77 results on '"Orgel E"'
Search Results
2. 20. A growing risk in a growing population: obesity's impact on leukemia incidence and survival
- Author
-
Orgel, E., primary
- Published
- 2016
- Full Text
- View/download PDF
3. Recommendations for ototoxicity surveillance for childhood, adolescent, and young adult cancer survivors: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group in collaboration with the PanCare Consortium
- Author
-
Clemens, Eva, Van den Heuvel - Eibrink, Marry, Mulder, RL, Kremer, LCM (Leontien), Hudson, MM, Skinner, R, Constine, LS, Bass, JK, Kuehni, CE, Langer, T, van Dalen, EC, Bardi, E, Bonne, NX, Brock, PR, Brooks, B, Carleton, B, Caron, E, Chang, KW, Johnston, K, Knight, K, Nathan, PC, Orgel, E, Prasad, PK, Rottenberg, J, Scheinemann, K, de Vries, A.C.H., Walwyn, T, Weiss, A, Zehnhoff-Dinnesen, AA, Cohn, RJ, Landier, W, Kadan-Lottick, N, Levitt, G, Hoetink, A, Mussman, J, Princess Máxima Center for Pediatric Oncology [Utrecht, Netherlands], Erasmus MC-Sophia Hospital [Rotterdam, Netherlands], Emma Children's Hospital [Amsterdam, Netherlands] (Academic Medical Center), University of Amsterdam [Amsterdam] (UvA), St Jude Children's Research Hospital, Great North Children’s Hospital [Newcastle Upon Tyne, UK], Northern Institute for Cancer Research [Newcastle] (NICR), Newcastle University [Newcastle], University of Rochester Medical Center (URMC), Institute of Social and Preventive Medicine [Bern] (ISPM), Universität Bern [Bern] (UNIBE), University Hospital for Children and Adolescents [Lübeck, Germany], Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM) - U 1192 (PRISM), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Service d'Otologie et d'Otoneurologie [CHU Lille], Centre Hospitalier Universitaire de Lille (CHU de Lille), Great Ormond Street Hospital for Children NHS Foundation Trust [London, UK] (GOSHC), University of British Columbia (UBC), Stanford University, Sydney Children's hospital, Oregon Health and Science University [Portland] (OHSU), The Hospital for sick children [Toronto] (SickKids), Children’s Hospital Los Angeles [Los Angeles], University of Southern California (USC), Louisiana State University (LSU), Children's Hospital at New Orleans, Masaryk University [Brno] (MUNI), Kantonsspital Aarau [Aarau, Switzerland], University Hospital Basel [Basel], McMaster University [Hamilton, Ontario], Perth Children's Hospital [Nedlands, WA, Australia], The University of Western Australia (UWA), University of Regensburg, University Hospital Münster - Universitaetsklinikum Muenster [Germany] (UKM), University of New South Wales [Sydney] (UNSW), University of Alabama at Birmingham [ Birmingham] (UAB), SALZET, Michel, Pediatrics, Universität Bern [Bern], Université de Lille-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), and Great Ormond Street Hospital for Children NHS Foundation Trust [London, UK]
- Subjects
Shunt placement ,Pediatrics ,medicine.medical_specialty ,Adolescent ,[SDV]Life Sciences [q-bio] ,Childhood cancer ,Harmonization ,Antineoplastic Agents ,Platinum Compounds ,Article ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Ototoxicity ,SDG 3 - Good Health and Well-being ,Cancer Survivors ,Neoplasms ,otorhinolaryngologic diseases ,Medicine ,Humans ,Young adult ,030223 otorhinolaryngology ,Child ,Evidence-Based Medicine ,business.industry ,Cancer ,Guideline ,medicine.disease ,3. Good health ,[SDV] Life Sciences [q-bio] ,Oncology ,030220 oncology & carcinogenesis ,Population Surveillance ,Cranial Irradiation ,business ,Delivery of Health Care - Abstract
International audience; Childhood, adolescent, and young adult (CAYA) cancer survivors treated with platinum-based drugs, head or brain radiotherapy, or both have an increased risk of ototoxicity (hearing loss, tinnitus, or both). To ensure optimal care and reduce consequent problems—such as speech and language, social–emotional development, and learning difficulties—for these CAYA cancer survivors, clinical practice guidelines for monitoring ototoxicity are essential. The implementation of surveillance across clinical settings is hindered by differences in definitions of hearing loss, recommendations for surveillance modalities, and remediation. To address these deficiencies, the International Guideline Harmonization Group organised an international multidisciplinary panel, including 32 experts from ten countries, to evaluate the quality of evidence for ototoxicity following platinum-based chemotherapy and head or brain radiotherapy, and formulate and harmonise ototoxicity surveillance recommendations for CAYA cancer survivors.
- Published
- 2019
4. Reduction in Ototoxicity Using IMRT for Patients with Medulloblastoma/PNET
- Author
-
Ellis, L.R., primary, Olch, A.J., additional, Orgel, E., additional, and Wong, K., additional
- Published
- 2019
- Full Text
- View/download PDF
5. Not All Aspiration Is Dysphagia in Trisomy 21: Working with the Family When the Solution Is Difficult to Swallow
- Author
-
Miyakawa, R., primary, Mostmand, S., additional, Orgel, E., additional, Zheng, Y., additional, and Kato, R.M., additional
- Published
- 2019
- Full Text
- View/download PDF
6. The genetic basis and cell of origin of mixed phenotype acute leukaemia.
- Author
-
Alexander, TB, Gu, Z, Iacobucci, I, Dickerson, K, Choi, JK, Xu, B, Payne-Turner, D, Yoshihara, H, Loh, ML, Horan, J, Buldini, B, Basso, G, Elitzur, S, de Haas, V, Zwaan, CM, Yeoh, A, Reinhardt, D, Tomizawa, D, Kiyokawa, N, Lammens, T, De Moerloose, B, Catchpoole, D, Hori, H, Moorman, A, Moore, AS, Hrusak, O, Meshinchi, S, Orgel, E, Devidas, M, Borowitz, M, Wood, B, Heerema, NA, Carrol, A, Yang, Y-L, Smith, MA, Davidsen, TM, Hermida, LC, Gesuwan, P, Marra, MA, Ma, Y, Mungall, AJ, Moore, RA, Jones, SJM, Valentine, M, Janke, LJ, Rubnitz, JE, Pui, C-H, Ding, L, Liu, Y, Zhang, J, Nichols, KE, Downing, JR, Cao, X, Shi, L, Pounds, S, Newman, S, Pei, D, Guidry Auvil, JM, Gerhard, DS, Hunger, SP, Inaba, H, Mullighan, CG, Alexander, TB, Gu, Z, Iacobucci, I, Dickerson, K, Choi, JK, Xu, B, Payne-Turner, D, Yoshihara, H, Loh, ML, Horan, J, Buldini, B, Basso, G, Elitzur, S, de Haas, V, Zwaan, CM, Yeoh, A, Reinhardt, D, Tomizawa, D, Kiyokawa, N, Lammens, T, De Moerloose, B, Catchpoole, D, Hori, H, Moorman, A, Moore, AS, Hrusak, O, Meshinchi, S, Orgel, E, Devidas, M, Borowitz, M, Wood, B, Heerema, NA, Carrol, A, Yang, Y-L, Smith, MA, Davidsen, TM, Hermida, LC, Gesuwan, P, Marra, MA, Ma, Y, Mungall, AJ, Moore, RA, Jones, SJM, Valentine, M, Janke, LJ, Rubnitz, JE, Pui, C-H, Ding, L, Liu, Y, Zhang, J, Nichols, KE, Downing, JR, Cao, X, Shi, L, Pounds, S, Newman, S, Pei, D, Guidry Auvil, JM, Gerhard, DS, Hunger, SP, Inaba, H, and Mullighan, CG
- Abstract
Mixed phenotype acute leukaemia (MPAL) is a high-risk subtype of leukaemia with myeloid and lymphoid features, limited genetic characterization, and a lack of consensus regarding appropriate therapy. Here we show that the two principal subtypes of MPAL, T/myeloid (T/M) and B/myeloid (B/M), are genetically distinct. Rearrangement of ZNF384 is common in B/M MPAL, and biallelic WT1 alterations are common in T/M MPAL, which shares genomic features with early T-cell precursor acute lymphoblastic leukaemia. We show that the intratumoral immunophenotypic heterogeneity characteristic of MPAL is independent of somatic genetic variation, that founding lesions arise in primitive haematopoietic progenitors, and that individual phenotypic subpopulations can reconstitute the immunophenotypic diversity in vivo. These findings indicate that the cell of origin and founding lesions, rather than an accumulation of distinct genomic alterations, prime tumour cells for lineage promiscuity. Moreover, these findings position MPAL in the spectrum of immature leukaemias and provide a genetically informed framework for future clinical trials of potential treatments for MPAL.
- Published
- 2018
7. TACL'ing supportive care needs in pediatric early phase clinical trials for acute leukemia: A report from the therapeutic advances in childhood leukemia & lymphoma (TACL) consortium supportive care committee
- Author
-
Orgel, E, primary and Auletta, J J, additional
- Published
- 2017
- Full Text
- View/download PDF
8. Prospective External Validation of the Esbenshade Vanderbilt Models Accurately Predicts Bloodstream Infection Risk in Febrile Non-Neutropenic Children With Cancer.
- Author
-
Zhao Z, Patel PA, Slatnick L, Sitthi-Amorn A, Bielamowicz KJ, Nunez FA, Walsh AM, Hess J, Rossoff J, Elgarten C, Myers R, Saab R, Basbous M, Mccormick M, Aftandilian C, Richards R, Nessle CN, Tribble AC, Sheth Bhutada JK, Coven SL, Runco D, Wilkes J, Gurunathan A, Guinipero T, Belsky JA, Lee K, Wong V, Malhotra M, Armstrong A, Jerkins LP, Cross SJ, Fisher L, Stein MT, Wu NL, Yi T, Orgel E, Haeusler GM, Wolf J, Demedis JM, Miller TP, and Esbenshade AJ
- Subjects
- Humans, Child, Prospective Studies, Fever diagnosis, Fever etiology, Anti-Bacterial Agents therapeutic use, Bacteremia diagnosis, Bacteremia epidemiology, Bacteremia microbiology, Infections, Neoplasms complications, Bacterial Infections, Sepsis diagnosis
- Abstract
Purpose: The optimal management of fever without severe neutropenia (absolute neutrophil count [ANC] ≥500/µL) in pediatric patients with cancer is undefined. The previously proposed Esbenshade Vanderbilt (EsVan) models accurately predict bacterial bloodstream infections (BSIs) in this population and provide risk stratification to aid management, but have lacked prospective external validation., Materials and Methods: Episodes of fever with a central venous catheter and ANC ≥500/µL occurring in pediatric patients with cancer were prospectively collected from 18 academic medical centers. Variables included in the EsVan models and 7-day clinical outcomes were collected. Five versions of the EsVan models were applied to the data with calculation of C-statistics for both overall BSI rate and high-risk organism BSI (gram-negative and Staphylococcus aureus BSI), as well as model calibration., Results: In 2,565 evaluable episodes, the BSI rate was 4.7% (N = 120). Complications for the whole cohort were rare, with 1.1% (N = 27) needing intensive care unit (ICU) care by 7 days, and the all-cause mortality rate was 0.2% (N = 5), with only one potential infection-related death. C-statistics ranged from 0.775 to 0.789 for predicting overall BSI, with improved accuracy in predicting high-risk organism BSI (C-statistic 0.800-0.819). Initial empiric antibiotics were withheld in 14.9% of episodes, with no deaths or ICU admissions attributable to not receiving empiric antibiotics., Conclusion: The EsVan models, especially EsVan2b, perform very well prospectively across multiple academic medical centers and accurately stratify risk of BSI in episodes of non-neutropenic fever in pediatric patients with cancer. Implementation of routine screening with risk-stratified management for non-neutropenic fever in pediatric patients with cancer could safely reduce unnecessary antibiotic use.
- Published
- 2024
- Full Text
- View/download PDF
9. An Exome Capture-Based RNA-Sequencing Assay for Genome-Wide Identification and Prioritization of Clinically Important Fusions in Pediatric Tumors.
- Author
-
Buckley J, Schmidt RJ, Ostrow D, Maglinte D, Bootwalla M, Ruble D, Govindarajan A, Ji J, Kovach AE, Orgel E, Raca G, Navid F, Mascarenhas L, Pawel B, Robison N, Gai X, and Biegel JA
- Subjects
- Child, Humans, High-Throughput Nucleotide Sequencing methods, Software, RNA, Gene Fusion, Exome genetics, Neoplasms diagnosis, Neoplasms genetics
- Abstract
This study reports the development of an exome capture-based RNA-sequencing assay to detect recurring and novel fusions in hematologic, solid, and central nervous system tumors. The assay used Twist Comprehensive Exome capture with either fresh or formalin-fixed samples and a bioinformatic platform that provides fusion detection, prioritization, and downstream curation. A minimum of 50 million uniquely mapped reads, a consensus read alignment/fusion calling approach using four callers (Arriba, FusionCatcher, STAR-Fusion, and Dragen), and custom software were used to integrate, annotate, and rank the candidate fusion calls. In an evaluation of 50 samples, the number of calls varied substantially by caller, from a mean of 24.8 with STAR-Fusion to 259.6 with FusionCatcher; only 1.1% of calls were made by all four callers. Therefore a filtering and ranking algorithm was developed based on multiple criteria, including number of supporting reads, calling consensus, genes involved, and cross-reference against databases of known cancer-associated or likely false-positive fusions. This approach was highly effective in pinpointing known clinically relevant fusions, ranking them first in 47 of 50 samples (94%). Detection of pathogenic gene fusions in three diagnostically challenging cases highlights the importance of a genome-wide and nontargeted method for fusion detection in pediatric cancer., Competing Interests: Disclosure Statement None declared., (Copyright © 2024 Association for Molecular Pathology and American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Apixaban versus no anticoagulation for the prevention of venous thromboembolism in children with newly diagnosed acute lymphoblastic leukaemia or lymphoma (PREVAPIX-ALL): a phase 3, open-label, randomised, controlled trial.
- Author
-
O'Brien SH, Rodriguez V, Lew G, Newburger JW, Schultz CL, Orgel E, Derr K, Ranalli MA, Esbenshade AJ, Hochberg J, Kang HJ, Dinikina Y, Mills D, Donovan M, Dyme JL, Favatella NA, and Mitchell LG
- Subjects
- Humans, Male, Female, Child, Anticoagulants adverse effects, Epistaxis chemically induced, Epistaxis complications, Epistaxis drug therapy, Treatment Outcome, Venous Thromboembolism etiology, Venous Thromboembolism prevention & control, Venous Thromboembolism drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Thrombosis drug therapy, Lymphoma drug therapy, Heart Arrest chemically induced, Heart Arrest complications, Heart Arrest drug therapy
- Abstract
Background: Paediatric patients with acute lymphoblastic leukaemia or lymphoma are at increased risk of venous thromboembolism resulting in increased mortality and morbidity. We hypothesised that apixaban, a direct oral anticoagulant, would safely reduce venous thromboembolism in this patient population., Methods: PREVAPIX-ALL was a phase 3, open-label, randomised, controlled trial conducted in 74 paediatric hospitals in 9 countries. Participants aged 1 year or older to younger than 18 years with newly diagnosed acute lymphoblastic leukaemia (pre-B cell or T cell) or lymphoblastic lymphoma (B cell or T cell immunophenotype) and a central venous line in place throughout induction were randomly assigned 1:1 to standard of care (SOC, ie, no systemic anticoagulation) or weight-adjusted twice-daily apixaban during induction. Randomisation was performed centrally and stratified by age (those <10 years or those ≥10 years). Participants weighing 35 kg or less were administered 2·5 mg twice daily of apixaban as a 2·5 mg tablet, 0·5 mg tablets, or 0·4 mg/mL oral solution, while those weighing more than 35 kg were administered weight-adjusted prophylactic doses using 0·5 mg tablets or the 0·4 mg/mL oral solution twice daily. Primary outcomes were assessed by a blinded central adjudication committee. The primary efficacy outcome for the intention to treat population was the composite of symptomatic or clinically unsuspected venous thromboembolism, the primary safety outcome was major bleeding, and secondary safety outcomes included clinically relevant non-major (CRNM) bleeding. Patients were screened for venous thromboembolism by ultrasound and echocardiogram at the end of induction. The trial was registered with ClinicalTrials.gov (NCT02369653) and is now complete., Findings: Between Oct 22, 2015, and June 4, 2021, 512 participants were randomly assigned and included in analyses (222 [43%] female and 290 [57%] male; 388 [76%] White, 52 [10%] Asian, 24 [5%] Black or African American, and 48 [9%] other races; and 122 [24%] Hispanic or Latino ethnicity). During a median follow-up period of 27 days (IQR 26-28), 31 (12%) of 256 patients on apixaban had a composite venous thromboembolism compared with 45 (18%) of 256 participants receiving SOC (relative risk [RR] 0·69, 95% CI 0·45-1·05; p=0·080). Two major bleeding events occurred in each group (RR 1·0, 95% CI 0·14-7·01; p=1·0). A higher incidence of CRNM bleeding, primarily grade 1 or 2 epistaxis, occurred in the apixaban group (11 [4%] of 256 participants) compared with the SOC group (3 [1%] of 256; RR 3·67, 95% CI 1·04-12·97, p=0·030). The most frequent grade 3-5 adverse events in both groups were thrombocytopenia (n=28 for the apixaban group and n=20 for the SOC group) or platelet count decreased (n=49 and n=45), anaemia (n=77 and n=74), febrile neutropenia (n=27 and n=20), and neutropenia (n=16 and n=17) or neutrophil count decreased (n=22 and n=25). Five deaths occurred, which were due to infection (n=3 in the SOC group), cardiac arrest (n=1 in apixaban group), and haemorrhagic cerebral sinus vein thrombosis (n=1 in the SOC group). There was one apixaban-related death (coagulopathy and haemorrhage after cardiac arrest of unknown cause)., Interpretation: PREVAPIX-ALL is, to our knowledge, the first trial assessing primary thromboprophylaxis using a direct oral anticoagulant in paediatric patients with acute lymphoblastic leukaemia or lymphoma. No statistically significant treatment benefit was identified in participants receiving apixaban. Major and CRNM bleeding were infrequent overall, but a higher incidence of CRNM bleeding (primarily epistaxis in younger children) occurred in participants receiving apixaban. For patients deemed to be at particularly high risk of thrombosis, PREVAPIX-ALL provides encouraging safety data for the use of apixaban in clinical settings in which the potential benefits are thought to outweigh the risk of bleeding., Funding: Bristol Myers Squibb-Pfizer Alliance., Competing Interests: Declaration of interests GL, DM, MD, JLD, and NAF are employees of Bristol Myers Squibb. SHO'B and VR's institutions received salary support from the Children's Oncology Group for their roles as study chair and study vice-chair. LGM and JWN receive honoraria for their role as members of the study advisory committee, and JWN's institution receives compensation for her role as chair of the events adjudication committee. LGM has received research funding from Bristol Myers Squibb. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Lack of benefit from premedication for pegylated asparaginase during pediatric acute lymphoblastic leukemia/lymphoma therapy: A side-by-side comparison.
- Author
-
Menig S, Dinh A, Angus J, Tucker S, Leger KJ, Rushing T, and Orgel E
- Subjects
- Adolescent, Young Adult, Child, Humans, Infant, Asparaginase therapeutic use, Retrospective Studies, Polyethylene Glycols, Premedication, Antineoplastic Agents therapeutic use, Drug Hypersensitivity etiology, Drug Hypersensitivity prevention & control, Drug Hypersensitivity drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Hypersensitivity, Lymphoma drug therapy
- Abstract
Background: Pegylated l-asparaginase (PEG) is integral to treatment regimens for acute lymphoblastic leukemia (ALL) and lymphoma. Hypersensitivity reactions (HSRs) to PEG are common and can preclude continued administration. Data supporting recommendations for universal premedication (UPM) prior to PEG infusion to reduce incidence of HSRs are limited; UPM has become common practice., Procedures: Two free-standing children's hospitals independently implemented UPM prior to PEG infusions in 2016 and 2019, respectively. In a side-by-side retrospective analysis, incidence and severity of HSRs were analyzed pre- and postimplementation of UPM in youth ≥1 years old treated with frontline PEG-containing ALL regimens (2015-2018, 2016-2020). All HSRs were centrally reviewed within each institution to confirm and grade the HSR (Common Terminology Criteria for Adverse Events, v5). Planned analyses of subsets at potentially greater risk for HSRs included intensive PEG regimens (≥5 doses), adolescent and young adults (AYA), Hispanic/Latinx ethnicity, and obesity., Results: In 410 patients (by institution, n = 282 and n = 128), the overall incidence of Grade ≥ 3 HSRs was 20% (56 out of 282) and 18% (23 out of 128), respectively. No difference in incidence of Grade ≥ 3 HSRs in patients with versus without UPM was found at either institution (23 vs. 19%, p = .487 and 19 vs. 17%, p = .845). UPM also did not reduce the severity of HSRs, nor influence HSR risk within any patient subset., Conclusions: UPM prior to PEG infusion did not alter incidence or severity of HSRs at either institution. HSR remains a common complication of PEG therapy, impacting the patient experience. Alternative strategies to reduce HSRs are urgently required., (© 2023 The Authors. Pediatric Blood & Cancer published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
12. Dose-limiting mucositis: friend or foe?
- Author
-
Thornton CP and Orgel E
- Subjects
- Humans, Mucositis chemically induced, Neoplasms drug therapy
- Abstract
Dose-limiting toxicities are ubiquitous to cancer-directed therapy, presenting with severity to a degree that necessitates therapy de-escalation, pause, or discontinuation. To date, there is incredible limited understanding if these therapy de-escalations present risk for survival by limiting delivery of intensive therapy, or if they indicate physiologic susceptibility and are a favorable prognostic indicator. Mucositis is an excellent illustration of the current paradox of dose-limiting toxicities-it has existed alongside therapy for eight decades, but despite its presence, there is an incomplete understanding of how it develops, why it varies between oncologic populations, and if it relates to cancer survival. Rigorous methodologic approaches in symptom science holds potential to better understand mucositis, to determine if it is a marker of response or threat, and evaluate if it holds potential to guide therapy delivery., (© 2023. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
13. Reevaluation of sodium thiosulfate otoprotection using the consensus International Society of Paediatric Oncology Ototoxicity Scale: A report from the Children's Oncology Group study ACCL0431.
- Author
-
Orgel E, Knight KR, Villaluna D, Krailo M, Esbenshade AJ, Sung L, and Freyer DR
- Abstract
In two randomized trials (Children's Oncology Group ACCL0431 and International Childhood Liver Tumour Strategy Group SIOPEL-6), sodium thiosulfate (STS) demonstrated efficacy in preventing cisplatin-induced hearing loss (CIHL). However, the measures used in those trials have been superseded by the consensus International Society of Paediatric Oncology (SIOP) Ototoxicity Scale. To provide benchmark data for STS efficacy when using this contemporary scale, we reanalyzed ACCL0431 hearing outcomes with the SIOP scale and using multiple timepoints. Compared to the control arm, STS significantly reduced CIHL when assessed by the SIOP scale across these different approaches. These results provide critical data to inform treatment discussions and support future potential trial designs comparing otoprotectants., (© 2023 The Authors. Pediatric Blood & Cancer published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
14. Intravenous N-Acetylcysteine to Prevent Cisplatin-Induced Hearing Loss in Children: A Nonrandomized Controlled Phase I Trial.
- Author
-
Orgel E, Knight KR, Chi YY, Malvar J, Rushing T, Mena V, Eisenberg LS, Rassekh SR, Ross CJD, Scott EN, Neely M, Neuwelt EA, Muldoon LL, and Freyer DR
- Subjects
- Adolescent, Humans, Child, Cisplatin adverse effects, Acetylcysteine therapeutic use, Acetylcysteine adverse effects, Administration, Intravenous, Hearing Loss chemically induced, Hearing Loss prevention & control, Neoplasms drug therapy, Neoplasms chemically induced
- Abstract
Purpose: Cisplatin-induced hearing loss (CIHL) is common and permanent. As compared with earlier otoprotectants, we hypothesized N-acetylcysteine (NAC) offers potential for stronger otoprotection through stimulation of glutathione (GSH) production. This study tested the optimal dose, safety, and efficacy of NAC to prevent CIHL., Patients and Methods: In this nonrandomized, controlled phase Ia/Ib trial, children and adolescents newly diagnosed with nonmetastatic, cisplatin-treated tumors received NAC intravenously 4 hours post-cisplatin. The trial performed dose-escalation across three dose levels to establish a safe dose that exceeded the targeted peak serum NAC concentration of 1.5 mmol/L (as identified from preclinical models). Patients with metastatic disease or who were otherwise ineligible were enrolled in an observation-only/control arm. To evaluate efficacy, serial age-appropriate audiology assessments were performed. Integrated biology examined genes involved in GSH metabolism and post-NAC GSH concentrations., Results: Of 52 patients enrolled, 24 received NAC and 28 were in the control arm. The maximum tolerated dose was not reached; analysis of peak NAC concentration identified 450 mg/kg as the recommended phase II dose (RP2D). Infusion-related reactions were common. No severe adverse events occurred. Compared with the control arm, NAC decreased likelihood of CIHL at the end of cisplatin therapy [OR, 0.13; 95% confidence interval (CI), 0.021-0.847; P = 0.033] and recommendations for hearing intervention at end of study (OR, 0.082; 95% CI, 0.011-0.60; P = 0.014). NAC increased GSH; GSTP1 influenced risk for CIHL and NAC otoprotection., Conclusions: NAC was safe at the RP2D, with strong evidence for efficacy to prevent CIHL, warranting further development as a next-generation otoprotectant., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
15. Efficacy and safety of FLAG-IDA as front-line therapy in de novo paediatric acute myeloid leukaemia population.
- Author
-
Doan A, Huang HKT, Hadar AJ, Malvar J, Rushing T, Raca G, Kovach AE, Freyer DR, Parekh C, Stokke J, Posch LC, Dao J, Bhojwani D, Gaynon P, and Orgel E
- Subjects
- Child, Humans, Antineoplastic Combined Chemotherapy Protocols, Cytarabine therapeutic use, Granulocyte Colony-Stimulating Factor therapeutic use, Vidarabine therapeutic use, Idarubicin therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Published
- 2023
- Full Text
- View/download PDF
16. The International Consensus Classification of acute leukemias of ambiguous lineage.
- Author
-
Weinberg OK, Arber DA, Döhner H, Mullighan CG, Orgel E, Porwit A, Stone RM, and Borowitz MJ
- Subjects
- Humans, Consensus, Acute Disease, Immunophenotyping, Leukemia
- Published
- 2023
- Full Text
- View/download PDF
17. Symptom management care pathway adaptation process and specific adaptation decisions.
- Author
-
Vettese E, Sherani F, King AA, Yu L, Aftandilian C, Baggott C, Agarwal V, Nagasubramanian R, Kelly KM, Freyer DR, Orgel E, Bradfield SM, Kyono W, Roth M, Klesges LM, Beauchemin M, Grimes A, Tomlinson G, Dupuis LL, and Sung L
- Subjects
- Child, Humans, Palliative Care, Critical Pathways, Neoplasms
- Abstract
Background: There is substantial heterogeneity in symptom management provided to pediatric patients with cancer. The primary objective was to describe the adaptation process and specific adaptation decisions related to symptom management care pathways based on clinical practice guidelines. The secondary objective evaluated if institutional factors were associated with adaptation decisions., Methods: Fourteen previously developed symptom management care pathway templates were reviewed by an institutional adaptation team composed of two clinicians at each of 10 institutions. They worked through each statement for all care pathway templates sequentially. The institutional adaptation team made the decision to adopt, adapt or reject each statement, resulting in institution-specific symptom management care pathway drafts. Institutional adaption teams distributed the 14 care pathway drafts to their respective teams; their feedback led to care pathway modifications., Results: Initial care pathway adaptation decision making was completed over a median of 4.2 (interquartile range 2.0-5.3) weeks per institution. Across all institutions and among 1350 statements, 551 (40.8%) were adopted, 657 (48.7%) were adapted, 86 (6.4%) were rejected and 56 (4.1%) were no longer applicable because of a previous decision. Most commonly, the reason for rejection was not agreeing with the statement (70/86, 81.4%). Institutional-level factors were not significantly associated with statement rejection., Conclusions: Acceptability of the 14 care pathways was evident by most statements being adopted or adapted. The adaptation process was accomplished over a relatively short timeframe. Future work should focus on evaluation of care pathway compliance and determination of the impact of care pathway-consistent care on patient outcomes., Trial Registration: clinicaltrials.gov, NCT04614662. Registered 04/11/2020, https://clinicaltrials.gov/ct2/show/NCT04614662?term=NCT04614662&draw=2&rank=1 ., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
18. Special considerations in the design and implementation of pediatric otoprotection trials.
- Author
-
Freyer DR, Orgel E, Knight K, and Krailo M
- Subjects
- Child, Humans, Cisplatin adverse effects, Carboplatin adverse effects, Quality of Life, Antineoplastic Agents adverse effects, Cancer Survivors, Hearing Loss chemically induced
- Abstract
Purpose: Cisplatin-induced hearing loss (CIHL) is a common late effect after childhood cancer treatment having profound, lifelong consequences that lower quality of life. The recent identification of intravenous sodium thiosulfate (STS) as an effective agent for preventing pediatric CIHL represents a paradigm shift that has created new opportunities for expanding STS usage and developing additional otoprotectants. The purpose of this paper is to discuss key considerations and recommendations for the design and implementation of future pediatric otoprotection trials., Methods: An approach synthesizing published data and collective experience was used., Results: Key issues were identified in the categories of translational research, trial designs for systemic and intratympanic agents, measurement of ototoxicity, and biostatistical challenges., Conclusions: Future pediatric otoprotection trials should emphasize (1) deep integration of preclinical and early-phase studies; (2) an embedded or free-standing design for systemic agents based on mechanistic considerations; (3) use of suitable audiologic testing batteries for children, SIOP grading criteria, and submission of raw audiologic data for central review; and (4) novel endpoints and innovative study designs that maximize trial efficiency for limited sample sizes. Additional recommendations include routine collection of DNA specimens for assessing modifying effects of genetic susceptibility and meaningful inclusion of patient/family advocates for informing trial development., Implications for Cancer Survivors: Changing the historical paradigm from acceptance to prevention of pediatric CIHL through expanded research with existing and emerging otoprotectants will dramatically improve quality of life for future childhood cancer survivors exposed to cisplatin., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
19. Asparaginase toxicity in Hispanic adult and pediatric patients with acute lymphoblastic leukemia: current understanding.
- Author
-
Alqahtani A, Alhousari D, Ali A, Yaghmour G, Orgel E, Curran E, Stock W, Bhojwani D, and Alachkar H
- Subjects
- Humans, Child, Adult, Asparaginase adverse effects, Antineoplastic Agents adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Hypertriglyceridemia drug therapy, Chemical and Drug Induced Liver Injury drug therapy
- Abstract
Introduction: Asparaginase is essential to chemotherapy regimens for acute lymphoblastic leukemia (ALL). Survival of patients with ALL has improved since incorporating asparaginase into chemotherapy backbones. Hispanic patients have a higher incidence of ALL than other ethnicities and suffer inferior outcomes. The inferior outcome of Hispanics is due to several factors, including the increased incidence of high-risk genetic subtypes and susceptibility to treatment-related toxicity., Areas Covered: We summarize the current knowledge of asparaginase-related toxicity by comparing their incidence between Hispanic and non-Hispanic patients. These toxicities include hypersensitivity, hepatotoxicity, pancreatitis, thrombosis, and hypertriglyceridemia. The PubMed database and Google Scholar were used to search for this review from October 2022 to June 2023., Expert Opinion: Except for hepatotoxicity and hypertriglyceridemia secondary to asparaginase-based treatments, which may develop more frequently among Hispanic patients with ALL, other toxicities were comparable between Hispanic and non-Hispanic patients. Nevertheless, studies with larger cohorts and more accurate capturing of Hispanic ethnicity should be conducted to fill the gaps in the current knowledge.
- Published
- 2023
- Full Text
- View/download PDF
20. From soup to nuts: Obesity impairs chemotherapy during early and late phases of acute lymphoblastic leukemia treatment.
- Author
-
Orgel E and Mittelman SD
- Subjects
- Humans, Obesity drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Published
- 2023
- Full Text
- View/download PDF
21. Temsirolimus combined with cyclophosphamide and etoposide for pediatric patients with relapsed/refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium trial (TACL 2014-001).
- Author
-
Tasian SK, Silverman LB, Whitlock JA, Sposto R, Loftus JP, Schafer ES, Schultz KR, Hutchinson RJ, Gaynon PS, Orgel E, Bateman CM, Cooper TM, Laetsch TW, Sulis ML, Chi YY, Malvar J, Wayne AS, and Rheingold SR
- Subjects
- Adolescent, Alanine Transaminase therapeutic use, Child, Cyclophosphamide therapeutic use, Etoposide, Humans, MTOR Inhibitors, Phosphatidylinositol 3-Kinases, Phosphoinositide-3 Kinase Inhibitors, Phosphoproteins, Sirolimus analogs & derivatives, TOR Serine-Threonine Kinases, Antineoplastic Combined Chemotherapy Protocols adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling is commonly dysregulated in acute lymphoblastic leukemia (ALL). The TACL2014-001 phase I trial of the mTOR inhibitor temsirolimus in combination with cyclophosphamide and etoposide was performed in children and adolescents with relapsed/refractory ALL. Temsirolimus was administered intravenously (IV) on days 1 and 8 with cyclophosphamide 440 mg/m2 and etoposide 100 mg/m2 IV daily on days 1-5. The starting dose of temsirolimus was 7.5 mg/m2 (DL1) with escalation to 10 mg/m2 (DL2), 15 mg/m2 (DL3), and 25 mg/m2 (DL4). PI3K/mTOR pathway inhibition was measured by phosphoflow cytometry analysis of peripheral blood specimens from treated patients. Sixteen heavily-pretreated patients were enrolled with 15 evaluable for toxicity. One dose-limiting toxicity of grade 4 pleural and pericardial effusions occurred in a patient treated at DL3. Additional dose-limiting toxicities were not seen in the DL3 expansion or DL4 cohort. Grade 3/4 non-hematologic toxicities occurring in three or more patients included febrile neutropenia, elevated alanine aminotransferase, hypokalemia, mucositis, and tumor lysis syndrome and occurred across all doses. Response and complete were observed at all dose levels with a 47% overall response rate and 27% complete response rate. Pharmacodynamic correlative studies demonstrated dose-dependent inhibition of PI3K/mTOR pathway phosphoproteins in all studied patients. Temsirolimus at doses up to 25 mg/m2 with cyclophosphamide and etoposide had an acceptable safety profile in children with relapsed/refractory ALL. Pharmacodynamic mTOR target inhibition was achieved and appeared to correlate with temsirolimus dose. Future testing of next-generation PI3K/mTOR pathway inhibitors with chemotherapy may be warranted to increase response rates in children with relapsed/refractory ALL.
- Published
- 2022
- Full Text
- View/download PDF
22. Sodium thiosulfate for prevention of cisplatin-induced hearing loss: updated survival from ACCL0431.
- Author
-
Orgel E, Villaluna D, Krailo MD, Esbenshade A, Sung L, and Freyer DR
- Subjects
- Humans, Thiosulfates therapeutic use, Cisplatin adverse effects, Hearing Loss chemically induced, Hearing Loss prevention & control
- Abstract
Competing Interests: Funding supporting the ACCL0431 study was provided by the National Institutes of Health (NIH) through the National Cancer Institute Community Oncology Research Program (NCORP; grant UG1CA189955), the National Cancer Institute Clinical Trials Network (NCTN) Statistics and Data Center (grant U10CA180899), and by St Baldrick's Foundation. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. We declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
23. Erratum to: Cohen-Cutler S, Wong K, Mena V, et al. Hearing Loss Risk in Pediatric Patients Treated with Cranial Irradiation and Cisplatin-Based Chemotherapy. Int J Radiat Oncol Biol Phys 2021:110(5);1488-1495.
- Author
-
Cohen-Cutler S, Wong K, Mena V, Sianto K, Wright MA, Olch A, and Orgel E
- Published
- 2022
- Full Text
- View/download PDF
24. Ultra-High Dose Vitamin D in Pediatric Hematopoietic Stem Cell Transplantation: A Nonrandomized Controlled Trial.
- Author
-
Bhandari R, Aguayo-Hiraldo P, Malvar J, Cheng K, Sacapano A, Abdel-Azim H, Chi YY, Wallace G, Asgharzadeh S, Jodele S, and Orgel E
- Subjects
- Calcifediol, Child, Humans, Vitamin D, Vitamins, Hematopoietic Stem Cell Transplantation, Vitamin D Deficiency drug therapy
- Abstract
Vitamin D is essential for bone health and has immunomodulatory properties. Most pediatric patients are vitamin D insufficient (<30 ng/mL) before hematopoietic stem cell transplantation (HSCT). Standard supplementation strategies fail to achieve vitamin D sufficiency in the acute post-transplantation period, and there are scarce data to support optimal vitamin D supplementation in this patient population. This study aimed to evaluate whether a single, oral, weight-based ultra-high dose of vitamin D (Stoss dosing) was more effective than standard supplementation to achieve pre-HSCT vitamin D sufficiency and reduce the incidence of HSCT-related complications (acute graft-versus-host disease, veno-occlusive disease, and/or transplant-associated thrombotic microangiopathy) that are associated with immune-mediated endothelial damage. Secondary endpoints examined the immunomodulatory properties of vitamin D. We conducted a nonrandomized controlled clinical trial of Stoss-dosed vitamin D in pediatric patients receiving HSCT. The study prospectively enrolled 33 patients, 29 of whom successfully received Stoss-dosed vitamin D and were compared to 136 patients in a historical control. Patient characteristics were compared using Fisher's exact test or t-test. The one-sided Fisher's exact test was used for cohort comparison of the primary endpoints. Logistic regression was used to examine the association between patient-specific factors and total 25-hydroxy vitamin D (25-OHD) levels and the compiled HSCT complications. In the Stoss cohort, 97% (n = 28/29) of patients achieved pre-HSCT vitamin D sufficiency compared to 67% (n = 10/15) of patients in the historical control who were on standard supplementation at the time the total 25-OHD level was assessed (P = .013). The mean total 25-OHD level in the Stoss cohort was significantly higher than patients in the historical control who received standard supplementation (72.2 ng/mL versus 35.8 ng/mL, P < .001). Nine patients in the Stoss cohort maintained vitamin D sufficiency throughout the first 100 days after HSCT, and the remaining 19 patients maintained sufficiency for a median of 63 days (range 6-105 days) from the Stoss dose. Patients receiving Stoss-dosed vitamin D developed a lower combined incidence of HSCT-related complications than the historical control (25% [n = 7/28] versus 42% [n = 57/136], P = .055). After Stoss dosing, immunophenotyping studies found a significant decrease in subsets of CD8+ T cells and mononuclear cells (P = .040 and.013, respectively), and, in a subset of cells, larger decreases in phosphoprotein expression were seen with greater increases in total 25-OHD levels. Inflammatory cytokines did not change significantly after Stoss dosing. Stoss dosing is therefore a safe and effective approach to maintain vitamin D sufficiency in the immediate post-HSCT period and may be associated with decreased HSCT-related complications. Randomized studies are warranted to further investigate the efficacy of Stoss-dosed vitamin D to improve bone health and reduce complications in pediatric patients receiving HSCT., (Copyright © 2021 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
25. Levocarnitine for pegaspargase-induced hepatotoxicity in older children and young adults with acute lymphoblastic leukemia.
- Author
-
Schulte R, Hinson A, Huynh V, Breese EH, Pierro J, Rotz S, Mixon BA, McNeer JL, Burke MJ, and Orgel E
- Subjects
- Adolescent, Adult, Chemical and Drug Induced Liver Injury prevention & control, Child, Female, Humans, Induction Chemotherapy, Male, Pediatric Obesity complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Survival Analysis, Young Adult, Antineoplastic Agents adverse effects, Asparaginase adverse effects, Carnitine therapeutic use, Chemical and Drug Induced Liver Injury drug therapy, Chemical and Drug Induced Liver Injury etiology, Polyethylene Glycols adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Background: Pegaspargase (PEG-ASP) is an integral component of therapy for acute lymphoblastic leukemia (ALL) but is associated with hepatotoxicity that may delay or limit future therapy. Obese and adolescent and young adult (AYA) patients are at high risk. Levocarnitine has been described as potentially beneficial for the treatment or prevention of PEG-ASP-associated hepatotoxicity., Methods: We collected data for patients age ≥10 years who received levocarnitine during induction therapy for ALL, compared to a similar patient cohort who did not receive levocarnitine. The primary endpoint was conjugated bilirubin (c.bili) >3 mg/dl. Secondary endpoints were transaminases >10× the upper limit of normal and any Grade ≥3 hepatotoxicity., Results: Fifty-two patients received levocarnitine for prophylaxis (n = 29) or rescue (n = 32) of hepatotoxicity. Compared to 109 patients without levocarnitine, more patients receiving levocarnitine were obese and/or older and had significantly higher values for some hepatotoxicity markers at diagnosis and after PEG-ASP. Levocarnitine regimens varied widely; no adverse effects of levocarnitine were identified. Obesity and AYA status were associated with an increased risk of conjugated hyperbilirubinemia and severe transaminitis. Multivariable analysis identified a protective effect of levocarnitine on the development of c.bili >3 mg/dl (OR 0.12, p = 0.029). There was no difference between groups in CTCAE Grade ≥3 hepatotoxicity. C.bili >3 mg/dl during induction was associated with lower event-free survival., Conclusions: This real-world data on levocarnitine supplementation during ALL induction highlights the risk of PEG-ASP-associated hepatotoxicity in obese and AYA patients, and hepatotoxicity's potential impact on survival. Levocarnitine supplementation may be protective, but prospective studies are needed to confirm these findings., (© 2021 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
26. Hearing Loss Risk in Pediatric Patients Treated with Cranial Irradiation and Cisplatin-Based Chemotherapy.
- Author
-
Cohen-Cutler S, Wong K, Mena V, Sianto K, Wright MA, Olch A, and Orgel E
- Subjects
- Adolescent, Audiometry, Autografts, Bone Marrow Transplantation, Child, Child, Preschool, Cisplatin therapeutic use, Female, Humans, Infant, Logistic Models, Male, Odds Ratio, Organs at Risk radiation effects, Ototoxicity etiology, Radiation Dosage, Radiation-Sensitizing Agents therapeutic use, Radiotherapy Dosage, Retrospective Studies, Risk Factors, Young Adult, Cisplatin adverse effects, Cochlea radiation effects, Cranial Irradiation adverse effects, Hearing Loss etiology, Radiation-Sensitizing Agents adverse effects
- Abstract
Purpose: Cranial radiation therapy (RT) and cisplatin-based chemotherapy are essential to treating many pediatric cancers but cause significant ototoxicity. The objective of this study is to determine the relationship between the RT dose and the risk of subsequent hearing loss in pediatric patients treated with cisplatin., Methods and Materials: This retrospective study of cisplatin-treated pediatric patients examined ototoxicity from cranial RT. Ototoxicity was graded for each ear according to the International Society of Pediatric Oncology (SIOP) consensus ototoxicity scale. The RT dose to the cochlea was calculated using the mean, median, maximum, and minimum dose received to determine the most predictive parameter for hearing loss. Multivariable logistic regression models then examined risk factors for hearing loss., Results: In 96 children (161 ears) treated with RT + cisplatin, the minimum cochlear RT dose was most predictive of hearing loss. A higher cochlear RT dose was associated with increased hearing loss (odds ratio per 10 Gy dose increase = 1.64; P = .043), with an added risk in those receiving an autologous bone marrow transplantation (hazard ratio = 10.47; P < .001)., Conclusions: This research supports further testing of the minimum cochlear RT dose as a more predictive dose parameter for risk of ototoxicity. The cochlear RT dose was additive to the risk of hearing loss from underlying cisplatin-based chemotherapy. Exposure to autologous bone marrow transplantation was the strongest predictor of developing hearing loss, placing these children at particularly high risk for hearing loss across all cochlear doses. Future prospective studies are crucial to further inform RT dose thresholds and minimize the risk of hearing loss in childhood cancer survivors., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
27. Increased prevalence of CRLF2 rearrangements in obesity-associated acute lymphoblastic leukemia.
- Author
-
Mittelman SD, Kim J, Raca G, Li G, Oberley MJ, and Orgel E
- Subjects
- Adipose Tissue, Adolescent, Child, Cohort Studies, Female, Hispanic or Latino genetics, Humans, Male, Gene Rearrangement genetics, Obesity complications, Obesity genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Receptors, Cytokine genetics
- Published
- 2021
- Full Text
- View/download PDF
28. Effect of Body Fat on Population Pharmacokinetics of High-Dose Methotrexate in Pediatric Patients With Acute Lymphoblastic Leukemia.
- Author
-
Orgel E, Nabais T, Douglas C, Mittelman SD, and Neely M
- Subjects
- Adolescent, Antimetabolites, Antineoplastic administration & dosage, Area Under Curve, Child, Creatinine blood, Dose-Response Relationship, Drug, Female, Humans, Male, Methotrexate administration & dosage, Models, Biological, Obesity epidemiology, Prospective Studies, Young Adult, Adipose Tissue physiology, Antimetabolites, Antineoplastic pharmacokinetics, Methotrexate pharmacokinetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Nearly all international regimens for pediatric acute lymphoblastic leukemia (ALL) incorporate intravenous "high-dose" methotrexate (HDMTX, ≥1 g/m
2 ) to penetrate the central nervous system. Dosing is routinely adjusted for body surface area (BSA), but limited data describe the pharmacokinetics of HDMTX, particularly in obese and/or large patients. To understand the impact of body size (BSA) and body fat percentage (BFP) on HDMTX pharmacokinetics, we performed a secondary analysis of 36 children and adolescents 10-21 years old treated for newly diagnosed ALL and who were enrolled in a prospective study examining body composition. All patients received 5 g/m2 of HDMTX infused over 24 hours. Plasma methotrexate concentrations were measured at 24, 42, and 48 hours. At 48 hours, ≥0.4 μmol/L was defined as "delayed elimination," necessitating prolonged supportive care. BFP was measured using dual-energy x-ray absorptiometry. A nonparametric population pharmacokinetic model was constructed with subsequent simulations to explore effects of BSA and BFP extremes. Despite standard BSA-adjusted dosing, we found significant intrapatient variability in mean MTX concentration (38%; range, 1.2%-86%). BSA and BFP were not linearly associated with increased area under the curve (AUC, P = 0.74 and P = 0.12), but both larger size (BSA) and greater obesity (BFP) were associated with an approximately 2-fold higher risk for delayed elimination at 48 hours. HDMTX AUC was not associated with toxicity. MTX pharmacokinetics vary among and even within patients despite BSA-adjusted dosing. Obesity and large size are identified as new risk factors for delayed elimination, requiring further investigation., (© 2020, The American College of Clinical Pharmacology.)- Published
- 2021
- Full Text
- View/download PDF
29. Adipocytes Provide Fatty Acids to Acute Lymphoblastic Leukemia Cells.
- Author
-
Tucci J, Chen T, Margulis K, Orgel E, Paszkiewicz RL, Cohen MD, Oberley MJ, Wahhab R, Jones AE, Divakaruni AS, Hsu CC, Noll SE, Sheng X, Zare RN, and Mittelman SD
- Abstract
Background: There is increasing evidence that adipocytes play an active role in the cancer microenvironment. We have previously reported that adipocytes interact with acute lymphoblastic leukemia (ALL) cells, contributing to chemotherapy resistance and treatment failure. In the present study, we investigated whether part of this resistance is due to adipocyte provision of lipids to ALL cells., Methods: We cultured 3T3-L1 adipocytes, and tested whether ALL cells or ALL-released cytokines induced FFA release. We investigated whether ALL cells took up these FFA, and using fluorescent tagged BODIPY-FFA and lipidomics, evaluated which lipid moieties were being transferred from adipocytes to ALL. We evaluated the effects of adipocyte-derived lipids on ALL cell metabolism using a Seahorse XF analyzer and expression of enzymes important for lipid metabolism, and tested whether these lipids could protect ALL cells from chemotherapy. Finally, we evaluated a panel of lipid synthesis and metabolism inhibitors to determine which were affected by the presence of adipocytes., Results: Adipocytes release free fatty acids (FFA) when in the presence of ALL cells. These FFA are taken up by the ALL cells and incorporated into triglycerides and phospholipids. Some of these lipids are stored in lipid droplets, which can be utilized in states of fuel deprivation. Adipocytes preferentially release monounsaturated FFA, and this can be attenuated by inhibiting the desaturating enzyme steroyl-CoA decarboxylase-1 (SCD1). Adipocyte-derived FFA can relieve ALL cell endogenous lipogenesis and reverse the cytotoxicity of pharmacological acetyl-CoA carboxylase (ACC) inhibition. Further, adipocytes alter ALL cell metabolism, shifting them from glucose to FFA oxidation. Interestingly, the unsaturated fatty acid, oleic acid, protects ALL cells from modest concentrations of chemotherapy, such as those that might be present in the ALL microenvironment. In addition, targeting lipid synthesis and metabolism can potentially reverse adipocyte protection of ALL cells., Conclusion: These findings uncover a previously unidentified interaction between ALL cells and adipocytes, leading to transfer of FFA for use as a metabolic fuel and macromolecule building block. This interaction may contribute to ALL resistance to chemotherapy, and could potentially be targeted to improve ALL treatment outcome., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Tucci, Chen, Margulis, Orgel, Paszkiewicz, Cohen, Oberley, Wahhab, Jones, Divakaruni, Hsu, Noll, Sheng, Zare and Mittelman.)
- Published
- 2021
- Full Text
- View/download PDF
30. Caloric and nutrient restriction to augment chemotherapy efficacy for acute lymphoblastic leukemia: the IDEAL trial.
- Author
-
Orgel E, Framson C, Buxton R, Kim J, Li G, Tucci J, Freyer DR, Sun W, Oberley MJ, Dieli-Conwright C, and Mittelman SD
- Subjects
- Adolescent, Child, Humans, Neoplasm, Residual, Nutrients, Obesity, Prospective Studies, Young Adult, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Being overweight or obese (OW/OB) during B-cell acute lymphoblastic leukemia (B-ALL) induction is associated with chemoresistance as quantified by minimal residual disease (MRD). We hypothesized that caloric and nutrient restriction from diet/exercise could lessen gains in fat mass (FM) and reduce postinduction MRD. The Improving Diet and Exercise in ALL (IDEAL) trial enrolled patients 10 to 21 years old, newly diagnosed with B-ALL (n = 40), in comparison with a recent historical control (n = 80). Designed to achieve caloric deficits ≥20% during induction, reduce fat intake/glycemic load, and increase activity, IDEAL's end points were FM gain (primary), MRD ≥0.01%, and adherence/feasibility. Integrated biology explored biomarkers of OW/OB physiology. IDEAL intervention did not significantly reduce median FM change from baseline overall (+5.1% [interquartile range [IQR], 15.8] vs +10.7% [IQR, 16.0]; P = .13), but stratified analysis showed benefit in those OW/OB (+1.5% [IQR, 6.6] vs +9.7% [IQR, 11.1]; P = .02). After accounting for prognostic factors, IDEAL intervention significantly reduced MRD risk (odds ratio, 0.30; 95% confidence interval, 0.09-0.92; P = .02). The trial exceeded its adherence (≥75% of overall diet) and feasibility (≥80% completed visits) thresholds. Integrated biology found the IDEAL intervention increased circulating adiponectin and reduced insulin resistance. The IDEAL intervention was feasible, decreased fat gain in those OW/OB, and reduced MRD. This is the first study in any hematologic malignancy to demonstrate potential benefit from caloric restriction via diet/exercise to augment chemotherapy efficacy and improve disease response. A prospective, randomized trial is warranted for validation. These trials were registered at www.clinicaltrials.gov as #NCT02708108 (IDEAL trial) and #NCT01317940 (historical control)., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
31. Association of body mass index with toxicity and survival in pediatric patients treated with cisplatin-containing regimens.
- Author
-
Bhandari R, Scott E, Yeh MY, Wong K, Rushing T, Huh W, and Orgel E
- Subjects
- Adolescent, Antineoplastic Agents therapeutic use, Antineoplastic Agents toxicity, Body Mass Index, Child, Cisplatin therapeutic use, Cisplatin toxicity, Female, Humans, Male, Progression-Free Survival, Proportional Hazards Models, Retrospective Studies, Survival Analysis, Antineoplastic Agents adverse effects, Cisplatin adverse effects, Malnutrition complications, Neoplasms complications, Neoplasms drug therapy, Obesity complications
- Abstract
Malnutrition is associated with treatment-related toxicities (TRT) in adults with solid tumors and in children with leukemia. Few studies have assessed whether malnutrition in pediatric patients treated for solid tumors impacts risk for TRT, relapse, and/or survival. To address this knowledge gap, this retrospective study evaluated the association between body mass index (BMI) at diagnosis, and imputed BMI during therapy, on the prevalence of TRT, specific toxicities, relapse, and survival in pediatric patients with solid tumors treated with cisplatin-containing regimens. Kaplan-Meier curves and regression models evaluated the association between patient-specific characteristics (including BMI) and TRT, relapse, and survival. The cohort included 221 patients, of whom 22% were malnourished at diagnosis (10% were underweight and 12% were obese). Most patients (60%) experienced at least one severe TRT, and 30% developed more than one severe TRT. Most patients with obesity at diagnosis remained obese during therapy (62%). In multivariable analysis, obesity at diagnosis was significantly associated with a more than threefold greater risk for developing severe TRT (p = 0.037), specifically for acute or chronic kidney injury ( p = 0.014). Obesity at diagnosis and adolescent and young adult age (≥15 years at diagnosis) were associated with worse event-free survival (hazard ratio [HR] 2.32, p = 0.024 and HR 2.28, p = 0.010, respectively) and overall survival (HR 3.69, p = 0.006 and HR 2.6, p = 0.012, respectively). Obese and older patients therefore constitute populations at risk for poorer outcomes. Prospective studies are warranted to gain further insight into the mechanism and role of obesity and adolescence in developing TRT and/or treatment failure.
- Published
- 2021
- Full Text
- View/download PDF
32. Prevalence and risk factors for cisplatin-induced hearing loss in children, adolescents, and young adults: a multi-institutional North American cohort study.
- Author
-
Moke DJ, Luo C, Millstein J, Knight KR, Rassekh SR, Brooks B, Ross CJD, Wright M, Mena V, Rushing T, Esbenshade AJ, Carleton BC, and Orgel E
- Subjects
- Adolescent, Adult, Age Distribution, Bone Neoplasms drug therapy, Bone Neoplasms epidemiology, Canada epidemiology, Central Nervous System Neoplasms drug therapy, Central Nervous System Neoplasms epidemiology, Child, Child, Preschool, Cisplatin therapeutic use, Dose-Response Relationship, Drug, Female, Hearing Loss, Sensorineural chemically induced, Hepatoblastoma drug therapy, Hepatoblastoma epidemiology, Humans, Infant, Infant, Newborn, Liver Neoplasms drug therapy, Liver Neoplasms epidemiology, Male, Neoplasms, Germ Cell and Embryonal drug therapy, Neoplasms, Germ Cell and Embryonal epidemiology, Neuroblastoma drug therapy, Neuroblastoma epidemiology, Odds Ratio, Osteosarcoma drug therapy, Osteosarcoma epidemiology, Ototoxicity etiology, Prevalence, Risk Factors, Severity of Illness Index, United States epidemiology, Young Adult, Antineoplastic Agents adverse effects, Cisplatin adverse effects, Hearing Loss, Sensorineural epidemiology, Neoplasms drug therapy, Ototoxicity epidemiology, Vincristine therapeutic use
- Abstract
Background: Cisplatin is used to treat a wide range of childhood cancers and cisplatin-induced hearing loss (CIHL) is a common and debilitating toxicity. We aimed to address persistent knowledge gaps in CIHL by establishing benchmarks for the prevalence of and risk factors for CIHL., Methods: In this multi-institutional cohort study, children (age 0-14 years), adolescents, and young adults (age 15-39 years) diagnosed with a cisplatin-treated tumour from paediatric cancer centres, who had available cisplatin dosing information, and primary audiology data for central review from consortia located in Canada and the USA were eligible for inclusion. Audiology was centrally reviewed and CIHL graded using the consensus International Society of Pediatric Oncology (SIOP) Boston Ototoxicity Scale. We assessed the prevalence of moderate or severe CIHL (SIOP grade ≥2) at latest follow-up and end of therapy, in each demographic, diagnosis, and treatment group and their relative contributions to risk for CIHL. Secondary endpoints explored associations of cisplatin dose reductions and CIHL with survival. We also examined whether cisplatin dose reductions and CIHL were associated with survival outcomes., Findings: We included 1481 patients who received cisplatin. Of the 1414 (95·5%) participants who had audiometry at latest follow-up (mean 3·9 years [SD 4·2] since diagnosis), 620 (43·8%) patients developed moderate or severe CIHL. The highest prevalence of CIHL was seen in the youngest patients (aged <5 years; 360 [59·4%] of 606 patients) and those with a CNS tumour (221 [50·9%] of 434 patients), hepatoblastoma (110 [65·9%] of 167 patients), or neuroblastoma (154 [62·1%] of 248 patients). After accounting for cumulative cisplatin dose, higher fractionated doses were associated with risk for CIHL (for each 10mg/m
2 increase per day, adjusted odds ratio [aOR] 1·15 [95% CI 1·07-1·25]; for each 50 mg/m2 increase per cycle aOR 2·16 [1·37-3·51]). Vincristine exposure was newly identified as a risk factor for CIHL (aOR 3·55 [2·19-5·84]). Dose reductions and moderate or severe CIHL were not significantly associated with survival differences., Interpretation: Using this large, multicentre cohort, benchmarks were established for the prevalence of CIHL in patients treated with cisplatin. Variations in cisplatin dosing confer additive risk for developing CIHL and warrant investigation as a potential approach to decrease the burden of therapy., Funding: US National Institutes of Health and National Institute on Deafness and Other Communication Disorders, US National Institutes of Health and National Cancer institute, St Baldrick's Foundation, Genome Canada, Genome British Columbia, Canadian Institutes of Health Research, the Canada Foundation for Innovation, University of British Columbia, British Columbia Children's Hospital Research Institute, British Columbia Provincial Health Services Authority, Health Canada, and C17 Research Network., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
33. Mixed Phenotype Acute Leukemia: Current Approaches to Diagnosis and Treatment.
- Author
-
Alexander TB and Orgel E
- Subjects
- Humans, Prognosis, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Purpose of Review: Mixed phenotype acute leukemia (MPAL) is a rare subtype of acute leukemia with features of both acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). The review examines current definitions and controversies in classification of MPAL, new insights into genomic drivers and pathogenesis, recent evidence to support treatment recommendations, and opportunities for future research., Recent Findings: Recent collaborative efforts have made progress in understanding the genomic landscape and optimal therapy for MPAL. The preponderance of retrospective data supports beginning therapy with ALL directed regimens. Differences in prognosis for adult and children with MPAL have led to divergent approaches for therapy intensity, including use of stem cell transplantation consolidation. MPAL remains a challenging leukemia to understand, research, and treat due to low incidence, shifting and subjective approaches to classification, and innate biological heterogeneity. Ongoing research hopes to surmount these obstacles through prospective studies within large cooperative groups to provide new insight into targetable biology and further refine optimal therapy.
- Published
- 2021
- Full Text
- View/download PDF
34. Preexisting or therapy-induced mutations in relapsed acute lymphoblastic leukemia?
- Author
-
Gaynon PS, Orgel E, and Ji L
- Subjects
- Genomics, Humans, Mutation, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2020
- Full Text
- View/download PDF
35. Assessment of provider perspectives on otoprotection research for children and adolescents: A Children's Oncology Group Cancer Control and Supportive Care Committee survey.
- Author
-
Orgel E, Freyer DR, Ullrich NJ, Hardy KK, Thomas SM, Dvorak CC, and Esbenshade AJ
- Subjects
- Adolescent, Antioxidants therapeutic use, Child, Follow-Up Studies, Hearing Loss chemically induced, Hearing Loss pathology, Humans, Neoplasms pathology, Prognosis, Surveys and Questionnaires, Antineoplastic Agents adverse effects, Cisplatin adverse effects, Hearing Loss prevention & control, Neoplasms drug therapy, Thiosulfates therapeutic use
- Abstract
Background: Cisplatin-induced hearing loss (CIHL) is a common and debilitating toxicity for childhood cancer survivors. Understanding provider perspectives is crucial to developing otoprotection studies that are both informative and feasible. Two international trials (ACCL0431 and SIOPEL6) investigated the drug sodium thiosulfate (STS) as an otoprotectant, but definitive interpretation of the findings of these trials has been challenging. Adoption of STS has therefore been uneven, and provider perspectives on its role are unknown., Procedure: The Children's Oncology Group (COG) Cancer Control and Supportive Care Neurotoxicity Subcommittee therefore conducted a survey of providers at COG institutions to determine perspectives on pediatric otoprotection practices and research surrounding three major themes: (1) prevalence of routine use of STS with cisplatin-based regimens, (2) application of audiometry to cisplatin therapy, and (3) preferred modalities for otoprotection research., Results: Survey respondents (45%, 44/98 surveyed institutions) were of diverse institutional sizes, practice settings, and geographical locations primarily in the United States and Canada. Overall, respondents considered CIHL an important toxicity and indicated strong enthusiasm for future studies (98%, 40/41). Results indicated that while STS was the current or planned standard of care in a minority of responding institutions (36%, 16/44), most sites were receptive to its inclusion in appropriate study designs. Application of audiometry for ototoxicity monitoring varied widely across sites. For otoprotection research, systemic agents were preferred (68%, 28/41) as compared with intratympanic approaches., Conclusion: These results suggest that pediatric otoprotection trials remain of interest to providers; the emphasis of these trials should remain on systemic and not intratympanic therapy., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
36. Integrin α6 mediates the drug resistance of acute lymphoblastic B-cell leukemia.
- Author
-
Gang EJ, Kim HN, Hsieh YT, Ruan Y, Ogana HA, Lee S, Pham J, Geng H, Park E, Klemm L, Willman CL, Carroll WL, Mittelman SD, Orgel E, Oberley MJ, Parekh C, Abdel-Azim H, Bhojwani D, Wayne AS, De Arcangelis A, Georges-Labouesse E, Wayner E, Bonig H, Minasyan A, Ten Hoeve J, Graeber TG, Müschen M, Heisterkamp N, and Kim YM
- Subjects
- Animals, Antibodies, Neoplasm pharmacology, Antibodies, Neutralizing pharmacology, Apoptosis drug effects, Apoptosis genetics, Female, Humans, Male, Mice, Mice, Knockout, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Gene Deletion, Integrin alpha6 genetics, Integrin alpha6 metabolism, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, Pyrimidines pharmacology
- Abstract
Resistance to multimodal chemotherapy continues to limit the prognosis of acute lymphoblastic leukemia (ALL). This occurs in part through a process called adhesion-mediated drug resistance, which depends on ALL cell adhesion to the stroma through adhesion molecules, including integrins. Integrin α6 has been implicated in minimal residual disease in ALL and in the migration of ALL cells to the central nervous system. However, it has not been evaluated in the context of chemotherapeutic resistance. Here, we show that the anti-human α6-blocking Ab P5G10 induces apoptosis in primary ALL cells in vitro and sensitizes primary ALL cells to chemotherapy or tyrosine kinase inhibition in vitro and in vivo. We further analyzed the underlying mechanism of α6-associated apoptosis using a conditional knockout model of α6 in murine BCR-ABL1+ B-cell ALL cells and showed that α6-deficient ALL cells underwent apoptosis. In vivo deletion of α6 in combination with tyrosine kinase inhibitor (TKI) treatment was more effective in eradicating ALL than treatment with a TKI (nilotinib) alone. Proteomic analysis revealed that α6 deletion in murine ALL was associated with changes in Src signaling, including the upregulation of phosphorylated Lyn (pTyr507) and Fyn (pTyr530). Thus, our data support α6 as a novel therapeutic target for ALL., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
37. Significance of minimal residual disease in pediatric mixed phenotype acute leukemia: a multicenter cohort study.
- Author
-
Oberley MJ, Raikar SS, Wertheim GB, Malvar J, Sposto R, Rabin KR, Punia JN, Seif AE, Cahen VC, Schore RJ, Luca DC, Guinipero T, Woods WG, O'Gorman MRG, and Orgel E
- Subjects
- Child, Cohort Studies, Female, Follow-Up Studies, Hematopoietic Stem Cell Transplantation adverse effects, Humans, Leukemia classification, Leukemia pathology, Leukemia therapy, Male, Neoplasm, Residual epidemiology, Neoplasm, Residual pathology, Phenotype, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Prognosis, Survival Rate, United States epidemiology, Hematopoietic Stem Cell Transplantation mortality, Induction Chemotherapy mortality, Leukemia mortality, Neoplasm, Residual mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality
- Abstract
The rarity of mixed phenotype acute leukemia (MPAL) has precluded adequate data to incorporate minimal residual disease (MRD) monitoring into therapy. Fluidity in MPAL classification systems further complicates understanding its biology and outcomes; this includes uncertainty surrounding the impact of shifting diagnostic requirements even between iterations of the World Health Organization (WHO) classification. Our primary objective was to address these knowledge gaps. To do so, we analyzed clinicopathologic features, therapy, MRD, and survival in a centrally-reviewed, multicenter cohort of MPAL uniformly diagnosed by the WHO classification and treated with acute lymphoblastic leukemia (ALL) regimens. ALL induction therapy achieved an EOI MRD negative (<0.01%) remission in most patients (70%). EOI MRD positivity was predictive of 5-year EFS (HR = 6.00, p < 0.001) and OS (HR = 9.57, p = 0.003). Patients who cleared MRD by EOC had worse survival compared with those EOI MRD negative. In contrast to adults with MPAL, ALL therapy without transplantation was adequate to treat most pediatric patients. Earlier MRD clearance was associated with better treatment success and survival. Prospective trials are now necessary to validate and refine MRD thresholds within the pediatric MPAL population and to identify salvage strategies for those with poor predicted survival.
- Published
- 2020
- Full Text
- View/download PDF
38. Clinical features and outcomes of patients with Shwachman-Diamond syndrome and myelodysplastic syndrome or acute myeloid leukaemia: a multicentre, retrospective, cohort study.
- Author
-
Myers KC, Furutani E, Weller E, Siegele B, Galvin A, Arsenault V, Alter BP, Boulad F, Bueso-Ramos C, Burroughs L, Castillo P, Connelly J, Davies SM, DiNardo CD, Hanif I, Ho RH, Karras N, Manalang M, McReynolds LJ, Nakano TA, Nalepa G, Norkin M, Oberley MJ, Orgel E, Pastore YD, Rosenthal J, Walkovich K, Larson J, Malsch M, Elghetany MT, Fleming MD, and Shimamura A
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Combined Modality Therapy, Female, Follow-Up Studies, Humans, Infant, Infant, Newborn, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy, Male, Middle Aged, Myelodysplastic Syndromes pathology, Myelodysplastic Syndromes therapy, Prognosis, Retrospective Studies, Shwachman-Diamond Syndrome pathology, Shwachman-Diamond Syndrome therapy, Survival Rate, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Hematopoietic Stem Cell Transplantation mortality, Leukemia, Myeloid, Acute mortality, Myelodysplastic Syndromes mortality, Shwachman-Diamond Syndrome mortality
- Abstract
Background: Data to inform surveillance and treatment for leukaemia predisposition syndromes are scarce and recommendations are largely based on expert opinion. This study aimed to investigate the clinical features and outcomes of patients with myelodysplastic syndrome or acute myeloid leukaemia and Shwachman-Diamond syndrome, an inherited bone marrow failure disorder with high risk of developing myeloid malignancies., Methods: We did a multicentre, retrospective, cohort study in collaboration with the North American Shwachman-Diamond Syndrome Registry. We reviewed patient medical records from 17 centres in the USA and Canada. Patients with a genetic (biallelic mutations in the SBDS gene) or clinical diagnosis (cytopenias and pancreatic dysfunction) of Shwachman-Diamond syndrome who developed myelodysplastic syndrome or acute myeloid leukaemia were eligible without additional restriction. Medical records were reviewed between March 1, 2001, and Oct 5, 2017. Masked central review of bone marrow pathology was done if available to confirm leukaemia or myelodysplastic syndrome diagnosis. We describe the clinical features and overall survival of these patients., Findings: We initially identified 37 patients with Shwachman-Diamond syndrome and myelodysplastic syndrome or acute myeloid leukaemia. 27 patients had samples available for central pathology review and were reclassified accordingly (central diagnosis concurred with local in 15 [56%] cases), 10 had no samples available and were classified based on the local review data, and 1 patient was excluded at this stage as not eligible. 36 patients were included in the analysis, of whom 10 (28%) initially presented with acute myeloid leukaemia and 26 (72%) initially presented with myelodysplastic syndrome. With a median follow-up of 4·9 years (IQR 3·9-8·4), median overall survival for patients with myelodysplastic syndrome was 7·7 years (95% CI 0·8-not reached) and 0·99 years (95% CI 0·2-2·4) for patients with acute myeloid leukaemia. Overall survival at 3 years was 11% (95% CI 1-39) for patients with leukaemia and 51% (29-68) for patients with myelodysplastic syndrome. Management and surveillance were variable. 18 (69%) of 26 patients with myelodysplastic syndrome received upfront therapy (14 haematopoietic stem cell transplantation and 4 chemotherapy), 4 (15%) patients received no treatment, 2 (8%) had unavailable data, and 2 (8%) progressed to acute myeloid leukaemia before receiving treatment. 12 patients received treatment for acute myeloid leukaemia-including the two patients initially diagnosed with myelodysplastic who progressed- two (16%) received HSCT as initial therapy and ten (83%) received chemotherapy with intent to proceed with HSCT. 33 (92%) of 36 patients (eight of ten with leukaemia and 25 of 26 with myelodysplastic syndrome) were known to have Shwachman-Diamond syndrome before development of a myeloid malignancy and could have been monitored with bone marrow surveillance. Bone marrow surveillance before myeloid malignancy diagnosis was done in three (33%) of nine patients with leukaemia for whom surveillance status was confirmed and 11 (46%) of 24 patients with myelodysplastic syndrome. Patients monitored had a 3-year overall survival of 62% (95% CI 32-82; n=14) compared with 28% (95% CI 10-50; n=19; p=0·13) without surveillance. Six (40%) of 15 patients with available longitudinal data developed myelodysplastic syndrome in the setting of stable blood counts., Interpretation: Our results suggest that prognosis is poor for patients with Shwachman-Diamond syndrome and myelodysplastic syndrome or acute myeloid leukaemia owing to both therapy-resistant disease and treatment-related toxicities. Improved surveillance algorithms and risk stratification tools, studies of clonal evolution, and prospective trials are needed to inform effective prevention and treatment strategies for leukaemia predisposition in patients with Shwachman-Diamond syndrome., Funding: National Institute of Health., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Mixed-phenotype acute leukemia: A cohort and consensus research strategy from the Children's Oncology Group Acute Leukemia of Ambiguous Lineage Task Force.
- Author
-
Orgel E, Alexander TB, Wood BL, Kahwash SB, Devidas M, Dai Y, Alonzo TA, Mullighan CG, Inaba H, Hunger SP, Raetz EA, Gamis AS, Rabin KR, Carroll AJ 3rd, Heerema NA, Berman JN, Woods WG, Loh ML, Zweidler-McKay PA, and Horan JT
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Clinical Trials as Topic, Disease-Free Survival, Female, Humans, Immunophenotyping methods, Infant, Leukemia, Biphenotypic, Acute pathology, Male, Pediatrics trends, World Health Organization, Young Adult, Hematopoietic Stem Cell Transplantation methods, Leukemia, Biphenotypic, Acute epidemiology, Leukemia, Biphenotypic, Acute therapy, Prognosis
- Abstract
Background: Optimal chemotherapy for treating mixed-phenotype acute leukemia (MPAL) and the role of hematopoietic stem cell transplantation (HSCT) remain uncertain. Major limitations in interpreting available data are MPAL's rarity and the use of definitions other than the currently widely accepted criteria: the World Health Organization 2016 (WHO2016) classification., Methods: To assess the relative efficacy of chemotherapy types for treating pediatric MPAL, the Children's Oncology Group (COG) Acute Leukemia of Ambiguous Lineage Task Force assembled a retrospective cohort of centrally reviewed WHO2016 MPAL cases selected from banking studies for acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). Patients were not treated in COG trials; treatment and outcome data were captured separately. The findings were then integrated with the available, mixed literature to develop a prospective trial in pediatric MPAL., Results: The central review confirmed that 54 of 70 cases fulfilled WHO2016 criteria for MPAL. ALL induction regimens achieved remission in 72% of the cases (28 of 39), whereas AML regimens achieved remission in 69% (9 of 13). The 5-year event-free survival (EFS) and overall survival (OS) rates for the entire cohort were 72% ± 8% and 77% ± 7%, respectively. EFS and OS were 75% ± 13% and 84% ± 11%, respectively, for those receiving ALL chemotherapy alone without HSCT (n = 21)., Conclusions: The results of the COG MPAL cohort and a literature review suggest that ALL chemotherapy without HSCT may be the preferred initial therapy. A prospective trial within the COG is proposed to investigate this approach; AML chemotherapy and/or HSCT will be reserved for those with treatment failure as assessed by minimal residual disease. Embedded biology studies will provide further insight into MPAL genomics., (© 2019 American Cancer Society.)
- Published
- 2020
- Full Text
- View/download PDF
40. Association between Vitamin D and Risk for Early and Late Post-Transplant Complications.
- Author
-
Bhandari R, Malvar J, Sacapano A, Aguayo-Hiraldo P, Jodele S, and Orgel E
- Subjects
- Child, Humans, Prospective Studies, Retrospective Studies, Vitamin D, Graft vs Host Disease etiology, Graft vs Host Disease prevention & control, Hematopoietic Stem Cell Transplantation adverse effects, Vitamin D Deficiency
- Abstract
Vitamin D (VD) deficiency is a well-described phenomenon in pediatric patients undergoing hematopoietic stem cell transplant (HSCT). VD modulates inflammation, and deficiency pre-HSCT and at day +100 has been associated with graft-versus-host disease (GVHD) and poorer survival. However, a paucity of data has specifically described the association between VD status and immune-mediated complications including GVHD and veno-occlusive disease (VOD). Additionally, data to guide recommendations for VD monitoring and supplementation during HSCT are scarce. Our primary objective was to evaluate the association between VD and post-HSCT complications. The key secondary aim was to evaluate the routine use and efficacy of VD monitoring and supplementation practices. To our knowledge, this is the largest study of its kind in the pediatric population. This retrospective study evaluated VD level (VDL) before and 1 year after HSCT, VD supplementation practices, and their association with acute GVHD, VOD, and survival in pediatric patients who received autologous and allogeneic HSCT for both malignant and nonmalignant diseases from January 2013 to April 2018. Of 314 HSCTs, 43% of patients (n = 136) had VDL measured before HSCT; 61% of this cohort had pre-HSCT VD insufficiency (<30 ng/mL). Neither pre-HSCT nor follow-up VDL was associated with the incidence of GVHD or VOD. Supplementation did not result in significantly different post-HSCT VDL.VDL was correlated with overall survival; every 10-ng/mL increase in VDL was associated with a 28% decreased risk of death (P = .01). Current accepted VD supplementation regimens for pediatric HSCT do not achieve sufficient VDL in most patients after HSCT. VD status was associated with all-cause mortality but not with individual comorbidities; prospective studies are required to establish the connection between VD status, inflammatory-mediated HSCT complications, and potential benefit of VD supplementation before and after HSCT. These studies are needed to inform evidence-based guidelines for monitoring and supplementing VD during HSCT., (Copyright © 2019 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
41. Levocarnitine does not impair chemotherapy cytotoxicity against acute lymphoblastic leukemia.
- Author
-
Sea JL, Orgel E, Chen T, Paszkiewicz RL, Krall AS, Oberley MJ, Stiles L, and Mittelman SD
- Subjects
- Acute Disease, Animals, Antineoplastic Combined Chemotherapy Protocols adverse effects, Asparaginase therapeutic use, Humans, Induction Chemotherapy, Mice, Carnitine therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Asparaginase (ASNase) is an integral part of pediatric induction chemotherapy that has also been shown to improve adult survival rates; however, pegylated (PEG)-ASNase induces severe hepatotoxicity in this population. Recent case reports describe the incorporation of levocarnitine (LC) supplementation into PEG-ASNase-containing induction regimens to prevent or treat hepatotoxicity. Because LC facilitates the metabolism of free fatty acids (FFA), a primary fuel source for ALL cells, LC could potentially interfere with ALL chemotherapy efficacy. To test this, we employed in vitro and in vivo models of ALL. We show in vitro that LC supplementation does not impact cytotoxicity from vincristine, daunorubicin, dexamethasone, or ASNase on human ALL cells nor lead to an increase in ALL cell metabolic rate. In vivo , we demonstrate LC does not impair PEG-ASNase monotherapy in mice with syngeneic ALL. Together, our findings show that LC supplementation is a safe strategy to prevent/reverse ASNase-induced toxicities in preclinical models.
- Published
- 2020
- Full Text
- View/download PDF
42. Improving the Timeliness of Chemotherapy Administration in the Bone Marrow Transplant Unit.
- Author
-
Bhandari R, Orgel E, Rushing T, Malicse K, Evangelista V, Jodele S, and Dandoy CE
- Subjects
- Adult, Female, Humans, Male, Time Factors, Antineoplastic Agents administration & dosage, Drug Chronotherapy, Hematopoietic Stem Cell Transplantation, Hospitalization, Quality Improvement, Transplantation Conditioning
- Abstract
Patients undergoing hematopoietic stem cell transplantation (HSCT) are often admitted to the hospital the day they are due to begin their conditioning regimen. Timely initiation of chemotherapy during regular work hours is important for patient safety, because during the night shift fewer physicians and pharmacists are available for urgent or unexpected matters. A review of the data at our institution from October 2017 to August 2018 showed that approximately one-third of our chemotherapy was started during the night shift (after 19:00), and the average time from admission to start of chemotherapy was over 8 hours. There are currently no well-defined benchmarks for timeliness of chemotherapy initiation. The aim of this quality improvement initiative was to increase the percentage of patients who start chemotherapy in the bone marrow transplant unit before 19:00 from 65% to >80% by March 31, 2019. We identified barriers to timely initiation of chemotherapy through process mapping and analysis of failures. The primary barriers were late admissions (after 12:00 pm) and time from admission to preparation of chemotherapy. We addressed mechanisms to mitigate these barriers through Plan-Do-Study-Act testing. Interventions included providing families specific admission times and their rationales and process for notifying pharmacy of admissions immediately on arrival. We used standardized control charts to measure the impact of the interventions on change. We also monitored medication errors before and during the intervention. From September 2018 to March 2019 the percentage of patients who started preparative chemotherapy before 19:00 increased from 65% to 85%, the percentage of patients who were admitted after 12:00 remained similar before (31%) and after the interventions (33%), and the average time from admission to start of chemotherapy decreased from 8.6 hours (513 minutes) to 6.4 hours (382 minutes). Medication errors were similar before (n = 50) and after the interventions (n = 43). Using standardized processes, we demonstrated a substantial decrease in the percentage of HSCT patients starting their preparative regimen after 19:00 without a concurrent increase in errors. We believe these interventions and measurements can be used in all transplant centers and have the potential to influence patient safety and outcomes., (Copyright © 2019 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
43. Myosteatosis in adolescents and young adults treated for acute lymphoblastic leukemia.
- Author
-
Mueske NM, Mittelman SD, Wren TAL, Gilsanz V, and Orgel E
- Subjects
- Absorptiometry, Photon, Adipose Tissue diagnostic imaging, Adipose Tissue drug effects, Adolescent, Bone and Bones diagnostic imaging, Bone and Bones drug effects, Child, Female, Humans, Male, Muscle, Skeletal diagnostic imaging, Muscle, Skeletal drug effects, Muscle, Skeletal radiation effects, Muscular Diseases diagnosis, Muscular Diseases etiology, Muscular Diseases pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Remission Induction, Sarcopenia diagnosis, Sarcopenia etiology, Sarcopenia pathology, Tomography, X-Ray Computed, United States epidemiology, Young Adult, Adipose Tissue pathology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Muscle, Skeletal pathology, Muscular Diseases epidemiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Sarcopenia epidemiology
- Abstract
Myosteatosis refers to fat deposition within muscle and is linked to risk of cardiovascular disease and metabolic disorders. Though these comorbidities are common during and after therapy for acute lymphoblastic leukemia (ALL), little is known about tissue distribution, including myosteatosis, in this population. Using quantitative computed tomography, we assessed the impact of ALL therapy on bone, muscle, subcutaneous, and muscle-associated (MA) fat in 12 adolescents and young adults (AYA) treated for ALL as compared to a healthy control group without ALL ( n = 116). AYA had a marked loss of muscle with a gain in MA fat between ALL diagnosis and end of induction. These changes persisted throughout intensive therapy. Lower bone and muscle and higher MA fat were also observed during and after treatment in comparison to controls. Altered lower extremity tissue distribution, specifically myosteatosis and sarcopenia, may contribute to functional declines and increased risk of metabolic disorders and cardiovascular diseases.
- Published
- 2019
- Full Text
- View/download PDF
44. Mechanisms by Which Obesity Impacts Survival from Acute Lymphoblastic Leukemia.
- Author
-
Orgel E, Sea JL, and Mittelman SD
- Subjects
- Energy Metabolism, Hormones metabolism, Humans, Inflammation complications, Inflammation metabolism, Mortality, Obesity drug therapy, Obesity epidemiology, Obesity metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma etiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Prognosis, Public Health Surveillance, Obesity complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality
- Abstract
The prevalence of obesity has steadily risen over the past decades, even doubling in more than 70 countries. High levels of body fat (adiposity) and obesity are associated with endocrine and hormonal dysregulation, cardiovascular compromise, hepatic dysfunction, pancreatitis, changes in drug metabolism and clearance, inflammation, and metabolic stress. It is thus unsurprising that obesity can affect the development of and survival from a wide variety of malignancies. This review focuses on acute lymphoblastic leukemia, the most common malignancy in children, to explore the multiple mechanisms connecting acute lymphoblastic leukemia, obesity, and adipocytes, and the implications for leukemia therapy., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
45. Recommendations for ototoxicity surveillance for childhood, adolescent, and young adult cancer survivors: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group in collaboration with the PanCare Consortium.
- Author
-
Clemens E, van den Heuvel-Eibrink MM, Mulder RL, Kremer LCM, Hudson MM, Skinner R, Constine LS, Bass JK, Kuehni CE, Langer T, van Dalen EC, Bardi E, Bonne NX, Brock PR, Brooks B, Carleton B, Caron E, Chang KW, Johnston K, Knight K, Nathan PC, Orgel E, Prasad PK, Rottenberg J, Scheinemann K, de Vries ACH, Walwyn T, Weiss A, Am Zehnhoff-Dinnesen A, Cohn RJ, and Landier W
- Subjects
- Adolescent, Antineoplastic Agents therapeutic use, Child, Cranial Irradiation adverse effects, Evidence-Based Medicine, Humans, Neoplasms radiotherapy, Ototoxicity etiology, Ototoxicity therapy, Platinum Compounds adverse effects, Population Surveillance, Young Adult, Antineoplastic Agents adverse effects, Cancer Survivors statistics & numerical data, Delivery of Health Care standards, Neoplasms drug therapy, Ototoxicity diagnosis, Ototoxicity prevention & control
- Abstract
Childhood, adolescent, and young adult (CAYA) cancer survivors treated with platinum-based drugs, head or brain radiotherapy, or both have an increased risk of ototoxicity (hearing loss, tinnitus, or both). To ensure optimal care and reduce consequent problems-such as speech and language, social-emotional development, and learning difficulties-for these CAYA cancer survivors, clinical practice guidelines for monitoring ototoxicity are essential. The implementation of surveillance across clinical settings is hindered by differences in definitions of hearing loss, recommendations for surveillance modalities, and remediation. To address these deficiencies, the International Guideline Harmonization Group organised an international multidisciplinary panel, including 32 experts from ten countries, to evaluate the quality of evidence for ototoxicity following platinum-based chemotherapy and head or brain radiotherapy, and formulate and harmonise ototoxicity surveillance recommendations for CAYA cancer survivors., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
46. The genetic basis and cell of origin of mixed phenotype acute leukaemia.
- Author
-
Alexander TB, Gu Z, Iacobucci I, Dickerson K, Choi JK, Xu B, Payne-Turner D, Yoshihara H, Loh ML, Horan J, Buldini B, Basso G, Elitzur S, de Haas V, Zwaan CM, Yeoh A, Reinhardt D, Tomizawa D, Kiyokawa N, Lammens T, De Moerloose B, Catchpoole D, Hori H, Moorman A, Moore AS, Hrusak O, Meshinchi S, Orgel E, Devidas M, Borowitz M, Wood B, Heerema NA, Carrol A, Yang YL, Smith MA, Davidsen TM, Hermida LC, Gesuwan P, Marra MA, Ma Y, Mungall AJ, Moore RA, Jones SJM, Valentine M, Janke LJ, Rubnitz JE, Pui CH, Ding L, Liu Y, Zhang J, Nichols KE, Downing JR, Cao X, Shi L, Pounds S, Newman S, Pei D, Guidry Auvil JM, Gerhard DS, Hunger SP, Inaba H, and Mullighan CG
- Subjects
- Cell Lineage genetics, DNA Mutational Analysis, Female, Genetic Variation genetics, Genome, Human genetics, Genomics, Humans, Immunophenotyping, Leukemia, Biphenotypic, Acute classification, Male, Models, Genetic, Mutation genetics, Neoplastic Stem Cells immunology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Phenotype, Trans-Activators genetics, Leukemia, Biphenotypic, Acute genetics, Leukemia, Biphenotypic, Acute pathology
- Abstract
Mixed phenotype acute leukaemia (MPAL) is a high-risk subtype of leukaemia with myeloid and lymphoid features, limited genetic characterization, and a lack of consensus regarding appropriate therapy. Here we show that the two principal subtypes of MPAL, T/myeloid (T/M) and B/myeloid (B/M), are genetically distinct. Rearrangement of ZNF384 is common in B/M MPAL, and biallelic WT1 alterations are common in T/M MPAL, which shares genomic features with early T-cell precursor acute lymphoblastic leukaemia. We show that the intratumoral immunophenotypic heterogeneity characteristic of MPAL is independent of somatic genetic variation, that founding lesions arise in primitive haematopoietic progenitors, and that individual phenotypic subpopulations can reconstitute the immunophenotypic diversity in vivo. These findings indicate that the cell of origin and founding lesions, rather than an accumulation of distinct genomic alterations, prime tumour cells for lineage promiscuity. Moreover, these findings position MPAL in the spectrum of immature leukaemias and provide a genetically informed framework for future clinical trials of potential treatments for MPAL.
- Published
- 2018
- Full Text
- View/download PDF
47. Therapy for children and adults with mixed phenotype acute leukemia: a systematic review and meta-analysis.
- Author
-
Maruffi M, Sposto R, Oberley MJ, Kysh L, and Orgel E
- Subjects
- Adult, Animals, Child, Combined Modality Therapy, Disease Management, Humans, Leukemia, Biphenotypic, Acute mortality, Prognosis, Remission Induction, Treatment Outcome, Leukemia, Biphenotypic, Acute diagnosis, Leukemia, Biphenotypic, Acute therapy
- Abstract
The rarity of mixed-phenotype acute leukemia (MPAL) has resulted in diffuse literature consisting of small case series, thus precluding a consensus treatment approach. We conducted a meta-analysis and systematic review to investigate the association of treatment type (acute lymphoblastic leukemia [ALL], acute myeloid leukemia [AML], or "hybrid" regimens), disease response, and survival. We searched seven databases from inception through June 2017 without age or language restriction. Included studies reported sufficient treatment detail for de novo MPAL classified according to the well-established European Group for Immunological Characterization of Acute Leukemias (EGIL) or World Health Organization (WHO2008) criteria. Meta-analyses and multivariable analyses of a patient-level compiled case series were performed for the endpoints of complete remission (CR) and overall survival (OS). We identified 97 reports from 33 countries meeting criteria, resulting in 1,499 unique patients with data, of whom 1,351 had sufficient detail for quantitative analysis of the study endpoints. Using either definition of MPAL, meta-analyses revealed that AML induction was less likely to achieve a CR as compared to ALL regimens, (WHO2008 odds ratio [OR] = 0.33, 95% confidence interval [95% CI] 0.18-0.58; EGIL, OR = 0.18, 95% CI 0.08-0.40). Multivariable analysis of the patient-level data supported poorer efficacy for AML induction (versus ALL: OR = 0.45 95% CI 0.27-0.77). Meta-analyses similarly found better OS for those beginning with ALL versus AML therapy (WHO2008 OR = 0.45, 95% CI 0.26-0.77; EGIL, OR = 0.43, 95% CI 0.24-0.78), but multivariable analysis of patient-level data showed only those starting with hybrid therapy fared worse (hazard ratio [HR] = 2.11, 95% CI 1.30-3.43). MPAL definition did not impact trends within each endpoint and were similarly predictive of outcome. Using either definition of MPAL, ALL-therapy is associated with higher initial remission rates for MPAL and is at least equivalent to more intensive AML therapy for long-term survival. Prospective trials are needed to establish a uniform approach to this heterogeneous disease.
- Published
- 2018
- Full Text
- View/download PDF
48. Adipocyte metabolism of the chemotherapy daunorubicin.
- Author
-
Mittelman SD and Orgel E
- Abstract
Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest.
- Published
- 2018
- Full Text
- View/download PDF
49. Obesity and risk for venous thromboembolism from contemporary therapy for pediatric acute lymphoblastic leukemia.
- Author
-
Prasca S, Carmona R, Ji L, Ko RH, Bhojwani D, Rawlins YA, Mittelman SD, Young G, and Orgel E
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Female, Humans, Infant, Male, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Retrospective Studies, Venous Thromboembolism pathology, Young Adult, Obesity complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Venous Thromboembolism etiology
- Abstract
Introduction: Acute lymphoblastic leukemia (ALL) therapy confers risk for venous thromboembolism (VTE) and associated acute and long-term morbidity. Obesity increases VTE risk in the general population but its impact on ALL therapy-associated VTE is unknown., Methods: In a retrospective cohort of children treated for ALL between 2008 and 2016 (n = 294), we analyzed obesity at diagnosis (body mass index [BMI] ≥95%) and subsequent development of VTE. A subset participated in two concurrent prospective ALL trials studying body composition via dual-energy X-ray absorptiometry (DXA) (n = 35) and hypercoagulability via thromboelastography (TEG) (n = 46). Secondary analyses explored whether precise measurement of body fat and/or global hemostasis ex vivo by TEG could further delineate VTE risk in the obese., Results: Overall, we found 27/294 (9.2%) patients developed symptomatic VTE during therapy, 19/27 (70%) occurred during Induction. Study-defined "serious" VTE developed in 4/294 (1.4%) of patients. Obesity but not overweight was strongly predictive of symptomatic VTE (obesity odds ratio = 3.8, 95% confidence interval 1.5-9.6, p = 0.008). In the DXA subset, only 2/35 patients developed symptomatic VTE. However, within those prospectively screened during Induction, 30% (14/46) developed VTE; eight (17%) of these were asymptomatic and found only via screening., Conclusions: In this pediatric ALL cohort, obesity conferred more than a three-fold increased risk for symptomatic VTE. In a subgroup of patients who underwent active screening, up to a third were noted to have VTE (symptomatic and asymptomatic). TEG did not predict VTE. Additional studies are necessary to validate these findings and to further refine a risk-stratified approach to thrombo-prevention during ALL therapy., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
50. Predictors of hepatotoxicity and pancreatitis in children and adolescents with acute lymphoblastic leukemia treated according to contemporary regimens.
- Author
-
Denton CC, Rawlins YA, Oberley MJ, Bhojwani D, and Orgel E
- Subjects
- Adolescent, Age Factors, Chemical and Drug Induced Liver Injury epidemiology, Child, Child, Preschool, Cohort Studies, Disease-Free Survival, Female, Humans, Incidence, Infant, Male, Obesity epidemiology, Pancreatitis epidemiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Retrospective Studies, Risk Factors, Young Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Chemical and Drug Induced Liver Injury etiology, Pancreatitis chemically induced, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Background: Hepatotoxicity and pancreatitis are common treatment-related toxicities (TRTs) during contemporary treatment regimens for acute lymphoblastic leukemia (ALL). Limited detailed data from Children's Oncology Group (COG) regimens has been previously reported to enable identification of patient and treatment risk factors for these toxicities and their impact on outcomes., Procedure: We analyzed a retrospective pediatric ALL cohort treated at a single institution according to COG regimens from 2008 to 2015. The primary endpoint was cumulative incidence of study-defined "severe" hepatotoxicity (Common Terminology Criteria for Adverse Events [CTCAE] Grade ≥ 4 transaminitis or Grade ≥ 3 hyperbilirubinemia) and clinically significant pancreatitis (any grade). Pancreatitis was additionally classified using the Ponte di Legno (PdL) toxicity criteria. Secondary endpoints were chemotherapy interruptions, early disease response (end of induction [EOI] minimal residual disease [MRD]), and event-free survival (EFS)., Results: We identified 262 patients, of whom 71 (27%) and 28 (11%) developed hepatotoxicity and pancreatitis, respectively. Three cases of pancreatitis did not fulfill PdL criteria despite otherwise consistent presentations. Both TRTs occurred throughout therapy, but approximately 25% of hepatotoxicity (18/71) and pancreatitis (8/28) occurred during induction alone. Both obesity and age (≥10 years) were identified as predictors of hepatotoxicity (subdistribution hazard ratio [SHR] obesity = 1.75, 95% confidence interval [95% CI] 1.04-2.96; SHR age ≥10 = 1.9, 95% CI 1.19-3.10) and pancreatitis (SHR obesity = 2.18, 95% CI 1.01-4.67; SHR age ≥ 10 = 2.76, 95% CI 1.19-6.39, P = 0.018). Dose interruptions were common but neither toxicity influenced EOI MRD nor EFS., Conclusions: Obese and/or older children are particularly at risk for hepatotoxicity and pancreatitis, and may benefit from toxicity surveillance and chemoprotective strategies to prevent or mitigate associated morbidity., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.