author
-
Sharma, Rabin Kr., primary, Kotoky, Rumi, additional, and Bhattacharyya, Pranab R., additional
- Published
- 2006
- Full Text
- View/download PDF
6. Attacking remaining challenges in childhood leukemia.
- Author
-
Rabin KR
- Published
- 2012
7. Volatile oil from the leaves of Callistemon lanceolatus D.C. grown in north-eastern India.
- Author
-
Sharma, Rabin Kr., Kotoky, Rumi, and Bhattacharyya, Pranab R.
- Abstract
The leaf essential oil of Callistemon lanceolatus D.C., obtained by hydrodistillation from matured leaves grown at Jorhat, Assam, in the north-eastern region of India, were analysed by GC and GC-MS. A total of 16 compounds were identified and 1,8 cineole (58.3%), α-pinene (21.2%), α-phellandrene (5.8%), limonene (4.1%) and α-terpineol (3.9%) were the major components representing more than 93% of the oil. Copyright © 2005 John Wiley & Sons, Ltd. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
8. Genomic profiling in Down syndrome acute lymphoblastic leukemia identifies histone gene deletions associated with altered methylation profiles
- Author
-
Andrea Pession, Karen R. Rabin, Debra J. Morrison, Nyla A. Heerema, Deepa Bhojwani, Gregory Condos, Michael G. Loudin, Giuseppe Basso, Sivashankarappa Gurusiddappa, Hon-Chiu Eastwood Leung, Meenakshi Devidas, Sharon E. Plon, Julia Meyer, Stephen P. Hunger, Jinhua Wang, William L. Carroll, Anna Tsimelzon, Andrew J. Carroll, Loudin MG, Wang J, Leung HC, Gurusiddappa S, Meyer J, Condos G, Morrison D, Tsimelzon A, Devidas M, Heerema NA, Carroll AJ, Plon SE, Hunger SP, Basso G, Pession A, Bhojwani D, Carroll WL, and Rabin KR.
- Subjects
Cancer Research ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Histones ,Loss of heterozygosity ,Gene cluster ,Humans ,Cancer epigenetics ,DNA Primers ,Epigenomics ,Genetics ,Base Sequence ,Gene Expression Profiling ,acute lymphoblastic leukemia, CRLF2, Down syndrome, histone, JAK2 ,Hematology ,Methylation ,DNA Methylation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Molecular biology ,Gene expression profiling ,Histone ,Oncology ,Mutation ,DNA methylation ,biology.protein ,Down Syndrome ,Gene Deletion - Abstract
Patients with Down syndrome (DS) and acute lymphoblastic leukemia (ALL) have distinct clinical and biological features. Whereas most DS-ALL cases lack the sentinel cytogenetic lesions that guide risk assignment in childhood ALL, JAK2 mutations and CRLF2 overexpression are highly enriched. To further characterize the unique biology of DS-ALL, we performed genome-wide profiling of 58 DS-ALL and 68 non-Down syndrome (NDS) ALL cases by DNA copy number, loss of heterozygosity, gene expression, and methylation analyses. We report a novel deletion within the 6p22 histone gene cluster as significantly more frequent in DS-ALL, occurring in 11 DS (22%) and only two NDS cases (3.1%) (Fisher’s exact p = 0.002). Homozygous deletions yielded significantly lower histone expression levels, and were associated with higher methylation levels, distinct spatial localization of methylated promoters, and enrichment of highly methylated genes for specific pathways and transcription factor binding motifs. Gene expression profiling demonstrated heterogeneity of DS-ALL cases overall, with supervised analysis defining a 45-transcript signature associated with CRLF2 overexpression. Further characterization of pathways associated with histone deletions may identify opportunities for novel targeted interventions.
- Published
- 2011
- Full Text
- View/download PDF
9. Prevalence and clinical correlates ofJAK2mutations in Down syndrome acute lymphoblastic leukaemia
- Author
-
Andrea Pession, Karen R. Rabin, Giuseppe Basso, Meenakshi Devidas, Shai Izraeli, Cassia L. Rye, Amos Gaikwad, Nyla A. Heerema, Sharon E. Plon, Andrew J. Carroll, Gaikwad A., Rye C.L., Devidas M., Heerema NA., Carroll AJ., Izraeli S., Plon SE., Basso G., Pession A., and Rabin KR.
- Subjects
Male ,medicine.medical_specialty ,Down syndrome ,Aneuploidy ,Kaplan-Meier Estimate ,Biology ,medicine.disease_cause ,Gastroenterology ,Article ,Disease-Free Survival ,Sex Factors ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,Internal medicine ,medicine ,Humans ,Point Mutation ,Child ,Mutation ,Hematology ,Point mutation ,Age Factors ,Sequence Analysis, DNA ,Janus Kinase 2 ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Diploidy ,Enzyme Activation ,Child, Preschool ,Immunology ,Hyperdiploidy ,Down Syndrome ,Trisomy - Abstract
Recurrent, prognostically significant chromosomal abnormalities occur in approximately 75% of paediatric acute lymphoblastic leukaemia (ALL), but only infrequently in children with Down syndrome (DS) and ALL. Recently, novel somatic activating mutations in the gene Janus kinase 2 (JAK2) were reported in 18% of DS ALL. Here we report identification and clinical correlates of JAK2 mutations in an independent cohort. JAK2 activating mutations occurred in 10/53 DS ALL cases (18.9%). Mutations were overrepresented in males (P < 0.03), occurred once in association with high hyperdiploidy and were not significantly correlated with age, initial white blood count, or event-free survival. Our results confirm the significance of JAK-STAT pathway activation in DS ALL.
- Published
- 2009
- Full Text
- View/download PDF
10. Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia
- Author
-
Richard T. Williams, Julia Meyer, Nyla A. Heerema, Charles G. Mullighan, Stephen P. Hunger, Jing Ma, William L. Carroll, Cheryl L. Willman, Fady M. Mikhail, Wei Liu, Giuseppe Basso, Letha A. Phillips, Jinjun Cheng, Andrew J. Carroll, Jinghui Zhang, Richard C. Harvey, Andrea Pession, Karen R. Rabin, Susana C. Raimondi, Elaine Coustan-Smith, Michael G. Loudin, J. Racquel Collins-Underwood, James R. Downing, Ching-Hon Pui, Mullighan CG, Collins-Underwood JR, Phillips LA, Loudin MG, Liu W, Zhang J, Ma J, Coustan-Smith E, Harvey RC, Willman CL, Mikhail FM, Meyer J, Carroll AJ, Williams RT, Cheng J, Heerema NA, Basso G, Pession A, Pui CH, Raimondi SC, Hunger SP, Downing JR, Carroll WL, and Rabin KR.
- Subjects
Down syndrome ,Recombinant Fusion Proteins ,Pseudoautosomal region ,Aneuploidy ,Chromosomal translocation ,Biology ,Article ,Cell Line ,Exon ,Mice ,Acute lymphocytic leukemia ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Genetics ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Receptors, Cytokine ,Gene Rearrangement ,Acute leukemia ,Base Sequence ,Receptors, Purinergic P2 ,Gene rearrangement ,Janus Kinase 2 ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Cancer research ,Chromosome Deletion ,Down Syndrome - Abstract
Aneuploidy and translocations are hallmarks of B-progenitor acute lymphoblastic leukemia (ALL), but many patients lack a recurring chromosomal alteration. Here we report a recurring interstitial deletion of the pseudoautosomal region 1 of chromosomes X and Y in B-progenitor ALL that juxtaposes the first, non-coding exon of P2RY8 to the coding region of CRLF2 (which encodes cytokine receptor like factor 2, or thymic stromal lymphopoietin receptor). The P2RY8-CRLF2 fusion was identified in 7% of B-progenitor ALL cases, and was identified in over 50% of ALL cases arising in patients with Down syndrome (53% of 75 cases). CRLF2 alteration was associated with the presence of activating JAK mutations, and expression of P2RY8-CRLF2 together with JAK2 mutants resulted in constitutive Jak-Stat activation and cytokine-independent growth of Ba/F3-IL7R cells, indicating that these two genetic lesions together contribute to leukemogenesis in B-progenitor ALL.
- Published
- 2009
11. The impact of Indigenous American-like ancestry on risk of acute lymphoblastic leukemia in Hispanic/Latino children.
- Author
-
Langie J, Chan TF, Yang W, Kang AY, Morimoto L, Stram DO, Mancuso N, Ma X, Metayer C, Lupo PJ, Rabin KR, Scheurer ME, Wiemels JL, Yang JJ, de Smith AJ, and Chiang CWK
- Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer, with Hispanic/Latino children having a higher incidence of ALL than other racial/ethnic groups. Genetic variants, particularly ones found enriched in Indigenous American (IA)-like ancestry and inherited by Hispanics/Latinos, may contribute to this disparity. In this study, we characterized the impact of IA-like ancestry on overall ALL risk and the frequency and effect size of known risk alleles in a large cohort of self-reported Hispanic/Latino individuals. We also performed genome-wide admixture mapping analysis to identify potentially novel ALL risk loci. We found that global IA ancestry was positively associated with ALL risk, but the association was not significant after adjusting for socio-economic indicators. In a series of local ancestry analyses, we uncovered that at known ALL risk loci, increasing copies of the IA-like haplotype were positively and significantly associated with ALL case-control status. Further, the IA-like haplotype had ∼1.33 times the odds of harboring the risk allele compared to non-IA-like haplotypes. We found no evidence of interaction between genotype and ancestry (local or global) in relation to ALL risk. Admixture mapping identified association signals on chromosomes 2 (2q21.2), 7 (7p12.2), 10 (10q21.2), and 15 (15q22.31); however, only the variants at 7p12.2 and 10q21.2 replicated in additional cohorts. Taken together, our results suggest that increased risk of ALL in Hispanic/Latino children may be conferred by higher frequency of risk alleles within IA-like ancestry, which can be leveraged as targets of new precision health strategies and therapeutics.
- Published
- 2025
- Full Text
- View/download PDF
12. Imagine, Discover, Inspire: Proceedings of the 4th International Conference of the Trisomy 21 Research Society.
- Author
-
Flores-Aguilar L, Hamlett ED, Araya P, Barone E, Bhattacharyya A, Carmona-Iragui M, Chan L, Christian B, Costa ACS, Costanzo F, Del Hoyo Soriano L, Dierssen M, Eichler EE, Fisher E, Galbraith M, Ghosh S, Gimenez S, Guedj F, Guidi S, Iulita MF, Mobley W, Pelleri MC, Potier MC, Rabin KR, Rachubinski A, Rebillat AS, Rubenstein E, Saternos H, Sordo L, Strydom A, Valle-Tamayo N, Waugh KA, Yu E, Zeldich E, Busciglio J, and Head E
- Subjects
- Humans, Animals, Female, Adult, Quality of Life, Child, Biomedical Research, Down Syndrome psychology
- Abstract
Down syndrome (DS) or trisomy 21 (T21) is present in a significant number of children and adults around the world and is associated with cognitive and medical challenges. Through research, the T21 Research Society (T21RS), established in 2014, unites a worldwide community dedicated to understanding the impact of T21 on biological systems and improving the quality of life of people with DS across the lifespan. T21RS hosts an international conference every two years to support collaboration, dissemination, and information sharing for this goal. In 2022, T21RS hosted an international conference in Long Beach, California, from June 9 to 12. The conference, attended by 483 people including scientists, families, self-advocates, and industry representatives from 17 countries, was a dynamic and interactive meeting that shared discoveries from international research teams. This summary highlights the scientific discoveries shared at the 4th T21RS meeting with the Imagine, Discover, Inspire theme., Competing Interests: Declarations. Conflict of interests: E.E.E. is a member of the scientific advisory board of Variant Bio, Inc. The other authors have no conflicts of interest to declare., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
13. Delayed excretion of high-dose methotrexate in pediatric acute leukemia correlates with laxative and constipation management.
- Author
-
Belsky JA, Chavana A, Banerjee A, Zobeck M, Schafer ES, Rabin KR, Gramatges M, Sim A, Leisinger A, Reddy T, Parod MJ, Lupo P, Scheurer M, Brown AL, and Bernhardt MB
- Subjects
- Humans, Male, Female, Child, Child, Preschool, Adolescent, Retrospective Studies, Follow-Up Studies, Prognosis, Infant, Methotrexate administration & dosage, Methotrexate pharmacokinetics, Methotrexate adverse effects, Constipation chemically induced, Constipation drug therapy, Laxatives administration & dosage, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Antimetabolites, Antineoplastic administration & dosage, Antimetabolites, Antineoplastic adverse effects, Antimetabolites, Antineoplastic pharmacokinetics
- Abstract
Background: Delayed excretion of high-dose methotrexate (HD-MTX) in pediatric acute lymphoblastic leukemia (ALL) can result in significant morbidity. While methotrexate is primarily renally excreted, HD-MTX may overwhelm renal excretion and increase reliance on fecal elimination. This study evaluated the association between laxative use for constipation and delayed excretion of HD-MTX., Methods: This multisite chart review included pediatric patients with ALL (2010-2020) who received HD-MTX (5 g/m
2 ). Delayed excretion was defined as a serum MTX concentration greater than 0.4 µM at Hour 48. We identified use of laxative medications after each HD-MTX infusion, with receipt of two or more doses considered a proxy for constipation. Multilevel logistic regression models evaluated associations between clinical factors and delayed HD-MTX excretion to account for multiple MTX cycles per individual., Results: A total of 533 eligible patients received 1875 HD-MTX infusions. Patients were mostly male (59.8%), Hispanic (56.7%), with a median age of 9.5 years. Delayed excretion was observed following 42.7% of HD-MTX infusions, and patients received two or more laxative doses during 19.9% of infusions. Independent of other factors, individuals who received two or more laxative doses were nearly 60% (odds ratio 1.58; 95% confidence interval: 1.19-2.09; p = .002) more likely to experience delayed excretion compared to those receiving fewer than two laxative doses., Conclusion: Receipt of at least two laxative doses was independently associated with delayed methotrexate excretion in pediatric patients with ALL. Future prospective studies are needed to confirm the secondary effects of constipation and confirm the association with constipation and identify clinical benefits that optimize drug excretion., (© 2024 The Author(s). Pediatric Blood & Cancer published by Wiley Periodicals LLC.)- Published
- 2025
- Full Text
- View/download PDF
14. Reply to: Accurate Determinants of Outcome in ALL.
- Author
-
Chang TC, Chen W, Qu C, Cheng Z, Elsayed A, Pounds SB, Shago M, Rabin KR, Raetz EA, Devidas M, Cheng C, Angiolillo A, Baviskar P, Borowitz M, Burke MJ, Carroll A, Carroll WL, Chen IM, Harvey R, Heerema N, Iacobucci I, Wang JR, Jeha S, Larsen E, Mattano L, Maloney K, Pui CH, Ramirez NC, Salzer W, Willman C, Winick N, Wood B, Hunger SP, Wu G, Mullighan CG, and Loh ML
- Published
- 2024
- Full Text
- View/download PDF
15. Co-targeting of the thymic stromal lymphopoietin receptor to decrease immunotherapeutic resistance in CRLF2-rearranged Ph-like and Down syndrome acute lymphoblastic leukemia.
- Author
-
Balestra T, Niswander LM, Bagashev A, Loftus JP, Ross SL, Chen RK, McClellan SM, Junco JJ, Bárcenas López DA, Rabin KR, Fry TJ, and Tasian SK
- Abstract
CRLF2 rearrangements occur in >50% of Ph-like and Down syndrome (DS)-associated B-acute lymphoblastic leukemia (ALL) and induce constitutive kinase signaling targetable by the JAK1/2 inhibitor ruxolitinib under current clinical investigation. While chimeric antigen receptor T cell (CART) immunotherapies have achieved remarkable remission rates in children with relapsed/refractory B-ALL, ~50% of CD19CART-treated patients relapse again, many with CD19 antigen loss. We previously reported preclinical activity of thymic stromal lymphopoietin receptor-targeted cellular immunotherapy (TSLPRCART) against CRLF2-overexpressing ALL as an alternative approach. In this study, we posited that combinatorial TSLPRCART and ruxolitinib would have superior activity and first validated potent TSLPRCART-induced inhibition of leukemia proliferation in vitro in CRLF2-rearranged ALL cell lines and in vivo in Ph-like and DS-ALL patient-derived xenograft (PDX) models. However, simultaneous TSLPRCART/ruxolitinib or CD19CART/ruxolitinib treatment during initial CART expansion diminished T cell proliferation, blunted cytokine production, and/or facilitated leukemia relapse, which was abrogated by time-sequenced/delayed ruxolitinib co-exposure. Importantly, ruxolitinib co-administration prevented fatal TSLPRCART cytokine-associated toxicity in ALL PDX mice. Upon ruxolitinib withdrawal, TSLPRCART functionality recovered in vivo with clearance of subsequent ALL rechallenge. These translational studies demonstrate an effective two-pronged therapeutic strategy that mitigates acute CART-induced hyperinflammation and provides potential anti-leukemia 'maintenance' relapse prevention for CRLF2-rearranged Ph-like and DS-ALL., Competing Interests: Competing interests: AB is a current employee of Carisma Therapeutics. SLR is a current employee of Parexel International. KRR has received preclinical research support from Incyte Corporation. TJF was a prior part-time employee of and consultant for Sana Biotechnology. TJF is an inventor on patent US11834509B2 (‘Thymic stromal lymphopoietin receptor-specific chimeric antigen receptors and methods using same’). SKT receives clinical research funding from Incyte Corporation for conduction of the Children’s Oncology Group AALL1521 phase 2 clinical trial (NCT02723994). The remaining authors declare no competing interests. Ethics approval and consent to participate: Viably cryopreserved primary pediatric, adolescent, and young adult ALL specimens used to create PDX models for these studies were obtained from leukemia biorepositories of the Children’s Oncology Group, Children’s Hospital of Philadelphia (CHOP), or Texas Children’s Hospital under institutional review board (IRB)-approved research protocols following obtainment of informed consent in accordance with the Declaration of Helsinki. Use of coded leukemia specimens without identifying patient health information in these studies was deemed non-human subjects research and exempt from further review by the CHOP IRB and ethics committee. All animal studies were conducted under an Institutional Animal Care and Use Committee-approved protocol at CHOP in accordance with all guidelines and regulations., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Blinatumomab in Standard-Risk B-Cell Acute Lymphoblastic Leukemia in Children.
- Author
-
Gupta S, Rau RE, Kairalla JA, Rabin KR, Wang C, Angiolillo AL, Alexander S, Carroll AJ, Conway S, Gore L, Kirsch I, Kubaney HR, Li AM, McNeer JL, Militano O, Miller TP, Moyer Y, O'Brien MM, Okada M, Reshmi SC, Shago M, Wagner E, Winick N, Wood BL, Haworth-Wright T, Zaman F, Zugmaier G, Zupanec S, Devidas M, Hunger SP, Teachey DT, Raetz EA, and Loh ML
- Abstract
Background: B-cell acute lymphoblastic leukemia (B-cell ALL) is the most common childhood cancer. Despite a high overall cure rate, relapsed B-cell ALL remains a leading cause of cancer-related death among children. The addition of the bispecific T-cell engager molecule blinatumomab (an anti-CD19 and anti-CD3 single-chain molecule) to therapy for newly diagnosed standard-risk (as defined by the National Cancer Institute) B-cell ALL in children may improve outcomes., Methods: We conducted a phase 3 trial involving children with newly diagnosed standard-risk B-cell ALL who had an average or high risk of relapse. Patients were randomly assigned to receive chemotherapy alone or chemotherapy plus two nonsequential 28-day cycles of blinatumomab. The primary end point was disease-free survival., Results: The data and safety monitoring committee reviewed the results from the first interim efficacy analysis, which included 1440 patients who had undergone randomization (722 to chemotherapy alone and 718 to blinatumomab and chemotherapy) and recommended early termination of randomization. At a median follow-up of 2.5 years, the estimated 3-year disease-free survival (±SE) was 96.0±1.2% with blinatumomab and chemotherapy and 87.9±2.1% with chemotherapy alone (difference in restricted mean survival time, 72 days; 95% confidence interval, 36 to 108; P<0.001 by stratified log-rank test). The estimated 3-year disease-free survival among patients with an average relapse risk was 97.5±1.3% with blinatumomab and chemotherapy and 90.2±2.3% with chemotherapy alone; among those with a high relapse risk, the corresponding values were 94.1±2.5% and 84.8±3.8%. Cytokine release syndrome, seizures, and sepsis of grade 3 or higher were rare during blinatumomab cycles, but the overall incidence of nonfatal sepsis and catheter-related infections was significantly higher among patients with an average relapse risk who had been assigned to receive blinatumomab and chemotherapy than among those assigned to receive chemotherapy alone., Conclusions: Adding blinatumomab to combination chemotherapy in patients with newly diagnosed childhood standard-risk B-cell ALL of average or high risk of relapse significantly improved disease-free survival. (Funded by the National Institutes of Health and others; AALL1731 ClinicalTrials.gov number, NCT03914625.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
17. A transgenic mouse model of Down syndrome acute lymphoblastic leukemia identifies targetable vulnerabilities.
- Author
-
Junco JJ, Rochette M, Alozie M, Rashid R, Terrell M, Zorman B, Sumazin P, Rowland L, Dettorre G, Powell RT, Stephan CC, Davies PJ, Martinez-Moczygemba M, Yang JJ, and Rabin KR
- Published
- 2024
- Full Text
- View/download PDF
18. Genomic Determinants of Outcome in Acute Lymphoblastic Leukemia.
- Author
-
Chang TC, Chen W, Qu C, Cheng Z, Hedges D, Elsayed A, Pounds SB, Shago M, Rabin KR, Raetz EA, Devidas M, Cheng C, Angiolillo A, Baviskar P, Borowitz M, Burke MJ, Carroll A, Carroll WL, Chen IM, Harvey R, Heerema N, Iacobucci I, Wang JR, Jeha S, Larsen E, Mattano L, Maloney K, Pui CH, Ramirez NC, Salzer W, Willman C, Winick N, Wood B, Hunger SP, Wu G, Mullighan CG, and Loh ML
- Subjects
- Humans, Child, Male, Female, Case-Control Studies, Child, Preschool, Adolescent, Genomics, Infant, PAX5 Transcription Factor genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Purpose: 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: 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: Genomic subtype was associated with relapse, which occurred in approximately 50% of cases of PAX5 -altered ALL (odds ratio [OR], 3.31 [95% CI, 2.17 to 5.03]; P = 3.18 × 10
-8 ). Within high-hyperdiploid ALL, gain of chromosome 10 with disomy of chromosome 7 was associated with favorable outcome (OR, 0.27 [95% CI, 0.17 to 0.42]; P = 8.02 × 10-10 ; St Jude Children's Research Hospital validation cohort: OR, 0.22 [95% CI, 0.05 to 0.80]; P = .009), and disomy of chromosomes 10 and 17 with gain of chromosome 6 was associated with relapse (OR, 7.16 [95% CI, 2.63 to 21.51]; P = 2.19 × 10-5 ; validation cohort: OR, 21.32 [95% CI, 3.62 to 119.30]; P = .0004). Genomic alterations were associated with relapse in a subtype-dependent manner, including alterations of INO80 in ETV6::RUNX1 ALL, IKZF1 , and CREBBP in high-hyperdiploid ALL and FHIT in BCR::ABL1 -like ALL. Genomic alterations were also associated with the presence of minimal residual disease, including NRAS and CREBBP in high-hyperdiploid ALL., Conclusion: 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.- Published
- 2024
- Full Text
- View/download PDF
19. Genome-wide association studies of Down syndrome associated congenital heart defects.
- Author
-
Feldman ER, Li Y, Cutler DJ, Rosser TC, Wechsler SB, Sanclemente L, Rachubinski AL, Elliott N, Vyas P, Roberts I, Rabin KR, Wagner M, Gelb BD, Espinosa JM, Lupo PJ, de Smith AJ, Sherman SL, and Leslie EJ
- Abstract
Congenital heart defects (CHDs) are the most common structural birth defect and are present in 40-50% of children born with Down syndrome (DS). To characterize the genetic architecture of DS-associated CHD, we sequenced genomes of a multiethnic group of children with DS and a CHD (n=886: atrioventricular septal defects (AVSD), n=438; atrial septal defects (ASD), n=122; ventricular septal defects (VSD), n=170; other types of CHD, n=156) and DS with a structurally normal heart (DS+NH, n=572). We performed four GWAS for common variants (MAF>0.05) comparing DS with CHD, stratified by CHD-subtype, to DS+NH controls. Although no SNP achieved genome-wide significance, multiple loci in each analysis achieved suggestive significance (p<2×10
-6 ). Of these, the 1p35.1 locus (near RBBP4 ) was specifically associated with ASD risk and the 5q35.2 locus (near MSX2 ) was associated with any type of CHD. Each of the suggestive loci contained one or more plausible candidate genes expressed in the developing heart. While no SNP replicated (p<2×10-6 ) in an independent cohort of DS+CHD (DS+CHD: n=229; DS+NH: n=197), most SNPs that were suggestive in our GWASs remained suggestive when meta-analyzed with the GWASs from the replication cohort. These results build on previous work to identify genetic modifiers of DS-associated CHD.- Published
- 2024
- Full Text
- View/download PDF
20. Episodes of acute methotrexate-related neurotoxicity linked to compromised long-term neurocognitive function.
- Author
-
Harris RD, Taylor OA, Raghubar KP, Matheus Gonzalez M, Zobeck M, Gramatges MM, Rabin KR, Scheurer ME, and Brown AL
- Subjects
- Humans, Female, Male, Child, Child, Preschool, Adolescent, Follow-Up Studies, Neuropsychological Tests, Prognosis, Methotrexate adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Neurotoxicity Syndromes etiology, Antimetabolites, Antineoplastic adverse effects
- Abstract
Methotrexate is a critical component of curative chemotherapy for pediatric acute lymphoblastic leukemia (ALL), but is associated with neurotoxicity. Information on long-term outcomes following an acute neurotoxic event is limited. Therefore, this report compares neurocognitive performance more than 12 months post diagnosis (mean = 4 years) between ALL patients with (n = 25) and without (n = 146) a history of acute neurotoxicity. Compared to children with no documented on-treatment neurotoxic event, children who experienced a neurotoxic event during treatment exhibited poorer performance on measures of fine motor function (p = .02) and attention (p = .02). Children with ALL who experience acute neurotoxicity may be candidates for early neuropsychological screening and intervention., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
21. Enhanced Risk Stratification for Children and Young Adults with B-Cell Acute Lymphoblastic Leukemia: A Children's Oncology Group Report.
- Author
-
DelRocco NJ, Loh ML, Borowitz MJ, Gupta S, Rabin KR, Zweidler-McKay P, Maloney KW, Mattano LA, Larsen E, Angiolillo A, Schore RJ, Burke MJ, Salzer WL, Wood BL, Carroll AJ, Heerema NA, Reshmi SC, Gastier-Foster JM, Harvey R, Chen IM, Roberts KG, Mullighan CG, Willman C, Winick N, Carroll WL, Rau RE, Teachey DT, Hunger SP, Raetz EA, Devidas M, and Kairalla JA
- Subjects
- Child, Humans, Young Adult, Prognosis, Recurrence, Risk Assessment, Disease-Free Survival, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Burkitt Lymphoma
- Abstract
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 (PI
COG ) 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., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
22. Outcomes in Children, Adolescents, and Young Adults With Down Syndrome and ALL: A Report From the Children's Oncology Group.
- Author
-
Rabin KR, Devidas M, Chen Z, Ji L, Kairalla J, Hitzler JK, Yang JJ, Carroll AJ, Heerema NA, Borowitz MJ, Wood BL, Roberts KG, Mullighan CG, Harvey RC, Chen IM, Willman CL, Reshmi SC, Gastier-Foster JM, Bhojwani D, Rheingold SR, Maloney KW, Mattano LA, Larsen EC, Schore RJ, Burke MJ, Salzer WL, Winick NJ, Carroll WL, Raetz EA, Loh ML, Hunger SP, and Angiolillo AL
- Subjects
- Child, Humans, Adolescent, Young Adult, Infant, Child, Preschool, Adult, Treatment Outcome, Disease-Free Survival, Neoplasm Recurrence, Local complications, Recurrence, Neoplasm, Residual, Down Syndrome complications, Down Syndrome therapy
- Abstract
Purpose: Patients with Down syndrome (DS) and B-ALL experience increased rates of relapse, toxicity, and death. We report results for patients with DS B-ALL enrolled on Children's Oncology Group trials between 2003 and 2019., Methods: We analyzed data for DS (n = 743) and non-DS (n = 20,067) patients age 1-30 years on four B-ALL standard-risk (SR) and high-risk trials., Results: Patients with DS exhibited more frequent minimal residual disease (MRD) ≥0.01% at end induction (30.8% v 21.5%; P < .001). This difference persisted at end consolidation only in National Cancer Institute (NCI) high-risk patients (34.0% v 11.7%; P < .0001). Five-year event-free survival (EFS) and overall survival (OS) were significantly poorer for DS versus non-DS patients overall (EFS, 79.2% ± 1.6% v 87.5% ± 0.3%; P < .0001; OS, 86.8% ± 1.4% v 93.6% ± 0.2%; P < .0001), and within NCI SR and high-risk subgroups. Multivariable Cox regression analysis of the DS cohort for risk factors associated with inferior EFS identified age >10 years, white blood count >50 × 10
3 /μL, and end-induction MRD ≥0.01%, but not cytogenetics or CRLF2 overexpression. Patients with DS demonstrated higher 5-year cumulative incidence of relapse (11.5% ± 1.2% v 9.1% ± 0.2%; P = .0008), death in remission (4.9% ± 0.8% v 1.7% ± 0.1%; P < .0001), and induction death (3.4% v 0.8%; P < .0001). Mucositis, infections, and hyperglycemia were significantly more frequent in all patients with DS, while seizures were more frequent in patients with DS on high-risk trials (4.1% v 1.8%; P = .005)., Conclusion: Patients with DS-ALL exhibit an increased rate of relapse and particularly of treatment-related mortality. Novel, less-toxic therapeutic strategies are needed to improve outcomes.- Published
- 2024
- Full Text
- View/download PDF
23. Racial and ethnic disparities in acuity of presentation among children with newly diagnosed acute leukemia.
- Author
-
Winestone LE, Getz KD, Li Y, Burrows E, Scheurer ME, Tam V, Gramatges MM, Wilkes JJ, Miller TP, Seif AE, Rabin KR, Fisher BT, and Aplenc R
- Subjects
- Child, Humans, Racial Groups, Hospitals, Pediatric, Healthcare Disparities, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
We evaluated disparities in disease burden, organ dysfunction, vital signs, and timing of therapy in children newly presenting with acute leukemia. Among 899 patients with acute leukemia diagnosed at two large children's hospitals, a priori lab-based definitions of high disease burden, infection risk, renal dysfunction, and coagulopathy were applied to electronic health record data. Black patients with acute myeloid leukemia had increased prevalence of elevated white blood cell count and uric acid; Black patients with acute lymphoblastic leukemia demonstrated increased prevalence of coagulopathy. Black patients' presentation more frequently included multiple lab abnormalities consistent with advanced physiologic dysfunction. No differences were found in days to therapy initiation., (© 2023 The Authors. Pediatric Blood & Cancer published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
24. Prognostic significance of ETP phenotype and minimal residual disease in T-ALL: a Children's Oncology Group study.
- Author
-
Wood BL, Devidas M, Summers RJ, Chen Z, Asselin B, Rabin KR, Zweidler-McKay PA, Winick NJ, Borowitz MJ, Carroll WL, Raetz EA, Loh ML, Hunger SP, Dunsmore KP, Teachey DT, and Winter SS
- Subjects
- Child, Humans, Young Adult, Disease-Free Survival, Neoplasm, Residual diagnosis, Prognosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma diagnosis
- Abstract
The early thymic precursor (ETP) immunophenotype was previously reported to confer poor outcome in T-cell acute lymphoblastic leukemia (T-ALL). Between 2009 and 2014, 1256 newly diagnosed children and young adults enrolled in Children's Oncology Group (COG) AALL0434 were assessed for ETP status and minimal residual disease (MRD) using flow cytometry at a central reference laboratory. The subject phenotypes were categorized as ETP (n = 145; 11.5%), near-ETP (n = 209; 16.7%), or non-ETP (n = 902; 71.8%). Despite higher rates of induction failure for ETP (6.2%) and near-ETP (6.2%) than non-ETP (1.2%; P < .0001), all 3 groups showed excellent 5-year event-free survival (EFS) and overall survival (OS): ETP (80.4% ± 3.9% and 86.8 ± 3.4%, respectively), near-ETP (81.1% ± 3.3% and 89.6% ± 2.6%, respectively), and non-ETP (85.3% ± 1.4% and 90.0% ± 1.2%, respectively; P = .1679 and P = .3297, respectively). There was no difference in EFS or OS for subjects with a day-29 MRD <0.01% vs 0.01% to 0.1%. However, day-29 MRD ≥0.1% was associated with inferior EFS and OS for patients with near-ETP and non-ETP, but not for those with ETP. For subjects with day-29 MRD ≥1%, end-consolidation MRD ≥0.01% was a striking predictor of inferior EFS (80.9% ± 4.1% vs 52.4% ± 8.1%, respectively; P = .0001). When considered as a single variable, subjects with all 3 T-ALL phenotypes had similar outcomes and subjects with persistent postinduction disease had inferior outcomes, regardless of their ETP phenotype. This clinical trial was registered at AALL0434 as #NCT00408005., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
25. Acute and chronic kidney injury during therapy for pediatric acute leukemia: A report from the Leukemia Electronic Abstraction of Records Network (LEARN).
- Author
-
Hsiao W, Li Y, Getz K, Cao L, Krause E, Ramos M, Lee J, Gramatges MM, Rabin KR, Scheurer ME, Aplenc R, Denburg M, and Miller TP
- Subjects
- Child, Humans, Retrospective Studies, Hospital Mortality, Kidney, Electronics, Risk Factors, Acute Kidney Injury chemically induced, Acute Kidney Injury epidemiology, Renal Insufficiency, Chronic epidemiology, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute complications
- Abstract
Children with acute leukemia are at increased risk of kidney injury. Using electronic health record data from three centers between 2010 and 2018, this study retrospectively described acute kidney injury (AKI) and chronic kidney disease (CKD) prevalence in children with acute lymphoblastic or myeloid leukemia (ALL, AML) using Common Terminology Criteria for Adverse Events (CTCAE) and Kidney Disease Improving Global Outcomes (KDIGO) definitions. AKI during therapy was 25% (ALL) and 32% (AML) using CTCAE, versus 84% (ALL) and 74% (AML) using KDIGO. CKD prevalence was low and Grade 1/Stage 2. Further investigation is needed to optimally define kidney injury in acute leukemia., (© 2023 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
26. Survival outcomes of children with relapsed or refractory myeloid leukemia associated with Down syndrome.
- Author
-
Raghuram N, Hasegawa D, Nakashima K, Rahman S, Antoniou E, Skajaa T, Merli P, Verma A, Rabin KR, Aftandilian C, Kotecha RS, Cheuk D, Jahnukainen K, Kolenova A, Balwierz W, Norton A, O'Brien M, Cellot S, Chopek A, Arad-Cohen N, Goemans B, Rojas-Vasquez M, Ariffin H, Bartram J, Kolb EA, Locatelli F, Klusmann JH, Hasle H, McGuire B, Hasnain A, Sung L, and Hitzler J
- Subjects
- Humans, Child, Retrospective Studies, Recurrence, Down Syndrome complications, Down Syndrome therapy, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute
- Abstract
Children with Down syndrome (DS) are at a significantly higher risk of developing acute myeloid leukemia, also termed myeloid leukemia associated with DS (ML-DS). In contrast to the highly favorable prognosis of primary ML-DS, the limited data that are available for children who relapse or who have refractory ML-DS (r/r ML-DS) suggest a dismal prognosis. There are few clinical trials and no standardized treatment approach for this population. We conducted a retrospective analysis of international study groups and pediatric oncology centers and identified 62 patients who received treatment with curative intent for r/r ML-DS between year 2000 to 2021. Median time from diagnosis to relapse was 6.8 (range, 1.1-45.5) months. Three-year event-free survival (EFS) and overall survival (OS) were 20.9 ± 5.3% and 22.1 ± 5.4%, respectively. Survival was associated with receipt of hematopoietic stem cell transplantation (HSCT) (hazard ratio [HR], 0.28), duration of first complete remission (CR1) (HR, 0.31 for > 12 months) and attainment of remission after relapse (HR, 4.03). Patients who achieved complete remission (CR) before HSCT, had an improved OS and EFS of 56.0 ± 11.8% and 50.5 ± 11.9%, respectively compared to those who underwent HSCT without CR (3-year OS and EFS of 10.0 ± 9.5%). Treatment failure after HSCT was predominantly because of disease recurrence (52%) followed by treatment-related mortality (10%). The prognosis of r/r ML-DS remains dismal even in the current treatment period and serve as a reference point for current prognostication and future interventional studies. Clinical trials aimed at improving the survival of patients with r/r ML-DS are needed., (Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
27. Down syndrome and leukemia: from basic mechanisms to clinical advances.
- Author
-
Baruchel A, Bourquin JP, Crispino J, Cuartero S, Hasle H, Hitzler J, Klusmann JH, Izraeli S, Lane AA, Malinge S, Rabin KR, Roberts I, Ryeom S, Tasian SK, and Wagenblast E
- Subjects
- Child, Humans, Acute Disease, France, Down Syndrome complications, Down Syndrome genetics, Leukemia, Myeloid, Acute epidemiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Children with Down syndrome (DS, trisomy 21) are at a significantly higher risk of developing acute leukemia compared to the overall population. Many studies investigating the link between trisomy 21 and leukemia initiation and progression have been conducted over the last two decades. Despite improved treatment regimens and significant progress in iden - tifying genes on chromosome 21 and the mechanisms by which they drive leukemogenesis, there is still much that is unknown. A focused group of scientists and clinicians with expertise in leukemia and DS met in October 2022 at the Jérôme Lejeune Foundation in Paris, France for the 1st International Symposium on Down Syndrome and Leukemia. This meeting was held to discuss the most recent advances in treatment regimens and the biology underlying the initiation, progression, and relapse of acute lymphoblastic leukemia and acute myeloid leukemia in children with DS. This review provides a summary of what is known in the field, challenges in the management of DS patients with leukemia, and key questions in the field.
- Published
- 2023
- Full Text
- View/download PDF
28. Management of Down Syndrome-Associated Leukemias: A Review.
- Author
-
Verma A, Lupo PJ, Shah NN, Hitzler J, and Rabin KR
- Subjects
- Infant, Child, Adolescent, Young Adult, Humans, Child, Preschool, Down Syndrome complications, Down Syndrome genetics, Leukemoid Reaction complications, Leukemoid Reaction genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Importance: Down syndrome (DS), caused by an extra copy of material from chromosome 21, is one of the most common genetic conditions. The increased risk of acute leukemia in DS (DS-AL) has been recognized for decades, consisting of an approximately 150-fold higher risk of acute myeloid leukemia (AML) before age 4 years, and a 10- to 20-fold higher risk of acute lymphoblastic leukemia (ALL), compared with children without DS., Observations: A recent National Institutes of Health-sponsored conference, ImpacT21, reviewed research and clinical trials in children, adolescents, and young adults (AYAs) with DS-AL and are presented herein, including presentation and treatment, clinical trial design, and ethical considerations for this unique population. Between 10% to 30% of infants with DS are diagnosed with transient abnormal myelopoiesis (TAM), which spontaneously regresses. After a latency period of up to 4 years, 20% to 30% develop myeloid leukemia associated with DS (ML-DS). Recent studies have characterized somatic mutations associated with progression from TAM to ML-DS, but predicting which patients will progress to ML-DS remains elusive. Clinical trials for DS-AL have aimed to reduce treatment-related mortality (TRM) and improve survival. Children with ML-DS have better outcomes compared with non-DS AML, but outcomes remain dismal in relapse. In contrast, patients with DS-ALL have inferior outcomes compared with those without DS, due to both higher TRM and relapse. Management of relapsed leukemia poses unique challenges owing to disease biology and increased vulnerability to toxic effects. Late effects in survivors of DS-AL are an important area in need of further study because they may demonstrate unique patterns in the setting of chronic medical conditions associated with DS., Conclusions and Relevance: Optimal management of DS-AL requires specific molecular testing, meticulous supportive care, and tailored therapy to reduce TRM while optimizing survival. There is no standard approach to treatment of relapsed disease. Future work should include identification of biomarkers predictive of toxic effects; enhanced clinical and scientific collaborations; promotion of access to novel agents through innovative clinical trial design; and dedicated studies of late effects of treatment.
- Published
- 2023
- Full Text
- View/download PDF
29. Children's Oncology Group blueprint for research: Acute lymphoblastic leukemia.
- Author
-
Raetz EA, Bhojwani D, Devidas M, Gore L, Rabin KR, Tasian SK, Teachey DT, and Loh ML
- Subjects
- Humans, Child, Central Nervous System, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Treatment Outcome, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Cure rates for acute lymphoblastic leukemia (ALL), the most common childhood cancer have steadily improved over the past five decades. This is due to intensifying systemic therapy, recognizing and treating the central nervous system as a sanctuary site, and implementing modern risk stratification to deliver varying intensities of therapy based on age, presenting white blood count, sentinel somatic genetics, and therapy response. Recently, numerous Children's Oncology Group trials have demonstrated the lack of benefit of intensifying traditional chemotherapy, providing evidence that new approaches are needed to cure the patients for whom cure has been elusive. Distinguishing those who require intensive or novel therapeutic approaches from others who will be cured with minimal therapy is key for future trials. Incorporating new genomic biomarkers and more sensitive measures of minimal/measurable residual disease provide opportunities to achieve these goals., (© 2023 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
30. Insights into the genomics of iAMP21-ALL.
- Author
-
Rabin KR
- Subjects
- Humans, Genomics, Chromosomes, Human, Pair 21, Precursor Cell Lymphoblastic Leukemia-Lymphoma
- Published
- 2023
- Full Text
- View/download PDF
31. Flow cytometric assessment of leukemia-associated monocytes in childhood B-cell acute lymphoblastic leukemia outcome.
- Author
-
Contreras Yametti GP, Evensen NA, Schloss JM, Aldebert C, Duan E, Zhang Y, Hu J, Chambers TM, Scheurer ME, Teachey DT, Rabin KR, Raetz EA, Aifantis I, Carroll WL, and Witkowski MT
- Subjects
- Humans, Monocytes, Flow Cytometry, Precursor Cell Lymphoblastic Leukemia-Lymphoma, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Lymphoma, B-Cell
- Published
- 2023
- Full Text
- View/download PDF
32. Genomic landscape of Down syndrome-associated acute lymphoblastic leukemia.
- Author
-
Li Z, Chang TC, Junco JJ, Devidas M, Li Y, Yang W, Huang X, Hedges DJ, Cheng Z, Shago M, Carroll AJ, Heerema NA, Gastier-Foster J, Wood BL, Borowitz MJ, Sanclemente L, Raetz EA, Hunger SP, Feingold E, Rosser TC, Sherman SL, Loh ML, Mullighan CG, Yu J, Wu G, Lupo PJ, Rabin KR, and Yang JJ
- Subjects
- Animals, Mice, Mutation, Risk Factors, Genomics, Chromosome Aberrations, Down Syndrome complications, Down Syndrome genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Trisomy 21, the genetic cause of Down syndrome (DS), is the most common congenital chromosomal anomaly. It is associated with a 20-fold increased risk of acute lymphoblastic leukemia (ALL) during childhood and results in distinctive leukemia biology. To comprehensively define the genomic landscape of DS-ALL, we performed whole-genome sequencing and whole-transcriptome sequencing (RNA-Seq) on 295 cases. Our integrated genomic analyses identified 15 molecular subtypes of DS-ALL, with marked enrichment of CRLF2-r, IGH::IGF2BP1, and C/EBP altered (C/EBPalt) subtypes compared with 2257 non-DS-ALL cases. We observed abnormal activation of the CEBPD, CEBPA, and CEBPE genes in 10.5% of DS-ALL cases via a variety of genomic mechanisms, including chromosomal rearrangements and noncoding mutations leading to enhancer hijacking. A total of 42.3% of C/EBP-activated DS-ALL also have concomitant FLT3 point mutations or insertions/deletions, compared with 4.1% in other subtypes. CEBPD overexpression enhanced the differentiation of mouse hematopoietic progenitor cells into pro-B cells in vitro, particularly in a DS genetic background. Notably, recombination-activating gene-mediated somatic genomic abnormalities were common in DS-ALL, accounting for a median of 27.5% of structural alterations, compared with 7.7% in non-DS-ALL. Unsupervised hierarchical clustering analyses of CRLF2-rearranged DS-ALL identified substantial heterogeneity within this group, with the BCR::ABL1-like subset linked to an inferior event-free survival, even after adjusting for known clinical risk factors. These results provide important insights into the biology of DS-ALL and point to opportunities for targeted therapy and treatment individualization., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
33. Correction: Outstanding outcomes with two low intensity regimens in children with low-risk B-ALL: a report from COG AALL0932.
- Author
-
Schore RJ, Angiolillo AL, Kairalla JA, Devidas M, Rabin KR, Zweidler-McKay P, Borowitz MJ, Wood B, Carroll AJ, Heerema NA, Relling MV, Hitzler J, Kadan-Lottick NS, Maloney K, Wang C, Carroll WL, Winick NJ, Raetz EA, Loh ML, and Hunger SP
- Published
- 2023
- Full Text
- View/download PDF
34. Outstanding outcomes with two low intensity regimens in children with low-risk B-ALL: a report from COG AALL0932.
- Author
-
Schore RJ, Angiolillo AL, Kairalla JA, Devidas M, Rabin KR, Zweidler-McKay P, Borowitz MJ, Wood B, Carroll AJ, Heerema NA, Relling MV, Hitzler J, Kadan-Lottick NS, Maloney K, Wang C, Carroll WL, Winick NJ, Raetz EA, Loh ML, and Hunger SP
- Subjects
- Child, Humans, Risk, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Published
- 2023
- Full Text
- View/download PDF
35. SARS-CoV-2 infections in patients enrolled on the Children's Oncology Group standard-risk B-cell acute lymphoblastic leukemia trial, AALL1731.
- Author
-
Elgarten CW, Kairalla JA, Thompson JC, Miller TP, Wang C, Conway S, Loh ML, Raetz EA, Gupta S, Rau RE, Angiolillo A, Rabin KR, and Alexander S
- Abstract
Hematologic malignancy is a risk factor for severe coronavirus disease 2019 (COVID-19) in adults; however, data specific to children with leukemia are limited. High-quality infectious adverse event data from the ongoing Children's Oncology Group (COG) standard-risk B acute lymphoblastic leukemia/lymphoma (ALL/LLy) trial, AALL1731, were analyzed to provide a disease-specific estimate of SARS-CoV-2 infection outcomes in pediatric ALL. Of 253 patients with reported infections, the majority (77.1%) were asymptomatic or mildly symptomatic (CTCAE grade 1/2) and there was a single COVID-19-related death. These data suggest SARS-CoV-2 infection does not confer substantial morbidity among young patients with B-lymphoblastic leukemia/lymphoma (B-ALL/LLy)., Competing Interests: The authors declare they have no conflicts of interest., (© 2023 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
36. Patients with Down syndrome can be included in early phase clinical trials- a report from the T2016-003 Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) study.
- Author
-
Gossai N, Bhojwani D, Schafer ES, Chang BH, Pommert L, Verma A, Malvar J, Chi YY, Hitzler J, Burke MJ, and Rabin KR
- Subjects
- Humans, Down Syndrome complications, Down Syndrome drug therapy, Leukemia drug therapy, Leukemia pathology, Lymphoma drug therapy
- Published
- 2023
- Full Text
- View/download PDF
37. Ethnic-specific predictors of neurotoxicity among patients with pediatric acute lymphoblastic leukemia after high-dose methotrexate.
- Author
-
Harris RD, Bernhardt MB, Zobeck MC, Taylor OA, Gramatges MM, Schafer ES, Lupo PJ, Rabin KR, Scheurer ME, and Brown AL
- Subjects
- Child, Humans, Methotrexate, Antimetabolites, Antineoplastic therapeutic use, Creatinine, Retrospective Studies, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma epidemiology, Neurotoxicity Syndromes epidemiology, Neurotoxicity Syndromes etiology
- Abstract
Background: High-dose methotrexate (HD-MTX; 5000 mg/m
2 ) is an important component of curative therapy in many treatment regimens for high-risk pediatric acute lymphoblastic leukemia (ALL). However, methotrexate therapy can result in dose-limiting neurotoxicity, which may disproportionately affect Latino children. This study evaluated risk factors for neurotoxicity after HD-MTX in an ethnically diverse population of patients with ALL., Methods: The authors retrospectively reviewed the medical records of patients who were diagnosed with ALL and treated with HD-MTX at Texas Children's Cancer Center (2010-2017). Methotrexate neurotoxicity was defined as a neurologic episode (e.g., seizures or stroke-like symptoms) occurring within 21 days of HD-MTX that resulted in methotrexate treatment modifications. Mixed effects multivariable logistic regression was used to estimate the odds ratio (OR) and corresponding 95% confidence interval (CI) for the association between clinical factors and neurotoxicity., Results: Overall, 351 patients (58.1% Latino) who received 1183 HD-MTX infusions were evaluated. Thirty-five patients (10%) experienced neurotoxicity, 71% of whom were Latino. After adjusting for clinical risk factors, the authors observed that serum creatinine elevations ≥50% of baseline were associated with a three-fold increased odds (OR, 3.32; 95% CI, 0.98-11.21; p = .05) for neurotoxicity compared with creatinine elevation <25%. Notably, predictors of neurotoxicity differed by ethnicity. Specifically, Latino children experienced a nearly six-fold increase in neurotoxicity odds (OR, 5.80; 95% CI, 1.39-24.17; p = .02) with serum creatinine elevation ≥50% compared with creatinine elevation <25%., Conclusions: The current findings indicate that serum creatinine elevations ≥50% may be associated with an increased risk for neurotoxicity among Latino children with ALL and may identify potential candidates for therapeutic or supportive care interventions., (© 2023 American Cancer Society.)- Published
- 2023
- Full Text
- View/download PDF
38. Central nervous system status is prognostic in T-cell acute lymphoblastic leukemia: a Children's Oncology Group report.
- Author
-
Gossai NP, Devidas M, Chen Z, Wood BL, Zweidler-McKay PA, Rabin KR, Loh ML, Raetz EA, Winick NJ, Burke MJ, Carroll AJ, Esiashvili N, Heerema NA, Carroll WL, Hunger SP, Dunsmore KP, Winter SS, and Teachey DT
- Subjects
- Child, Humans, Infant, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Central Nervous System, Disease-Free Survival, Methotrexate, Prognosis, T-Lymphocytes, Treatment Outcome, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
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 clinical trials were examined. AALL0434 and AALL1231 tested efficacy of novel agents within augmented-Berlin-Frankfurt-Münster (aBFM) therapy. In addition to testing study-specific chemotherapy through randomization, the AALL0434 regimen delivered cranial radiation therapy (CRT) to most participants (90.8%), whereas AALL1231 intensified chemotherapy to eliminate CRT in 88.2% of participants. In an analysis of 2164 patients with T-ALL (AALL0434, 1550; AALL1231, 614), 1564 had 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 = .0004). Patients with CNS-1 and CNS-2 had similar 4-year overall survival (OS) (90.1% ± 0.8% and 90.5% ± 1.5%, respectively), with OS for CNS-3 being 82.7% ± 3.4% (P = .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 >2000 patients with T-ALL identified that CNS-1 and CNS-2 status at diagnosis had similar outcomes. Unlike B-ALL, CNS-2 status in T-ALL does not impact outcome with aBFM therapy, without additional intrathecal therapy, with or without CRT. Although nelarabine improved outcomes for those with CNS-3 status, novel approaches are needed. These trials were registered at www.clinicaltrials.gov as #NCT00408005 (AALL0434) and #NCT02112916 (AALL1231)., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
39. Novel and replicated clinical and genetic risk factors for toxicity from high-dose methotrexate in pediatric acute lymphoblastic leukemia.
- Author
-
Zobeck M, Bernhardt MB, Kamdar KY, Rabin KR, Lupo PJ, and Scheurer ME
- Subjects
- Humans, Child, Retrospective Studies, Bayes Theorem, Creatinine, Risk Factors, Liver-Specific Organic Anion Transporter 1, Methotrexate adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Study Objective: Methotrexate (MTX) is a key component of treatment for high-risk pediatric acute lymphoblastic leukemia (ALL) but may cause acute kidney injury and prolonged hospitalization due to delayed clearance. The purpose of this study is to identify clinical and genetic factors that may predict which children are at risk for creatinine increase and prolonged MTX clearance., Design: We conducted a single-center, retrospective cohort study of pediatric patients with ALL who received 4000-5000 mg/m
2 of MTX. Measurements We performed germline genotyping to determine genetic ancestry and allele status for 49 single nucleotide polymorphisms (SNPs) identified from the literature as related to MTX disposition. Bayesian hierarchical ordinal regression models for creatinine increase and for prolonged MTX clearance were developed., Main Results: Hispanic ethnicity, body mass index (BMI) < 3%, BMI between 85%-95%, and Native American genetic ancestry were found to be associated with an increased risk for creatinine elevation. Older age, Black race, and use of the intensive monitoring protocol were associated with a decreased risk for creatinine elevation. Older age, B- compared to T-ALL, and the minor alleles of rs2838958/SLC19A1 and rs7317112/ABCC4 were associated with an increased risk for delayed clearance. Black race, MTX dose reduction, and the minor allele of rs2306283/SLCO1B1 were found to be associated with a decreased risk for delayed clearance., Conclusions: These predictors of MTX toxicities may allow for more precise individualized toxicity risk prediction., (© 2023 Pharmacotherapy Publications, Inc.)- Published
- 2023
- Full Text
- View/download PDF
40. Prognostic impact of pretreatment immunoglobulin clonal composition in pediatric B-lymphoblastic leukemia.
- Author
-
Fries C, Lee LW, Devidas M, Dai Y, Rabin KR, Gupta S, Loh ML, Kirsch IR, Wood B, and Rau RE
- Subjects
- Child, Humans, Prognosis, Genes, Immunoglobulin, Immunoglobulins, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2023
- Full Text
- View/download PDF
41. Antileukemic properties of the kinase inhibitor OTSSP167 in T-cell acute lymphoblastic leukemia.
- Author
-
Bridges CS, Chen TJ, Puppi M, Rabin KR, and Lacorazza HD
- Subjects
- Humans, Mice, Animals, Child, Naphthyridines pharmacology, Naphthyridines therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, T-Lymphocytes metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Novel drugs are needed to increase treatment response in children with high-risk T-cell acute lymphoblastic leukemia (T-ALL). Following up on our previous report on the activation of the MAP2K7-JNK pathway in pediatric T-ALL, here we demonstrate that OTSSP167, recently shown to inhibit MAP2K7, has antileukemic capacity in T-ALL. OTSSP167 exhibited dose-dependent cytotoxicity against a panel of T-ALL cell lines with IC50 in the nanomolar range (10-50 nM). OTSSP167 induces apoptosis and cell cycle arrest in T-ALL cell lines, associated at least partially with the inhibition of MAP2K7 kinase activity and lower activation of its downstream substrate, JNK. Other leukemic T-cell survival pathways, such as mTOR and NOTCH1 were also inhibited. Daily intraperitoneal administration of 10 mg/kg OTSSP167 was well tolerated, with mice showing no hematological toxicity, and effective at reducing the expansion of human T-ALL cells in a cell-based xenograft model. The same dosage of OTSSP167 efficiently controlled the leukemia burden in the blood, bone marrow, and spleen of 3 patient-derived xenografts, which resulted in prolonged survival. OTSSP167 exhibited synergistic interactions when combined with dexamethasone, L-asparaginase, vincristine, and etoposide. Our findings reveal novel antileukemic properties of OTSSP167 in T-ALL and support the use of OTSSP167 as an adjuvant drug to increase treatment response and reduce relapses in pediatric T-ALL., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
42. Racial and ethnic disparities in childhood and young adult acute lymphocytic leukaemia: secondary analyses of eight Children's Oncology Group cohort trials.
- Author
-
Gupta S, Dai Y, Chen Z, Winestone LE, Teachey DT, Bona K, Aplenc R, Rabin KR, Zweidler-McKay P, Carroll AJ, Heerema NA, Gastier-Foster J, Borowitz MJ, Wood BL, Maloney KW, Mattano LA Jr, Larsen EC, Angiolillo AL, Burke MJ, Salzer WL, Winter SS, Brown PA, Guest EM, Dunsmore KP, Kairalla JA, Winick NJ, Carroll WL, Raetz EA, Hunger SP, Loh ML, and Devidas M
- Subjects
- Adolescent, Humans, Child, Male, Female, Young Adult, White People, Black or African American, Ethnicity, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Leukemia, Lymphocytic, Chronic, B-Cell
- Abstract
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: 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: 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: National Cancer Institute and St Baldrick's Foundation., Competing Interests: Declaration of interests We declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
43. 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.
- Author
-
Elgarten CW, Thompson JC, Angiolillo A, Chen Z, Conway S, Devidas M, Gupta S, Kairalla JA, McNeer JL, O'Brien MM, Rabin KR, Rau RE, Rheingold SR, Wang C, Wood C, Raetz EA, Loh ML, Alexander S, and Miller TP
- Subjects
- Acute Disease, Adolescent, Child, Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma etiology
- Abstract
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., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
44. Residence in a Latinx enclave and end-induction minimal residual disease positivity among children with acute lymphoblastic leukemia.
- Author
-
Muñiz JP, Woodhouse JP, Hughes AE, Pruitt SL, Rabin KR, Scheurer ME, Lupo PJ, and Schraw JM
- Subjects
- Child, Humans, Incidence, Neoplasm, Residual, Texas, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma epidemiology
- Abstract
Racial and ethnic inequities in survival persist for children with acute lymphoblastic leukemia (ALL). In the US, there are strong associations between SES, race/ethnicity, and place of residence. This is evidenced by ethnic enclaves: neighborhoods with high concentrations of ethnic residents, immigrants, and language isolation. The Latinx enclave index (LEI) can be used to investigate how residence in a Latinx enclave is associated with health outcomes. We studied the association between LEI score and minimal residual disease (MRD) in 142 pediatric ALL patients treated at Texas Children's Hospital. LEI score was associated with end-induction MRD positivity (OR per unit increase 1.63, CI 1.12-2.46). There was also a significant trend toward increased odds of MRD positivity among children living in areas with the highest enclave index scores. MRD positivity at end of induction is associated with higher incidence of relapse and lower overall survival among children with ALL; future studies are needed to elucidate the exact causes of these findings and to improve ALL outcomes among children residing within Latinx enclaves.Supplemental data for this article is available online at https://doi.org/10.1080/08880018.2022.2047850.
- Published
- 2022
- Full Text
- View/download PDF
45. The genomic landscape of pediatric acute lymphoblastic leukemia.
- Author
-
Brady SW, Roberts KG, Gu Z, Shi L, Pounds S, Pei D, Cheng C, Dai Y, Devidas M, Qu C, Hill AN, Payne-Turner D, Ma X, Iacobucci I, Baviskar P, Wei L, Arunachalam S, Hagiwara K, Liu Y, Flasch DA, Liu Y, Parker M, Chen X, Elsayed AH, Pathak O, Li Y, Fan Y, Michael JR, Rusch M, Wilkinson MR, Foy S, Hedges DJ, Newman S, Zhou X, Wang J, Reilly C, Sioson E, Rice SV, Pastor Loyola V, Wu G, Rampersaud E, Reshmi SC, Gastier-Foster J, Guidry Auvil JM, Gesuwan P, Smith MA, Winick N, Carroll AJ, Heerema NA, Harvey RC, Willman CL, Larsen E, Raetz EA, Borowitz MJ, Wood BL, Carroll WL, Zweidler-McKay PA, Rabin KR, Mattano LA, Maloney KW, Winter SS, Burke MJ, Salzer W, Dunsmore KP, Angiolillo AL, Crews KR, Downing JR, Jeha S, Pui CH, Evans WE, Yang JJ, Relling MV, Gerhard DS, Loh ML, Hunger SP, Zhang J, and Mullighan CG
- Subjects
- Child, Chromosome Aberrations, Exome genetics, Genomics, Humans, Mutation, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
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., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
46. Rates of laboratory adverse events by course in paediatric leukaemia ascertained with automated electronic health record extraction: a retrospective cohort study from the Children's Oncology Group.
- Author
-
Miller TP, Getz KD, Li Y, Demissei BG, Adamson PC, Alonzo TA, Burrows E, Cao L, Castellino SM, Daves MH, Fisher BT, Gerbing R, Grundmeier RW, Krause EM, Lee J, Lupo PJ, Rabin KR, Ramos M, Scheurer ME, Wilkes JJ, Winestone LE, Hawkins DS, Gramatges MM, and Aplenc R
- Subjects
- Adolescent, Child, Child, Preschool, Clinical Trials as Topic, Electronic Health Records, Female, Humans, Hypokalemia epidemiology, Infant, Infant, Newborn, Male, Retrospective Studies, Young Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Background: Adverse events are often misreported in clinical trials, leading to an incomplete understanding of toxicities. We aimed to test automated laboratory adverse event ascertainment and grading (via the ExtractEHR automated package) to assess its scalability and define adverse event rates for children with acute myeloid leukaemia and acute lymphoblastic leukaemia., Methods: For this retrospective cohort study from the Children's Oncology Group (COG), we included patients aged 0-22 years treated for acute myeloid leukaemia or acute lymphoblastic leukaemia at Children's Healthcare of Atlanta (Atlanta, GA, USA) from Jan 1, 2010, to Nov 1, 2018, at the Children's Hospital of Philadelphia (Philadelphia, PA, USA) from Jan 1, 2011, to Dec 31, 2014, and at the Texas Children's Hospital (Houston, TX, USA) from Jan 1, 2011, to Dec 31, 2014. The ExtractEHR automated package acquired, cleaned, and graded laboratory data as per Common Terminology Criteria for Adverse Events (CTCAE) version 5 for 22 commonly evaluated grade 3-4 adverse events (fatal events were not evaluated) with numerically based CTCAE definitions. Descriptive statistics tabulated adverse event frequencies. Adverse events ascertained by ExtractEHR were compared to manually reported adverse events for patients enrolled in two COG trials (AAML1031, NCT01371981; AALL0932, NCT02883049). Analyses were restricted to protocol-defined chemotherapy courses (induction I, induction II, intensification I, intensification II, and intensification III for acute myeloid leukaemia; induction, consolidation, interim maintenance, delayed intensification, and maintenance for acute lymphoblastic leukaemia)., Findings: Laboratory adverse event data from 1077 patients (583 from Children's Healthcare of Atlanta, 200 from the Children's Hospital of Philadelphia, and 294 from the Texas Children's Hospital) who underwent 4611 courses (549 for acute myeloid leukaemia and 4062 for acute lymphoblastic leukaemia) were extracted, processed, and graded. Of the 166 patients with acute myeloid leukaemia, 86 (52%) were female, 80 (48%) were male, 96 (58%) were White, and 132 (80%) were non-Hispanic. Of the 911 patients with acute lymphoblastic leukaemia, 406 (45%) were female, 505 (55%) were male, 596 (65%) were White, and 641 (70%) were non-Hispanic. Patients with acute myeloid leukaemia had the most adverse events during induction I and intensification II. Hypokalaemia (one [17%] of six to 75 [48%] of 156 courses) and alanine aminotransferase (ALT) increased (13 [10%] of 134 to 27 [17%] of 156 courses) were the most prevalent non-haematological adverse events in patients with acute myeloid leukaemia, as identified by ExtractEHR. Patients with acute lymphoblastic leukaemia had the greatest number of adverse events during induction and maintenance (eight adverse events with prevalence ≥10%; induction and maintenance: anaemia, platelet count decreased, white blood cell count decreased, neutrophil count decreased, lymphocyte count decreased, ALT increased, and hypocalcaemia; induction: hypokalaemia; maintenance: aspartate aminotransferase [AST] increased and blood bilirubin increased), as identified by ExtractEHR. 187 (85%) of 220 total comparisons in 22 adverse events in four AAML1031 and six AALL0923 courses were substantially higher with ExtractEHR than COG-reported adverse event rates for adverse events with a prevalence of at least 2%., Interpretation: ExtractEHR is scalable and accurately defines laboratory adverse event rates for paediatric acute leukaemia; moreover, ExtractEHR seems to detect higher rates of laboratory adverse events than those reported in COG trials. These rates can be used for comparisons between therapies and to counsel patients treated on or off trials about the risks of chemotherapy. ExtractEHR-based adverse event ascertainment can improve reporting of laboratory adverse events in clinical trials., Funding: US National Institutes of Health, St Baldrick's Foundation, and Alex's Lemonade Stand Foundation., Competing Interests: Declaration of interests SMC and PCA disclose relationships with the Children's Oncology Group as group and study chairs. MHD received funding from Alex's Lemonade Stand Foundation for this study and funding from Epic Systems. BTF and DSH have received funding from Pfizer and Merck. BTF has also received funding from Astellas. MMG, TPM, and PCA have received research funding from the National Institutes of Health and National Cancer Institute. KDG has received research funding from the US National Institutes of Health (NIH) and the National Heart, Lung, and Blood Institute. MMG's institution has a grant to receive funding from the Cancer Prevention Research Institute of Texas, Hyundai Hope on Wheels, the American Society of Clinical Oncology, Children's Cancer Cause, and the American Society of Clinical Investigation. DSH has received funding from Incyte, Eli Lilly, E R Squibb & Sons, Jazz Pharmaceuticals, Bayer, and AstraZeneca. PCA is currently employed by Sanofi. RWG has leadership roles on the board of the American Academy of Pediatrics and the American Board of Pediatrics., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
47. Epigenome-wide association study of acute lymphoblastic leukemia in children with Down syndrome.
- Author
-
Li S, Sok P, Xu K, Muskens IS, Elliott N, Myint SS, Pandey P, Hansen HM, Morimoto LM, Kang AY, Metayer C, Ma X, Mueller BA, Roy A, Roberts I, Rabin KR, Brown AL, Lupo PJ, Wiemels JL, and de Smith AJ
- Subjects
- Acute Disease, Child, Epigenome, Humans, Down Syndrome complications, Down Syndrome genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2022
- Full Text
- View/download PDF
48. An updated assessment of 43,110 patients enrolled in the Childhood Cancer Research Network: A Children's Oncology Group report.
- Author
-
Brown AL, Sok P, Scheurer ME, Rabin KR, Marcotte EL, Hawkins DS, Spector LG, and Lupo PJ
- Subjects
- Censuses, Child, Forecasting, Humans, Incidence, Male, Registries, Neoplasms diagnosis, Neoplasms epidemiology, Neoplasms therapy
- Abstract
Background: The Childhood Cancer Research Network (CCRN) was established by the Children's Oncology Group (COG) as a resource for epidemiologic studies of childhood cancer. The objective of this study was to evaluate the representativeness of CCRN and identify factors associated with enrollment., Method: The number of US childhood patients with cancer diagnosed <20 years of age enrolled in CCRN (2008-2015) was compared to expected counts, calculated from Surveillance, Epidemiology, and End Results incidence rates and US Census population estimates. Observed-to-expected ratios and corresponding 95% confidence intervals (CI) were estimated across sex, race, diagnosis age, calendar year, and cancer diagnosis groups. Multivariable linear regression models were generated to evaluate the association between open COG phase 3 therapeutic trials and CCRN enrollment rates., Result: The 43,110 cases enrolled in CCRN represented 36% of the expected childhood cancers diagnosed from 2008 to 2015 (N = 120,118). CCRN enrollment ratios [95% CI] were highest among males (0.38 [95% CI, 0.37-0.38]), non-Hispanics (0.35 [95% CI, 0.35-0.36]), and those diagnosed from 1 to 4 years of age (0.50 [95% CI, 0.50-51]). Enrollment ratios varied by diagnosis group, with leukemia, myeloproliferative diseases, myelodysplastic diseases (0.55 [95% CI, 0.54-0.55]), and renal tumors (0.55 [95% CI, 0.53-0.58]) having the highest enrollment. After adjusting for year of diagnosis and cancer diagnosis, there was a 3.1% [95% CI, 0.6-5.6%] increase in CCRN enrollment during windows of open COG therapeutic trials., Conclusions: Despite enrolling only 36% of newly diagnosed cases, CCRN remains a valuable resource for investigators conducting childhood cancer etiology and survivorship research. The results of this study may inform efforts to improve enrollment on current and future COG nontherapeutic registry protocols., (© 2022 American Cancer Society.)
- Published
- 2022
- Full Text
- View/download PDF
49. CRLF2 overexpression results in reduced B-cell differentiation and upregulated E2F signaling in the Dp16 mouse model of Down syndrome.
- Author
-
Junco JJ, Zorman B, Gant VU Jr, Muñoz J, Lacorazza HD, Sumazin P, and Rabin KR
- Subjects
- Animals, Cell Differentiation genetics, Disease Models, Animal, Humans, Mice, Receptors, Cytokine genetics, Signal Transduction, Down Syndrome complications, Down Syndrome genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Children with Down syndrome (DS) are 10-fold more likely to develop B-cell acute lymphoblastic leukemia (B-ALL), with a higher frequency of rearrangements resulting in overexpression of cytokine receptor-like factor 2 (CRLF2). Here, we investigated the impact of CRLF2 overexpression on B-cell progenitor proliferation, immunophenotype, and gene expression profile in the Dp(16)1Yey (Dp16) mouse model of DS compared with wild-type (WT) mice. CRLF2 overexpression enhanced immature B-lymphoid colony development and increased the proportion of less differentiated pre-pro-B cells, with a greater effect in Dp16 versus WT. In CRLF2-rearranged (CRLF2-R) B-ALL patient samples, cells with higher CRLF2 expression exhibited a less differentiated B-cell immunophenotype. CRLF2 overexpression resulted in a gene expression signature associated with E2F signaling both in Dp16 B-progenitors and in DS-ALL patient samples, and PI3K/mTOR and pan-CDK inhibitors, which reduce E2F-mediated signaling, exhibited cytotoxicity in CRLF2-R B-ALL cell lines and patient samples. CRLF2 overexpression alone in Dp16 stem and progenitor cells did not result in leukemic transformation in recipient mice. Thus, CRLF2 overexpression results in reduced B-cell differentiation and enhanced E2F signaling in Dp16 B-progenitor cells and DS-ALL patient samples. These findings suggest a functional basis for the high frequency of CRLF2-R in DS-ALL as well as a potential therapeutically targetable pathway., (Copyright © 2022 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
50. Sex-based disparities in outcome in pediatric acute lymphoblastic leukemia: a Children's Oncology Group report.
- Author
-
Gupta S, Teachey DT, Chen Z, Rabin KR, Dunsmore KP, Larsen EC, Maloney KW, Mattano LA Jr, Winter SS, Carroll AJ, Heerema NA, Borowitz MJ, Wood BL, Carroll WL, Raetz EA, Winick NJ, Loh ML, Hunger SP, and Devidas M
- Subjects
- Adolescent, Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Marrow, Child, Child, Preschool, Disease-Free Survival, Female, Humans, Infant, Male, Recurrence, Treatment Outcome, Young Adult, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma
- Abstract
Background: Boys with acute lymphoblastic leukemia (ALL) have historically experienced inferior survival compared to girls. This study determined whether sex-based disparities persist with contemporary therapy and whether patterns of treatment failure vary by sex., Methods: Patients 1 to 30.99 years old were enrolled on frontline Children's Oncology Group trials between 2004 and 2014. Boys received an additional year of maintenance therapy. Sex-based differences in the distribution of various prognosticators, event-free survival (EFS) and overall survival (OS), and subcategories of relapse by site were explored., Results: A total of 8202 (54.4% male) B-cell ALL (B-ALL) and 1562 (74.3% male) T-cell ALL (T-ALL) patients were included. There was no sex-based difference in central nervous system (CNS) status. Boys experienced inferior 5-year EFS and OS (EFS, 84.6% ± 0.5% vs 86.0% ± 0.6%, P = .009; OS, 91.3% ± 0.4% vs 92.5% ± 0.4%, P = .02). This was attributable to boys with B-ALL, who experienced inferior EFS (hazard ratio [HR], 1.2; 95% confidence interval [95% CI], 1.1-1.3; P = .004) and OS (HR, 1.2; 95% CI, 1.0-1.4; P = .046) after adjustment for prognosticators. Inferior B-ALL outcomes in boys were attributable to more relapses (5-year cumulative incidence 11.2% ± 0.5% vs 9.6% ± 0.5%; P = .001), particularly involving the CNS (4.2% ± 0.3% vs 2.5% ± 0.3%; P < .0001). There was no difference in isolated bone marrow relapses (5.4% ± 0.4% vs 6.2% ± 0.4%; P = .49). There were no sex-based differences in EFS or OS in T-ALL., Conclusions: Sex-based disparities in ALL persist, attributable to increased CNS relapses in boys with B-ALL. Studies of potential mechanisms are warranted. Improved strategies to identify and modify treatment for patients at highest risk of CNS relapse may have particular benefit for boys., (© 2022 American Cancer Society.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.