51
|
Kuo MT, Chen HHW, Feun LG, Savaraj N. Targeting the Proline-Glutamine-Asparagine-Arginine Metabolic Axis in Amino Acid Starvation Cancer Therapy. Pharmaceuticals (Basel) 2021; 14:ph14010072. [PMID: 33477430 PMCID: PMC7830038 DOI: 10.3390/ph14010072] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/22/2022] Open
Abstract
Proline, glutamine, asparagine, and arginine are conditionally non-essential amino acids that can be produced in our body. However, they are essential for the growth of highly proliferative cells such as cancers. Many cancers express reduced levels of these amino acids and thus require import from the environment. Meanwhile, the biosynthesis of these amino acids is inter-connected but can be intervened individually through the inhibition of key enzymes of the biosynthesis of these amino acids, resulting in amino acid starvation and cell death. Amino acid starvation strategies have been in various stages of clinical applications. Targeting asparagine using asparaginase has been approved for treating acute lymphoblastic leukemia. Targeting glutamine and arginine starvations are in various stages of clinical trials, and targeting proline starvation is in preclinical development. The most important obstacle of these therapies is drug resistance, which is mostly due to reactivation of the key enzymes involved in biosynthesis of the targeted amino acids and reprogramming of compensatory survival pathways via transcriptional, epigenetic, and post-translational mechanisms. Here, we review the interactive regulatory mechanisms that control cellular levels of these amino acids for amino acid starvation therapy and how drug resistance is evolved underlying treatment failure.
Collapse
Affiliation(s)
- Macus Tien Kuo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| | - Helen H. W. Chen
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan;
| | - Lynn G. Feun
- Department of Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Niramol Savaraj
- Division of Hematology and Oncology, Miami Veterans Affairs Heaithcare System, Miami, FL 33136, USA;
| |
Collapse
|
52
|
Burke PW, Hoelzer D, Park JH, Schmiegelow K, Douer D. Managing toxicities with asparaginase-based therapies in adult ALL: summary of an ESMO Open-Cancer Horizons roundtable discussion. ESMO Open 2020; 5:e000858. [PMID: 33037033 PMCID: PMC7549445 DOI: 10.1136/esmoopen-2020-000858] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023] Open
Abstract
With recent prospective clinical trials that used paediatric regimens with multiple doses of pegylated form of asparaginase (PEG asparaginase) in adults reporting significantly improved survival compared with historical data with regimens that used less asparaginase, PEG asparaginase is increasingly being used in the treatment of adult acute lymphoblastic leukaemia (ALL). However, administering asparaginase still comes with its challenges, especially in adult patients. Therefore, it is important to understand how to manage its toxicities properly. An expert group met in November 2019 in London to discuss recent data of paediatric as well as adult studies using paediatric regimens with regard to the best management of several key toxicities that can occur in adults treated with asparaginase including hepatotoxicity, pancreatitis, hypertriglyceridaemia, thrombosis and hypersensitivity. Several recommendations were made for each one of these toxicities, with the goal of safe administration of the drug and to educate clinicians when the drug can be continued despite side effects.
Collapse
Affiliation(s)
- Patrick W Burke
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | - Dieter Hoelzer
- Internal Medicine, Onkologikum Frankfurt, Frankfurt, Germany
| | - Jae H Park
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Juliane Marie Center, Rigshospitalet University Hospital, and Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dan Douer
- Jane Anne Nohle Division of Hematology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
53
|
Hayashi RJ, Winter SS, Dunsmore KP, Devidas M, Chen Z, Wood BL, Hermiston ML, Teachey DT, Perkins SL, Miles RR, Raetz EA, Loh ML, Winick NJ, Carroll WL, Hunger SP, Lim MS, Gross TG, Bollard CM. Successful Outcomes of Newly Diagnosed T Lymphoblastic Lymphoma: Results From Children's Oncology Group AALL0434. J Clin Oncol 2020; 38:3062-3070. [PMID: 32552472 PMCID: PMC7479761 DOI: 10.1200/jco.20.00531] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The Children's Oncology Group (COG) protocol AALL0434 evaluated the safety and efficacy of multi-agent chemotherapy with Capizzi-based methotrexate/pegaspargase (C-MTX) in patients with newly diagnosed pediatric T-cell lymphoblastic lymphoma (T-LL) and gained preliminary data using nelarabine in high-risk patients. PATIENTS AND METHODS The trial enrolled 299 patients, age 1-31 years. High-risk (HR) patients had ≥ 1% minimal detectable disease (MDD) in the bone marrow at diagnosis or received prior steroid treatment. Induction failure was defined as failure to achieve a partial response (PR) by the end of the 4-week induction. All patients received the augmented Berlin-Frankfurt-Muenster (ABFM) C-MTX regimen. HR patients were randomly assigned to receive or not receive 6 5-day courses of nelarabine incorporated into ABFM. Patients with induction failure were nonrandomly assigned to ABFM C-MTX plus nelarabine. No patients received prophylactic cranial radiation; however, patients with CNS3 disease (CSF WBC ≥ 5/μL with blasts or cranial nerve palsies, brain/eye involvement, or hypothalamic syndrome) were ineligible. RESULTS At end-induction, 98.8% of evaluable participants had at least a PR. The 4-year event-free survival (EFS) and overall survival (OS) were 84.7% ± 2.3% and 89.0% ± 2.0%. The 4-year disease-free survival (DFS) from end-induction was 85.9% ± 2.6%. There was no difference in DFS observed between the HR and standard-risk groups (P = .29) or by treatment regimen (P = .55). Disease stage, tumor response, and MDD at diagnosis did not demonstrate thresholds that resulted in differences in EFS. Nelarabine did not show an advantage for HR patients. CNS relapse occurred in only 4 patients. CONCLUSION COG AALL0434 produced excellent outcomes in one of the largest trials ever conducted for patients with newly diagnosed T-LL. The COG ABFM regimen with C-MTX provided excellent EFS and OS without cranial radiation.
Collapse
Affiliation(s)
- Robert J. Hayashi
- Pediatric Hematology/Oncology, Washington School of Medicine, St Louis Children’s Hospital, St Louis, MO
| | - Stuart S. Winter
- Children’s Minnesota Cancer and Blood Disorders Program, Minneapolis, MN
| | | | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN
| | - Zhiguo Chen
- Department of Biostatistics, College of Medicine and College of Public Health and Health Professions, University of Florida, Gainesville, FL
| | - Brent L. Wood
- Laboratory Medicine, Seattle Children’s Hospital, Seattle, WA
| | - Michelle L. Hermiston
- Department of Pediatrics, UCSF Benioff Children’s Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - David T. Teachey
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sherrie L. Perkins
- Department of Pathology, University of Utah Health Sciences Center, ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, UT
| | - Rodney R. Miles
- Department of Pathology, University of Utah Health Sciences Center, ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, UT
| | - Elizabeth A. Raetz
- Department of Pediatrics and Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Mignon L. Loh
- Department of Pediatrics, UCSF Benioff Children’s Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Naomi J. Winick
- Pediatric Hematology/Oncology, University of Texas Southwestern/Simmons Cancer Center, Dallas, TX
| | - William L. Carroll
- Department of Pediatrics and Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Stephen P. Hunger
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Megan S. Lim
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Thomas G. Gross
- National Cancer Institute Center for Global Health, Rockville, MD
| | - Catherine M. Bollard
- Division of Blood and Marrow Transplantation, Children’s National Health System, Washington, DC
| |
Collapse
|
54
|
Patel AA, Thomas J, Rojek AE, Stock W. Biology and Treatment Paradigms in T Cell Acute Lymphoblastic Leukemia in Older Adolescents and Adults. Curr Treat Options Oncol 2020; 21:57. [PMID: 32468488 DOI: 10.1007/s11864-020-00757-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OPINION STATEMENT T cell acute lymphoblastic leukemia (T-ALL) occurs in approximately 25-30% of adult ALL diagnoses. Historically, B cell ALL (B-ALL) and T-ALL have been treated in the same fashion despite differences in the biology of disease. Outcomes in the adolescent/young adult (AYA) population have improved significantly with the utilization of pediatric-based regimens. In addition, there may now be a role for the addition of nelarabine to frontline treatment in the AYA population. In older adults, choices in which regimen to pursue should account for the potential toxicities associated with pediatric-based regimens. Measurable residual disease (MRD) has taken on increasing prognostic value in T-ALL and may help to identify which patients should receive an allogeneic stem cell transplant. T cell lymphoblastic lymphoma (T-LBL) has traditionally been treated similarly to T-ALL, but additional management questions must be considered. Mediastinal irradiation does not seem to clearly improve outcomes, and there is considerable heterogeneity in the central nervous system (CNS) prophylaxis strategy used in prospective trials. CNS prophylaxis in AYA patients with T-ALL, on the other hand, can be safely achieved with intrathecal chemotherapy alone. Prospective data regarding CNS prophylaxis strategies in older adults are currently not available. Nelarabine-based regimens currently remain the standard in relapsed/refractory T-ALL; however, novel therapies targeting molecular aberrations in T-ALL are actively being investigated.
Collapse
Affiliation(s)
- Anand A Patel
- Department of Medicine, Section of Hematology-Oncology, The University of Chicago Medicine, 5841 S. Maryland Avenue, MC 2115, Chicago, IL, 60637, USA
| | - Joseph Thomas
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Wendy Stock
- Department of Medicine, Section of Hematology-Oncology, The University of Chicago Medicine, 5841 S. Maryland Avenue, MC 2115, Chicago, IL, 60637, USA.
| |
Collapse
|
55
|
Gupta S, Wang C, Raetz EA, Schore R, Salzer WL, Larsen EC, Maloney KW, Mattano LA, Carroll WL, Winick NJ, Hunger SP, Loh ML, Devidas M. Impact of Asparaginase Discontinuation on Outcome in Childhood Acute Lymphoblastic Leukemia: A Report From the Children's Oncology Group. J Clin Oncol 2020; 38:1897-1905. [PMID: 32275469 DOI: 10.1200/jco.19.03024] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Asparaginase (ASNase) is an important component of acute lymphoblastic leukemia (ALL) treatment, but is often discontinued because of toxicity. Erwinia chrysanthemi ASNase (Erwinia) substitution was approved in 2011 for allergic reactions. Erwinia has, however, been intermittently unavailable because of drug supply issues. The impact of Erwinia substitution or complete ASNase discontinuation is unknown. METHODS Patients aged 1-30.99 years in frontline Children's Oncology Group trials for B-cell acute lymphoblastic leukemia between 2004 and 2011 (National Cancer Institute [NCI] standard risk [SR]: AALL0331; NCI high risk: AALL0232) were included. The number of prescribed pegaspargase (PEG-ASNase) doses varied by trial and strata. Maintenance therapy did not contain ASNase. Landmark analyses at maintenance compared disease-free survival (DFS) among those receiving all prescribed PEG-ASNase doses versus switching to Erwinia but receiving all doses versus not receiving all ASNase doses. RESULTS We included 5,195 AALL0331 and 3,001 AALL0232 patients. The cumulative incidence of PEG-ASNase discontinuation was 12.2% ± 4.6% in AALL0331 and 25.4% ± 0.8% in AALL0232. In multivariable analyses, NCI high-risk patients not receiving all prescribed ASNase doses had inferior DFS (hazard ratio [HR], 1.5; 95% CI, 1.2 to 1.9; P = .002) compared with those receiving all prescribed PEG-ASNase doses. Patients with Erwinia substitution who completed subsequent courses were not at increased risk (HR, 1.1; 95% CI, 0.7 to 1.6; P = .69). NCI SR patients who discontinued ASNase were not at elevated risk (HR, 1.2; 95% CI, 0.9 to 1.6; P = .23), except when restricted to those with slow early response, who were prescribed more ASNase because of therapy intensification (HR, 1.7; 95% CI, 1.1 to 2.7; P = .03). CONCLUSION Discontinuation of ASNase doses is associated with inferior DFS in higher-risk patients. Our results illustrate the severe consequences of Erwinia shortages.
Collapse
Affiliation(s)
- Sumit Gupta
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cindy Wang
- Department of Biostatistics, University of Florida, Gainesville, FL
| | | | | | - Wanda L Salzer
- US Army Medical Research and Materiel Command, Fort Detrick, MD
| | - Eric C Larsen
- Department of Pediatrics, Maine Children's Cancer Program, Scarborough, ME
| | | | | | - William L Carroll
- Department of Pediatrics and Perlmutter Cancer Center, New York University Langone Health, New York, NY
| | - Naomi J Winick
- University of Texas Southwestern/Simmons Cancer Center, Dallas, TX
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mignon L Loh
- Department of Pediatrics, UCSF Benoiff Childen's Hospital and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
56
|
Derman BA, Streck M, Wynne J, Christ TN, Curran E, Stock W, Knoebel RW. Efficacy and toxicity of reduced vs. standard dose pegylated asparaginase in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia. Leuk Lymphoma 2020; 61:614-622. [PMID: 31680584 PMCID: PMC7028458 DOI: 10.1080/10428194.2019.1680839] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/21/2019] [Accepted: 10/10/2019] [Indexed: 01/01/2023]
Abstract
Incorporation of asparaginase (ASNase) and pegylated asparaginase (PEG-ASP) into pediatric-inspired regimens for adults with acute lymphoblastic leukemia (ALL) has led to improved treatment outcomes albeit with increased toxicities. This study compared the efficacy and safety of the Children's Oncology Group standard PEG-ASP (SD) dosing (>1000, median 2500 IU/m2/dose) in adult Philadelphia chromosome-negative ALL patients receiving multiagent chemotherapy vs reduced dose PEG-ASP (RED) (≤1000, median 500 IU/m2/dose) during induction. 51 patients were included, 26 in RED and 25 in SD (median age 49 vs 37 years, p = .027). Median day 7 ASNase activity level for RED was 0.16 IU/mL. All 11 patients who received PEG-ASP 1000 IU/m2 and 9/11 patients who received 500 IU/m2 achieved an ASNase level ≥0.1 IU/mL. Patients receiving RED experienced fewer total grade 3/4 toxicities during induction compared to SD (p = .02) while still attaining therapeutic ASNase levels. RED permits safer ASNase use in adults with ALL and should be tested in a larger cohort prospectively.
Collapse
Affiliation(s)
| | - Mitchell Streck
- Simon Cancer Center, Indiana University Health, Indianapolis, IN
| | - Joseph Wynne
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Trevor N Christ
- Department of Pharmacy, University of Chicago, Chicago, IL
- Rush University Medical Center, Chicago, IL
| | - Emily Curran
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Wendy Stock
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | | |
Collapse
|
57
|
Mondelaers V, Ferster A, Uyttebroeck A, Brichard B, van der Werff Ten Bosch J, Norga K, Francotte N, Piette C, Vandemeulebroecke K, Verbeke C, Schmidt S, Benoit Y, Lammens T, De Moerloose B. Prospective, real-time monitoring of pegylated Escherichia coli and Erwinia asparaginase therapy in childhood acute lymphoblastic leukaemia and non-Hodgkin lymphoma in Belgium. Br J Haematol 2020; 190:105-114. [PMID: 32057100 DOI: 10.1111/bjh.16495] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/24/2019] [Indexed: 11/25/2022]
Abstract
Asparaginase (ASNase) is an important anti-leukaemic drug in the treatment of childhood acute lymphoblastic leukaemia (ALL) and non-Hodgkin lymphoma (NHL). A substantial proportion of patients develop hypersensitivity reactions with anti-ASNase neutralising antibodies, resulting in allergic reactions or silent inactivation (SI), and characterised by inactivation and rapid clearance of ASNase. We report results of a prospective, real-time therapeutic drug monitoring of pegylated Escherichia coli (PEG-)ASNase and Erwinia ASNase in children treated for ALL and NHL in Belgium. Erwinia ASNase was given as second-line after hypersensitivity to PEG-ASNase. In total, 286 children were enrolled in the PEG-ASNase cohort. Allergy was seen in 11·2% and SI in 5·2% of patients. Of the 42 patients treated with Erwinia ASNase, 7·1% experienced allergy and 2·4% SI. The median trough PEG-ASNase activity was high in all patients without hypersensitivity. After Erwinia administration significantly more day 3 samples had activities <100 IU/l (62·5% vs. 10% at day 2 (D2)). The median D2 activity was significantly higher for intramuscular (IM; 347 IU/l) than for intravenous Erwinia administrations (159 IU/l). This prospective, multicentre study shows that monitoring of ASNase activity during treatment of children with ALL and NHL is feasible and informative. Treatment with Erwinia ASNase warrants close monitoring and optimally adherence to a 2-day interval of IM administrations.
Collapse
Affiliation(s)
- Veerle Mondelaers
- Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital and Cancer Research Institute Ghent, Ghent, Belgium
| | - Alina Ferster
- Pediatric Hematology-Oncology, Hôpital Universitaire des Enfants Reine Fabiola (HUDERF-UKZKF), Brussels, Belgium
| | - Anne Uyttebroeck
- Pediatric Hematology-Oncology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Bénédicte Brichard
- Pediatric Hematology-Oncology, Cliniques Universitaires Saint-Luc (UCL), Brussels, Belgium
| | | | - Koenraad Norga
- Pediatric Hematology-Oncology, University Hospital Antwerp, Antwerp, Belgium
| | - Nadine Francotte
- Department of Pediatric Oncology, CHC- Hospital of Hope, Montegnée, Belgium
| | - Caroline Piette
- Department of Pediatric Oncology, CHR Citadelle, Liège, Belgium
| | - Katrien Vandemeulebroecke
- Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Charlotte Verbeke
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Susanne Schmidt
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Yves Benoit
- Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital and Cancer Research Institute Ghent, Ghent, Belgium
| | - Tim Lammens
- Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital and Cancer Research Institute Ghent, Ghent, Belgium
| | - Barbara De Moerloose
- Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital and Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
58
|
Kloos RQH, Pieters R, Jumelet FMV, de Groot-Kruseman HA, van den Bos C, van der Sluis IM. Individualized Asparaginase Dosing in Childhood Acute Lymphoblastic Leukemia. J Clin Oncol 2020; 38:715-724. [PMID: 31922920 DOI: 10.1200/jco.19.02292] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
PURPOSE In the DCOG ALL-11 protocol, polyethylene glycol-conjugated Escherichia coli asparaginase (PEGasparaginase) and Erwinia asparaginase treatment of pediatric acute lymphoblastic leukemia are individualized with therapeutic drug monitoring (TDM). The efficacy of TDM and its effect on asparaginase-associated toxicity are reported. PATIENTS AND METHODS After induction with 3 fixed intravenous doses of 1,500 IU/m2 PEGasparaginase, medium-risk patients (n = 243) received 14 individualized doses that targeted trough levels of 100-250 IU/L, standard-risk patients (n = 108) received 1 individualized dose, and high-risk patients (n = 18) received 2-5 fixed administrations (1,500 IU/m2). After a neutralizing hypersensitivity reaction, patients were started with 20,000 IU/m2 Erwinia asparaginase 3 times per week, and l-asparagine was measured to monitor asparaginase efficacy. Several asparaginase-associated toxicities were studied. RESULTS The final median PEGasparaginase dose was lowered to 450 IU/m2. Overall, 97% of all trough levels of nonallergic patients were > 100 IU/L. Asparagine was < 0.5 μM in 96% and 67% of the PEGasparaginase and Erwinia asparaginase levels > 100 IU/L, respectively. Ten percent developed a neutralizing hypersensitivity reaction to PEGasparaginase, of which 40% were silent inactivations. The cumulative incidence of grade 3-4 pancreatitis, central neurotoxicity, and thromboses was 12%, 4%, and 6%, respectively, and not associated with asparaginase activity levels. During medium-risk intensification, 50% had increased ALT and 3% hyperbilirubinemia (both grade 3/4 and correlated with asparaginase activity levels), and 37% had grade 3/4 hypertriglyceridemia. Hypertriglyceridemia occurred less in intensification compared with ALL-10 (37% v 47%), which is similar to ALL-11 but with higher asparaginase levels during intensification. CONCLUSION TDM of asparaginase results in a significant reduction of the PEGasparaginase dose with adequate asparaginase activity levels and sufficient asparagine depletion. In addition, with TDM, silent inactivation and allergic-like reactions were identified. However, the effect of reduced asparaginase activity levels on toxicity is limited.
Collapse
Affiliation(s)
- Robin Q H Kloos
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Florine M V Jumelet
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Hester A de Groot-Kruseman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Dutch Childhood Oncology Group, Utrecht, the Netherlands
| | - Cor van den Bos
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Pediatric Oncology and Hematology, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Inge M van der Sluis
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
59
|
Kloos R, van der Sluis IM, Mastrobattista E, Hennink W, Pieters R, Verhoef JJ. Acute lymphoblastic leukaemia patients treated with PEGasparaginase develop antibodies to PEG and the succinate linker. Br J Haematol 2019; 189:442-451. [PMID: 31883112 DOI: 10.1111/bjh.16254] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/29/2019] [Indexed: 12/30/2022]
Abstract
Polyethylene glycol (PEG) conjugated asparaginase (PEGasparaginase) is essential for treatment of paediatric acute lymphoblastic leukaemia. We developed an assay identifying antibodies against the PEG-moiety, the linker and the drug itself in patients experiencing hypersensitivity reactions to PEGasparaginase. Eighteen patients treated according to the DCOG ALL-11 protocol, with a neutralizing hypersensitivity reaction to PEGasparaginase to the first PEGasparaginase doses in induction (12 patients) or during intensification after interruption of several months (6 patients) were included. ELISA was used to measure antibodies, coating with the succinimidyl succinate linker conjugated to BSA, PEGfilgrastim and Escherichia coli asparaginase, and using hydrolysed PEGasparaginase and mPEG5,000 for competition. Anti-PEG antibodies were detected in all patients (IgG 100%; IgM 67%) of whom 39% had anti-PEG antibodies exclusively. Pre-existing anti-PEG antibodies were also detected in patients who not previously received a PEGylated therapeutic (58% IgG; 21% IgM). Antibodies against the SS-linker were predominantly detected during induction (50% IgG; 42% IgM). Anti-asparaginase antibodies were detected in only 11% during induction but 94% during intensification. In conclusion, anti-PEG and anti-SS-linker antibodies predominantly play a role in the immunogenic response to PEGasparaginase during induction. Thus, switching to native E. coli asparaginase would be an option for adequate asparaginase treatment.
Collapse
Affiliation(s)
- Robin Kloos
- Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | | | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Wim Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jan-Jaap Verhoef
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
60
|
Hernández-Marqués C, Andión M, Perez-Somarriba M, Madero L, Lassaletta A. Can monitoring asparaginase activity help us to manage toxicity in pediatric acute lymphoblastic leukemia? Leuk Lymphoma 2019; 61:990-992. [PMID: 31749392 DOI: 10.1080/10428194.2019.1691191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Carmen Hernández-Marqués
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Maitane Andión
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Marta Perez-Somarriba
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Luis Madero
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Alvaro Lassaletta
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| |
Collapse
|
61
|
Cecconello DK, Magalhães MRD, Werlang ICR, Lee MLDM, Michalowski MB, Daudt LE. Asparaginase: an old drug with new questions. Hematol Transfus Cell Ther 2019; 42:275-282. [PMID: 31801703 PMCID: PMC7417439 DOI: 10.1016/j.htct.2019.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/19/2019] [Accepted: 07/20/2019] [Indexed: 02/08/2023] Open
Abstract
The long-term outcome of acute lymphoblastic leukemia has improved dramatically due to the development of more effective treatment strategies. L-asparaginase (ASNase) is one of the main drugs used and causes death of leukemic cells by systematically depleting the non-essential amino acid asparagine. Three main types of ASNase have been used so far: native ASNase derived from Escherichia coli, an enzyme isolated from Erwinia chrysanthemi and a pegylated form of the native E. coli ASNase, the ASNase PEG. Hypersensitivity reactions are the main complication related to this drug. Although clinical allergies may be important, a major concern is that antibodies produced in response to ASNase may cause rapid inactivation of ASNase, leading to a worse prognosis. This reaction is commonly referred to as "silent hypersensitivity" or "silent inactivation". We are able to analyze hypersensitivity and inactivation processes by the measurement of the ASNase activity. The ability to individualize the ASNase therapy in patients, adjusting the dose or switching patients with silent inactivation to an alternate ASNase preparation may help improve outcomes in those patients. This review article aims to describe the pathophysiology of the inactivation process, how to diagnose it and finally how to manage it.
Collapse
Affiliation(s)
- Daiane Keller Cecconello
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, RS, Brazil
| | | | - Isabel Cristina Ribas Werlang
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, RS, Brazil
| | | | - Mariana Bohns Michalowski
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, RS, Brazil.
| | - Liane Esteves Daudt
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, RS, Brazil
| |
Collapse
|
62
|
Quist-Paulsen P, Toft N, Heyman M, Abrahamsson J, Griškevičius L, Hallböök H, Jónsson ÓG, Palk K, Vaitkeviciene G, Vettenranta K, Åsberg A, Frandsen TL, Opdahl S, Marquart HV, Siitonen S, Osnes LT, Hultdin M, Overgaard UM, Wartiovaara-Kautto U, Schmiegelow K. T-cell acute lymphoblastic leukemia in patients 1–45 years treated with the pediatric NOPHO ALL2008 protocol. Leukemia 2019; 34:347-357. [DOI: 10.1038/s41375-019-0598-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/16/2019] [Accepted: 09/30/2019] [Indexed: 01/16/2023]
|
63
|
Siegel SE, Stock W, Johnson RH, Advani A, Muffly L, Douer D, Reed D, Lewis M, Freyer DR, Shah B, Luger S, Hayes-Lattin B, Jaboin JJ, Coccia PF, DeAngelo DJ, Seibel N, Bleyer A. Pediatric-Inspired Treatment Regimens for Adolescents and Young Adults With Philadelphia Chromosome-Negative Acute Lymphoblastic Leukemia: A Review. JAMA Oncol 2019; 4:725-734. [PMID: 29450465 DOI: 10.1001/jamaoncol.2017.5305] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Importance The incidence of acute lymphoblastic leukemia (ALL) and lymphoblastic lymphoma (LBL) in adolescent and young adult (AYA) patients (age range, 15-39 years) in the United States is increasing at a greater rate than in younger or older persons. Their optimal treatment has been increasingly debated as pediatric regimens have become more widely used in the age group. This review compares the basic features of pediatric and adult chemotherapy regimens for ALL and LBL, recognizes and describes the challenges of the pediatric regimen, and suggests strategies to facilitate its adoption for AYAs with ALL and LBL. Observations All but 2 of 25 published comparisons of outcomes with pediatric and adult regimens for ALL and LBL in AYAs and 1 meta-analysis favor the pediatric regimen. After more than a half-century of clinical trials of the pediatric regimens, including at least 160 phase 3 trials in the United States, the pediatric regimens have become far more complex than most adult regimens. Asparaginase, a critical component of the pediatric regimens, is more difficult to administer to AYAs (and older patients) but nonetheless has a favorable benefit to toxicity ratio for AYAs. A dramatic reduction in outcome of ALL and LBL during the AYA years (the "survival cliff") is coincident with similar reductions in proportions of AYAs referred to academic centers and enrolled on clinical trials (the "accrual cliff" and "referral cliff"). Conclusions and Relevance The accumulating data increasingly support treating AYAs with ALL and LBL with a pediatric-inspired regimen or an approved institutional or national clinical trial tailored for this patient group. A need to develop clinical trials specifically for AYAs and to encourage their participation is paramount, with a goal to improve both the quantity and quality of survival.
Collapse
Affiliation(s)
| | - Wendy Stock
- Alliance for Clinical Trials in Oncology (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Section of Hematology/Oncology, University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| | - Rebecca H Johnson
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Children's Oncology Group (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,National Clinical Oncology Research Program (All in the National Cancer Institute National Clinical Trials Network).,Pediatric Hematology/Oncology, Mary Bridge Children's Hospital and Health Center and Tacoma General Hospital, Tacoma, Washington
| | - Anjali Advani
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Hematology/Oncology, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Lori Muffly
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Blood and Marrow Transplantation, Department of Medicine, Stanford University, Palo Alto, California
| | - Dan Douer
- ECOG-ACRIN Cancer Research Group (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Keck Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles
| | - Damon Reed
- National Pediatric Cancer Foundation, Tampa, Florida.,Moffitt Cancer Center, Tampa, Florida
| | - Mark Lewis
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Hematology/Oncology, Intermountain Healthcare, Salt Lake City, Utah
| | - David R Freyer
- Children's Oncology Group (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Keck Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles
| | - Bijal Shah
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Moffitt Cancer Center, Tampa, Florida.,National Comprehensive Cancer Network
| | - Selina Luger
- ECOG-ACRIN Cancer Research Group (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Brandon Hayes-Lattin
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Department of Radiation Medicine, Oregon Health and Science University, Portland
| | - Jerry J Jaboin
- Department of Radiation Medicine, Oregon Health and Science University, Portland.,NRG Oncology (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group), National Cancer Institute, Bethesda, Maryland
| | - Peter F Coccia
- Children's Oncology Group (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,National Comprehensive Cancer Network.,Department of Pediatrics, University of Nebraska Medical Center, Omaha
| | - Daniel J DeAngelo
- Alliance for Clinical Trials in Oncology (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nita Seibel
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Archie Bleyer
- SWOG (National Cancer Institute-Sponsored National Clinical Trials Network Cooperative Group).,Department of Radiation Medicine, Oregon Health and Science University, Portland
| |
Collapse
|
64
|
Albertsen BK, Grell K, Abrahamsson J, Lund B, Vettenranta K, Jónsson ÓG, Frandsen TL, Wolthers BO, Heyman M, Schmiegelow K. Intermittent Versus Continuous PEG-Asparaginase to Reduce Asparaginase-Associated Toxicities: A NOPHO ALL2008 Randomized Study. J Clin Oncol 2019; 37:1638-1646. [DOI: 10.1200/jco.18.01877] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Asparaginase is an essential drug in childhood acute lymphoblastic leukemia (ALL) therapy and is frequently given for months to obtain continuous asparagine depletion. We randomly assigned patients to continuous versus intermittent pegylated-asparaginase (PEG-asp) treatment, hypothesizing there would be decreased toxicity with unchanged efficacy. METHODS Children (median age, 4.2 years) treated for non–high-risk ALL according to the Nordic Society for Pediatric Hematology and Oncology ALL2008 protocol received five intramuscular PEG-asp injections (1,000 IU/m2) every two weeks and were then randomly assigned to additional three doses (6-week intervals [experimental arm], n = 309) versus 10 doses (2-week intervals [standard arm], n = 316). The primary end point was noninferior (6% margin) disease-free survival. Toxicity reduction was a secondary end point. Occurrence of asparaginase-associated hypersensitivity, pancreatitis, osteonecrosis, and thromboembolism were prospectively registered. RESULTS After a median follow-up of 4.1 years, the 5-year disease-free survival was 92.2% (95% CI, 88.6 to 95.8) and 90.8% (95% CI, 87.0 to 94.6) in the experimental and standard arms, respectively. The 3-year cumulative incidence of any first asparaginase-associated toxicity (hypersensitivity [n = 13]; osteonecrosis [n = 29]; pancreatitis [n = 24]; thromboembolism [n = 17]) was 9.3% in the experimental arm and 18.1% in the standard arm ( P = .001). Asparaginase-associated toxicity reduction was confirmed in sex- and risk-group–adjusted Cox regression analysis stratified by age (≥ 10 and < 10 years; hazard ratio, 0.48; P = .001). The experimental arm had the lowest incidences of all four toxicities, reaching significance for pancreatitis (6-month risk, 5.8% v 1.3%; P = .002). CONCLUSION The excellent cure rates and reduced toxicity risk support the use of intermittent PEG-asp therapy after the first 10 weeks in future childhood ALL trials that apply prolonged PEG-asp therapy.
Collapse
Affiliation(s)
| | - Kathrine Grell
- University of Copenhagen, Copenhagen, Denmark,
- Rigshospitalet, Copenhagen, Denmark
| | | | - Bendik Lund
- Trondheim University Hospital, Trondheim, Norway
| | | | | | | | | | - Mats Heyman
- Karolinska University Hospital, Stockholm, Sweden
| | - Kjeld Schmiegelow
- University of Copenhagen, Copenhagen, Denmark,
- Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
65
|
Merlen C, Bonnefoy A, Afeich C, Théorêt Y, Laverdière C, Leclerc JM, Rivard GE. Antithrombin and fibrinogen levels as predictors for plasma L-asparaginase activity in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2019; 66:e27729. [PMID: 30938058 DOI: 10.1002/pbc.27729] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/08/2019] [Accepted: 02/27/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND L-asparaginase is a cornerstone treatment for children with acute lymphoblastic leukemia (ALL). However, immune reaction to the drug may increase the clearance or impair the function of L-asparaginase and reduces its therapeutic efficacy. The objective of this study was to identify potential plasma proteins that could be used as proxies for L-asparaginase activity. METHODS Fibrinogen, von Willebrand factor antigen (VWF:Ag), total protein, and albumin levels as well as antithrombin (AT) and L-asparaginase activities were measured in 97 children with ALL treated for prolonged period of time with L-asparaginase. Binary logistic regression and a receiver operating characteristic (ROC) curve analysis were performed to evaluate the predictive value of plasma proteins for L-asparaginase activity. RESULTS Median E. coli L-asparaginase activity was 220 IU/L (range, 0-1308) throughout the treatment period. L-asparaginase activity was below 100 IU/L in 23% of measured samples. L-asparaginase activity was inversely associated with AT activity, fibrinogen, total protein, and albumin levels (r = -0.63, -0.62, -0.57, and -0.45, respectively; P < 0.0001), but not with VWF:Ag. ROC curve analyses showed an intermediate accuracy of AT activity (area under the ROC curve [AUC] = 0.77) to detect specimens with subtherapeutic level of L-asparaginase. An optimal accuracy was found when AT and fibrinogen were combined (AUC = 0.82; sensitivity = 75%; specificity = 82%; positive predictive value = 55%; negative predictive value = 92%) with cutoff values of 0.73 IU/mL and 1.85 g/L, respectively. CONCLUSIONS AT combined with fibrinogen levels could be used as a proxy to identify patients with therapeutic level of L-asparaginase activity in the absence of real-time asparaginase measurement during prolonged exposure to L-asparaginase.
Collapse
Affiliation(s)
- Clémence Merlen
- Department of Hematology/Oncology, CHU Sainte-Justine, Montreal, QC, Canada
| | - Arnaud Bonnefoy
- Department of Hematology/Oncology, CHU Sainte-Justine, Montreal, QC, Canada
| | - Cynthia Afeich
- Department of Pharmacology, CHU Sainte-Justine, Montreal, QC, Canada
| | - Yves Théorêt
- Department of Pharmacology, CHU Sainte-Justine, Montreal, QC, Canada
| | | | - Jean-Marie Leclerc
- Department of Hematology/Oncology, CHU Sainte-Justine, Montreal, QC, Canada
| | | |
Collapse
|
66
|
Kloos RQH, Pieters R, van den Bos C, van Eijkelenburg NKA, de Jonge R, van der Sluis IM. The effect of asparaginase therapy on methotrexate toxicity and efficacy in children with acute lymphoblastic leukemia. Leuk Lymphoma 2019; 60:3002-3010. [PMID: 31120351 DOI: 10.1080/10428194.2019.1613537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Asparaginase and methotrexate (MTX), both essential for pediatric acute lymphoblastic leukemia therapy, are often used concomitantly. Depending on the sequence, in vitro, asparaginase inhibits MTX-polyglutamate (MTXPG) formation, and side effects overlap. MTX toxicity and efficacy, reflected by intracellular erythrocyte MTXPG's, were compared between children treated with and without asparaginase during high dose MTX (HD-MTX) courses of the DCOG ALL-11 protocol (NL50250.078.14). Seventy-three patients, of whom 23 received asparaginase during the HD-MTX courses, were included. Grade 3-4 leukopenia and neutropenia occurred more often (59% and 86% vs. 30% and 62%). The number of infections, grade 3-4 hepatotoxicity, nephrotoxicity, and neurotoxicity did not differ. Patients with asparaginase had lower MTXPG levels, although to a lesser extent than in vitro studies. Although patients with asparaginase during HD-MTX courses showed more myelosuppression, this had no (serious) clinical consequences. Regarding the MTX efficacy, the schedule-related antagonism seen in in vitro seems less important in vivo.
Collapse
Affiliation(s)
- Robin Q H Kloos
- Department of Pediatric Oncology and Hematology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Cor van den Bos
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Academic Medical Center, Amsterdam, The Netherlands
| | | | - Robert de Jonge
- Department of Clinical Chemistry, Amsterdam UMC, Amsterdam, The Netherlands
| | - Inge M van der Sluis
- Department of Pediatric Oncology and Hematology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
67
|
Madariaga A, Rustin GJS, Buckanovich RJ, Trent JC, Oza AM. Wanna Get Away? Maintenance Treatments and Chemotherapy Holidays in Gynecologic Cancers. Am Soc Clin Oncol Educ Book 2019; 39:e152-e166. [PMID: 31099646 DOI: 10.1200/edbk_238755] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Epithelial ovarian cancer has a very high rate of relapse after primary therapy; historically approximately 70% of patients with a complete clinical response to surgery and adjuvant chemotherapy will relapse and die of the disease. Although this number has slowly improved, cure rates remain less than 50%. As such, maintenance therapy with the aim of preventing or delaying disease relapse and the goal of improving overall survival has been the subject of intense study. Numerous earlier studies with agents ranging from radioactive phosphorus to extended frontline therapy or to monthly taxol administration demonstrated encouraging improvements in progression-free survival (PFS) only to find, disappointingly, no benefit in overall survival. In addition, the PFS advantage of maintenance therapy was associated with disconcerting side effects such that maintenance therapy was not adapted as standard of care. Studies with bevacizumab and PARP inhibitors have demonstrated a PFS advantage with a manageable side-effect profile. However, an overall survival advantage remains unclear, and the use of these approaches thus remains controversial. Furthermore, in recurrent disease, the length of chemotherapy and benefits of extended chemotherapy is unclear. Thus, additional trials assessing maintenance strategies in ovarian and other gynecologic malignancies are needed.
Collapse
Affiliation(s)
- Ainhoa Madariaga
- 1 Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | | | | | - Amit M Oza
- 1 Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
68
|
Burkhardt B, Hermiston ML. Lymphoblastic lymphoma in children and adolescents: review of current challenges and future opportunities. Br J Haematol 2019; 185:1158-1170. [PMID: 30809797 DOI: 10.1111/bjh.15793] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lymphoblastic lymphoma (LBL) is the second most common type of Non-Hodgkin Lymphoma (NHL) in childhood and adolescence, accounting for 25-35% of all cases. The majority, 70-80%, is of T-lymphoblastic origin while 20-25% arise from B lymphoblasts. With current therapy, the event-free and overall survivals for paediatric LBL patients now exceeds 80%. Therapy, especially in T-LBL with large mediastinal tumours, is challenging, with both significant morbidity and late sequela. An additional challenge is the dismal prognosis of patients with refractory or relapsed disease. This review article will focus on the growing knowledge of the pathogenesis and biology of LBL, recent advances and challenges in the therapy of LBL, and ongoing and future efforts and opportunities in optimizing therapy and developing novel targeted treatment approaches.
Collapse
Affiliation(s)
- Birgit Burkhardt
- Paediatric Haematology and Oncology, University Hospital Muenster, Muenster, Germany
| | - Michelle L Hermiston
- Pediatric Hematology and Oncology, University of California, San Francisco, CA, USA
| |
Collapse
|
69
|
Yang KB, Sun XF, Zhen ZZ, Lu SY, Zhu J, Sun FF, Wang J, Huang JT, Chen RR, Ye LT, Liu Y, You ZY. [Impact of intensified maintenance therapy on the prognosis of children and adolescents with advanced lymphoblastic lymphoma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 38:778-783. [PMID: 29081195 PMCID: PMC7348356 DOI: 10.3760/cma.j.issn.0253-2727.2017.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
目的 探讨增加维持治疗强度对Ⅲ期和Ⅳ期儿童青少年淋巴母细胞淋巴瘤(Lymphoblastic lymphoma, LBL)患者预后的影响。 方法 回顾性分析接受BFM-NHL-90/-95方案治疗且未做纵隔和中枢预防性放疗的Ⅲ期和Ⅳ期儿童青少年LBL患者的治疗结果。研究分组:1998年至2005年收治的患者于维持治疗阶段,在口服巯基嘌呤和甲氨蝶呤的基础上,定期采用“足叶乙甙+阿糖胞苷”和大剂量甲氨蝶呤交替进行化疗,为强化维持治疗组;其余为非强化维持治疗组。 结果 187例LBL患者纳入研究,其中强化维持治疗组52例,非强化维持治疗组135例,两组患者的性别、年龄、免疫分型、临床分期、危险度分层、受累部位的差异均无统计学意义(P值均>0.05);中位随访48(0.5~221)个月,两组患者的5年无事件生存(EFS)率分别为(76.9±5.8)%和(77.9±4.3)%(χ2=0.249,P=0.617),5年总生存(OS)率分别为(78.8±5.7)%和(79.8±4.1)%(χ2=0.353,P=0.552),差异均无统计学意义;亚组分析结果显示,两组患者在不同临床分期(Ⅲ/Ⅳ期)、免疫分型(T/B-LBL)和危险分层(中/高危)中的EFS、OS率差异均无统计学意义(P值均>0.05)。维持治疗期间,强化维持治疗组和非强化维持治疗组患者Ⅲ、Ⅳ级骨髓抑制发生率分别为55.8%和18.5%(χ2=25.363,P<0.05)。 结论 提高维持治疗强度并未提高Ⅲ期和Ⅳ期儿童青少年LBL患者的长期生存且可增加骨髓抑制等治疗相关不良反应。
Collapse
Affiliation(s)
- K B Yang
- State Key Laboratory of Oncology in Southern China; Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Osteonecrosis in pediatric cancer survivors: Epidemiology, risk factors, and treatment. Surg Oncol 2019; 28:214-221. [PMID: 30851903 DOI: 10.1016/j.suronc.2019.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/25/2019] [Accepted: 02/02/2019] [Indexed: 01/30/2023]
Abstract
Several treatment regimens for childhood malignancies have been associated with the development of osteonecrosis, including radiation therapy, glucocorticoid medications, immunotherapy (including anti-angiogenic agents), and several chemotherapeutic agents. Adolescents older than 10 years are at greatest risk of developing osteonecrosis within 1 year of initiating therapy. Screening with magnetic resonance imaging in this high-risk population may be a useful method for detecting osteonecrosis. Surgery may be required for lesions that have progressed substantially despite nonoperative interventions.
Collapse
|
71
|
Touzart A, Lengliné E, Latiri M, Belhocine M, Smith C, Thomas X, Spicuglia S, Puthier D, Pflumio F, Leguay T, Graux C, Chalandon Y, Huguet F, Leprêtre S, Ifrah N, Dombret H, Macintyre E, Hunault M, Boissel N, Asnafi V. Epigenetic Silencing Affects l-Asparaginase Sensitivity and Predicts Outcome in T-ALL. Clin Cancer Res 2019; 25:2483-2493. [PMID: 30659025 DOI: 10.1158/1078-0432.ccr-18-1844] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/29/2018] [Accepted: 01/15/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Biological explanation for discrepancies in patient-related response to chemotherapy depending on the underlying oncogenic events is a promising research area. TLX1- or TLX3-deregulated T-cell acute lymphoblastic leukemias (T-ALL; TLX1/3+) share an immature cortical phenotype and similar transcriptional signatures. However, their prognostic impacts differ, and inconsistent clinical outcome has been reported for TLX3. We therefore hypothesized that the overlapping transcriptional profiles of TLX1+ and TLX3+ T-ALLs would allow identification of candidate genes, which might determine their distinct clinical outcomes. EXPERIMENTAL DESIGN We compared TLX1+ and TLX3+ adult T-ALL outcome in the successive French national LALA-94 and GRAALL-2003/2005 multicentric trials and analyzed transcriptomic data to identify differentially expressed genes. Epigenetic regulation of asparagine synthetase (ASNS) and in vitro l-asparaginase sensitivity were evaluated for T-ALL cell lines and primary samples. RESULTS We show that TLX1+ patients expressed low levels of ASNS when compared with TLX3+ and TLX-negative patients, due to epigenetic silencing of ASNS by both DNA methylation and a decrease of active histone marks. Promoter methylation of the ASNS gene correlated with l-asparaginase sensitivity in both T-ALL cell lines and patient-derived xenografts. Finally, ASNS promoter methylation was an independent prognostic factor for both event-free survival [HR, 0.42; 95% confidence interval (CI), 0.24-0.71; P = 0.001] and overall survival (HR, 0.40; 95% CI, 0.23-0.70; P = 0.02) in 160 GRAALL-2003/2005 T-ALL patients and also in an independent series of 47 LL03-treated T lymphoblastic lymphomas (P = 0.012). CONCLUSIONS We conclude that ASNS methylation status at diagnosis may allow individual adaptation of l-asparaginase dose.
Collapse
Affiliation(s)
- Aurore Touzart
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de recherche Médicale (INSERM) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Etienne Lengliné
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
| | - Mehdi Latiri
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de recherche Médicale (INSERM) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Mohamed Belhocine
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France; Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Charlotte Smith
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de recherche Médicale (INSERM) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Xavier Thomas
- Division of Hematology, Hospices Civils de Lyon, INSERM U1052-Centre National de la Recherche Scientifique UMR 5286, Centre Hospitalier Lyon Sud, Pierre Benite, France
| | - Salvatore Spicuglia
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France; Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Denis Puthier
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France; Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Françoise Pflumio
- INSERM UMR967, CEA/DSV/iRCM, Laboratory of Hematopoietic Stem Cells and Leukemic Cells, Equipe labellisée par la Ligue Nationale Contre le Cancer, Université Paris Diderot, Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Fontenay-aux-Roses, France
| | - Thibaut Leguay
- Division of Hematology, Hôpital du Haut-Levêque, Pessac, France
| | - Carlos Graux
- Department of Hematology, Mont-Godinne University Hospital, Yvoir, Belgium
| | - Yves Chalandon
- Hematology Division, Department of Oncology, University Hospital, Genève, Switzerland; Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Françoise Huguet
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Stéphane Leprêtre
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1245, Department of Hematology, Centre Henri-Becquerel and Normandie Univ UNIROUEN, Rouen, France
| | - Norbert Ifrah
- PRES LUNAM, CHU Angers Service des Maladies du Sang et INSERM U 892, Angers, France
| | - Hervé Dombret
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
| | - Elizabeth Macintyre
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de recherche Médicale (INSERM) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Mathilde Hunault
- Division of Hematology, Centre Hospitalier Universitaire d'Angers, Angers, France; INSERM U892/Centre National de la Recherche Scientifique 6299, Angers, CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Nicolas Boissel
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
| | - Vahid Asnafi
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de recherche Médicale (INSERM) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France.
| |
Collapse
|
72
|
Kloos RQH, van Litsenburg RRL, Wolf S, Wismans L, Kaspers GJL, Uyl-de Groot CA, Pieters R, van der Sluis IM. A cost-effectiveness analysis of Erwinia asparaginase therapy in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2019; 66:e27458. [PMID: 30246496 DOI: 10.1002/pbc.27458] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 11/08/2022]
Abstract
OBJECTIVES Erwinia asparaginase is used as a second-line formulation after a neutralizing hypersensitivity reaction to the first-line formulation of asparaginase. Here, we have performed a cost-effectiveness analysis of Erwinia asparaginase treatment. METHODS Children with acute lymphoblastic leukemia treated according to the Dutch Childhood Oncology ALL-10 or ALL-11 protocol were included and initially treated with PEGasparaginase in the intensification phase. The total treatment costs of this treatment phase, quality of life (QoL), and life years saved (LYS) were studied for two scenarios: (a) patients were switched to Erwinia asparaginase treatment after a hypersensitivity reaction, or (b) asparaginase would have been permanently stopped. RESULTS Sixty-eight patients were included. There was no difference in QoL between patients with and without a hypersensitivity reaction. The mean costs of the intensification phase per patient were $40,925 if PEGasparaginase could be continued, $175,632 if patients had to switch to Erwinia asparaginase, and $21,190 if asparaginase would have been permanently stopped. An extrapolation of the literature suggests that the 5-year event-free survival would be 10.3% lower without intensive asparaginase treatment if asparaginase is stopped after a reaction. Thus, the costs per LYS were $1892 for scenario 1 and $872 for scenario 2. CONCLUSIONS Switching to Erwinia asparaginase increases the costs per LYS by $1020, which is modest in view of the total costs. Moreover, when asparaginase treatment can be completed by switching to Erwinia asparaginase, relapses-and consequential costs-will be avoided. Therefore, from a cost perspective, we recommend a switch to Erwinia asparaginase to complete asparaginase treatment.
Collapse
Affiliation(s)
- Robin Q H Kloos
- Department of Pediatric Oncology and Hematology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Sarah Wolf
- Institute of Health Care Policy & Management/Institute for Medical Technology Assessment Erasmus University, Rotterdam, The Netherlands
| | - Leonoor Wismans
- Department of Pediatric Oncology and Hematology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Carin A Uyl-de Groot
- Institute of Health Care Policy & Management/Institute for Medical Technology Assessment Erasmus University, Rotterdam, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | |
Collapse
|
73
|
Abstract
Lymphomas in adolescents and young adults represent approximately one quarter of all cancers in this age group. Historically, adolescent and young adult cancer patients represent a unique population with diverging issues surrounding psychosocial hardships/barriers, economics, and lack of standardization of therapeutic approaches.Furthermore, the biologic differences within the adolescent and young adult population seen in various lymphoma subtypes likely play a role in overall outcomes for this group. Without an organized approach to clinical and translational research for adolescent and young adult patients within specialized treatment centers, this population may continue to experience inferior results. Here we look at the current perspectives of adolescent and young adult lymphomas with respect to disease biology, clinical characteristics, treatment, and prognosis of this unique lymphoma population.
Collapse
|
74
|
Faret M, de Morais SB, Zanchin NIT, de Souza TDACB. L-Asparaginase from Erwinia carotovora: insights about its stability and activity. Mol Biol Rep 2018; 46:1313-1316. [PMID: 30446961 DOI: 10.1007/s11033-018-4459-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/30/2018] [Indexed: 11/26/2022]
Abstract
Enzymatic prospection indicated that L-asparaginase from Erwinia carotovora (ECAR-LANS) posses low glutaminase activity and much effort has been made to produce therapeutic ECAR-LANS. However, its low stability precludes its use in therapy. Herein, biochemical and biophysical assays provided data highlighting the influence of solubilization and storage into ECAR-LANS structure, stability, and activity. Moreover, innovations in recombinant expression and purification guaranteed the purification of functional tetramers. According to solubilization condition, the L-asparaginase activity and temperature of melting ranged up to 25-32%, respectively. CD spectra indicate the tendency of ECAR-LANS to instability and the influence of β-structures in activity. These results provide relevant information to guide formulations with prolonged action in the bloodstream.
Collapse
Affiliation(s)
- Marcele Faret
- Instituto Carlos Chagas, ICC - FIOCRUZ/PR, Rua Algacyr Munhoz Mader, 3775, bloco C, Curitiba,, Paraná, 81350-010, Brazil
| | - Stephanie Bath de Morais
- Instituto Carlos Chagas, ICC - FIOCRUZ/PR, Rua Algacyr Munhoz Mader, 3775, bloco C, Curitiba,, Paraná, 81350-010, Brazil
| | - Nilson Ivo Tonin Zanchin
- Instituto Carlos Chagas, ICC - FIOCRUZ/PR, Rua Algacyr Munhoz Mader, 3775, bloco C, Curitiba,, Paraná, 81350-010, Brazil
| | | |
Collapse
|
75
|
Jeong HC, Kim T, Yang DH, Shin KH. Development of a UPLC-MS/MS method for the therapeutic monitoring of L-asparaginase. Transl Clin Pharmacol 2018; 26:134-140. [PMID: 32055563 PMCID: PMC6989229 DOI: 10.12793/tcp.2018.26.3.134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 12/02/2022] Open
Abstract
This study aimed to develop a UPLC-MS/MS method for determining plasma levels of L-aspartic acid and L-asparagine and the activity of L-asparaginase. L-aspartic acid, L-asparagine, and L-aspartic acid-2,3,3-d3 were extracted from human plasma by protein precipitation with sulfosalicylic acid (30%, v/v). The plasma samples were analyzed using an Imtakt Intrada amino acid analysis column with 25 mM ammonium formate and 0.5% formic acid in acetonitrile as the mobile phase with step gradient method at a flow rate of 0.5 mL/min. The injection volume was 5 µL, and the total run time was 15 min. Inter- and intra-batch accuracies (%) ranged from 96.62–106.0% for L-aspartic acid and 89.85–104.8%, for L-asparagine, and the coefficient of variation (CV%) did not exceed 7%. The validation results for L-aspartic acid and L-asparagine satisfied the specified criterion, however, the results for L-asparaginase activity assay showed a borderline validity. This study could be a foundation for further development of therapeutic drug monitoring systems using UPLC-MS/MS.
Collapse
Affiliation(s)
- Hyeon-Cheol Jeong
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Therasa Kim
- Department of Hematology, Chonnam National University Hwasun Hospital, Hwasun 58128, Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 03080, Korea
| | - Deok-Hwan Yang
- Department of Hematology, Chonnam National University Hwasun Hospital, Hwasun 58128, Korea
| | - Kwang-Hee Shin
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
76
|
Winter SS, Dunsmore KP, Devidas M, Wood BL, Esiashvili N, Chen Z, Eisenberg N, Briegel N, Hayashi RJ, Gastier-Foster JM, Carroll AJ, Heerema NA, Asselin BL, Gaynon PS, Borowitz MJ, Loh ML, Rabin KR, Raetz EA, Zweidler-Mckay PA, Winick NJ, Carroll WL, Hunger SP. Improved Survival for Children and Young Adults With T-Lineage Acute Lymphoblastic Leukemia: Results From the Children's Oncology Group AALL0434 Methotrexate Randomization. J Clin Oncol 2018; 36:2926-2934. [PMID: 30138085 DOI: 10.1200/jco.2018.77.7250] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Early intensification with methotrexate (MTX) is a key component of acute lymphoblastic leukemia (ALL) therapy. Two different approaches to MTX intensification exist but had not been compared in T-cell ALL (T-ALL): the Children's Oncology Group (COG) escalating dose intravenous MTX without leucovorin rescue plus pegaspargase escalating dose, Capizzi-style, intravenous MTX (C-MTX) regimen and the Berlin-Frankfurt-Muenster (BFM) high-dose intravenous MTX (HDMTX) plus leucovorin rescue regimen. PATIENTS AND METHODS COG AALL0434 included a 2 × 2 randomization that compared the COG-augmented BFM (ABFM) regimen with either C-MTX or HDMTX during the 8-week interim maintenance phase. All patients with T-ALL, except for those with low-risk features, received prophylactic (12 Gy) or therapeutic (18 Gy for CNS3) cranial irradiation during either the consolidation (C-MTX; second month of therapy) or delayed intensification (HDMTX; seventh month of therapy) phase. RESULTS AALL0434 accrued 1,895 patients from 2007 to 2014. The 5-year event-free survival and overall survival rates for all eligible, evaluable patients with T-ALL were 83.8% (95% CI, 81.2% to 86.4%) and 89.5% (95% CI, 87.4% to 91.7%), respectively. The 1,031 patients with T-ALL but without CNS3 disease or testicular leukemia were randomly assigned to receive ABFM with C-MTX (n = 519) or HDMTX (n = 512). The estimated 5-year disease-free survival ( P = .005) and overall survival ( P = .04) rates were 91.5% (95% CI, 88.1% to 94.8%) and 93.7% (95% CI, 90.8% to 96.6%) for C-MTX and 85.3% (95% CI, 81.0%-89.5%) and 89.4% (95% CI, 85.7%-93.2%) for HDMTX. Patients assigned to C-MTX had 32 relapses, six with CNS involvement, whereas those assigned to HDMTX had 59 relapses, 23 with CNS involvement. CONCLUSION AALL0434 established that ABFM with C-MTX was superior to ABFM plus HDMTX for T-ALL in approximately 90% of patients who received CRT, with later timing for those receiving HDMTX.
Collapse
Affiliation(s)
- Stuart S Winter
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Kimberly P Dunsmore
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Meenakshi Devidas
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Brent L Wood
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Natia Esiashvili
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Zhiguo Chen
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Nancy Eisenberg
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Nikki Briegel
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Robert J Hayashi
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Julie M Gastier-Foster
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Andrew J Carroll
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Nyla A Heerema
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Barbara L Asselin
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Paul S Gaynon
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Michael J Borowitz
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Mignon L Loh
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Karen R Rabin
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Elizabeth A Raetz
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Patrick A Zweidler-Mckay
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Naomi J Winick
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - William L Carroll
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Stephen P Hunger
- Stuart S. Winter, Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN; Kimberly P. Dunsmore, Carilion Clinic, Roanoke, VA; Meenakshi Devidas and Zhiguo Chen, University of Florida, Gainesville, FL; Brent L. Wood, Seattle Children's Hospital, Seattle, WA; Natia Esiashvili, Emory University, Atlanta, GA; Nancy Eisenberg, University of New Mexico Health Sciences Center, Albuquerque, NM; Nikki Briegel, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Robert J. Hayashi, St Louis Children's Hospital, St Louis, MO; Julie M. Gastier-Foster, Nationwide Children's Hospital; Julie M. Gastier-Foster and Nyla A. Heerema, Ohio State University, Columbus, OH; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Barbara L. Asselin, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester; Elizabeth A. Raetz and William L. Carroll, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Paul S. Gaynon, Children's Hospital of Los Angeles, Los Angeles; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Karen R. Rabin, Baylor College of Medicine, Houston; Naomi J. Winick, University of Texas Southwestern, Dallas, TX; Patrick A. Zweidler-Mckay, ImmunoGen, Waltham, MA; and Stephen P. Hunger, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
77
|
Hochberg J, Flower A, Brugieres L, Cairo MS. NHL in adolescents and young adults: A unique population. Pediatr Blood Cancer 2018; 65:e27073. [PMID: 29741220 DOI: 10.1002/pbc.27073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 02/07/2018] [Accepted: 03/04/2018] [Indexed: 12/21/2022]
Abstract
Non-Hodgkin lymphoma (NHL) is a heterogeneous group of lymphoid malignancies with high incidence in adolescents and young adults (AYAs). The most common diseases include diffuse large B-cell lymphoma, anaplastic large cell lymphoma, Burkitt lymphoma, lymphoblastic lymphoma, and primary mediastinal large B-cell lymphoma. In comparison to younger children and adults, AYAs (15-39 years) with NHL present a specific set of challenges including variations in tumor biology, inconsistent treatment, pharmacodynamics, and psychosocial barriers. While survival of AYAs with NHL has improved, cure rates remain suboptimal. Incorporation of novel agents into pediatric-inspired treatment regimens specifically designed for NHL in AYAs has led to improved outcomes. Consideration of AYAs as a distinct population in the diagnosis and treatment of NHL is encouraged.
Collapse
Affiliation(s)
- Jessica Hochberg
- Department of Pediatrics, New York Medical College, Valhalla, New York
| | - Allyson Flower
- Department of Pediatrics, New York Medical College, Valhalla, New York.,Department of Microbiology & Immunology, New York Medical College, Valhalla, New York
| | | | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, New York.,Department of Microbiology & Immunology, New York Medical College, Valhalla, New York.,Department of Medicine, New York Medical College, Valhalla, New York.,Department of Pathology, New York Medical College, Valhalla, New York.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York
| |
Collapse
|
78
|
Hashii Y, Yoshida M, Hara J, Nishimura S, Yumura-Yagi K, Horibe K, Nakahata T. Acid-suppressing Drugs and a Low 1 Level of Antithrombin as Risk Factors for L-Asparaginase-associated Pancreatitis: A Case-control Study in the Japan Association of Childhood Leukemia Study (JACLS). J Pediatr Hematol Oncol 2018; 40:374-378. [PMID: 29697579 DOI: 10.1097/mph.0000000000001193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
L-Asparaginase has significantly improved outcome for children with acute lymphoblastic leukemia and has become an essential component of multiagent chemotherapy. However, there are many adverse events due to L-asparaginase, including acute pancreatitis. The pathology of L-asparaginase-associated pancreatitis (AAP) remains unclear. We compared patients who developed AAP (n=29) and random matched controls (n=36) who had been enrolled in the Japan Association of Childhood Leukemia Study of the ALL-02 protocol. AAP and control patients were matched for age, sex, treatment, and protocol risk. We examined correlations between AAP development and clinical symptoms, laboratory data, and concomitant medication. Abdominal pain and nausea were common presenting symptoms for AAP. There was an increased risk of AAP in patients using gastric acid-suppressing agents and antithrombin (AT) supplementation. Mean fibrinogen and AT levels before the onset of AAP were lower in AAP patients than in controls. Decreased AT and fibrinogen levels resulting from the strong suppression of protein synthesis by L-asparaginase were predictive signs for AAP. Our epidemiological approach should prove clinically useful for the diagnosis the AAP as early as possible.
Collapse
Affiliation(s)
- Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya
| | - Makoto Yoshida
- Department of Pediatrics, Asahikawa Medical University, Asahikawa
| | - Junichi Hara
- Department of Pediatrics, Osaka City General Hospital, Osaka
| | | | | | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya
| | | |
Collapse
|
79
|
Thu Huynh V, Bergeron S. Asparaginase Toxicities: Identification and Management in Patients With Acute Lymphoblastic Leukemia
. Clin J Oncol Nurs 2018; 21:E248-E259. [PMID: 28945721 DOI: 10.1188/17.cjon.e248-e259] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Acute lymphoblastic leukemia (ALL) is a common cancer in children, and outcomes have greatly improved because of the refinement of multiagent chemotherapy regimens that include intensified asparaginase therapy. Asparaginase, a cornerstone of modern pediatric chemotherapy regimens for ALL and asparaginase-containing protocols, is increasingly used in adolescent and adult patients historically treated with asparaginase-free regimens.
. OBJECTIVES This article is an overview of commonly encountered asparaginase-
associated toxicities and offers recommendations for treatment management.
. METHODS A literature review was conducted, reviewing asparaginase and common toxicities, specifically hypersensitivity, pancreatitis, thrombosis, hyperbilirubinemia, and hyperglycemia.
. FINDINGS The rapid identification and management of common asparaginase-associated adverse events can reduce symptom severity and limit potential interruptions to therapy, possibly improving outcomes.
Collapse
|
80
|
Rau RE, Dreyer Z, Choi MR, Liang W, Skowronski R, Allamneni KP, Devidas M, Raetz EA, Adamson PC, Blaney SM, Loh ML, Hunger SP. Outcome of pediatric patients with acute lymphoblastic leukemia/lymphoblastic lymphoma with hypersensitivity to pegaspargase treated with PEGylated Erwinia asparaginase, pegcrisantaspase: A report from the Children's Oncology Group. Pediatr Blood Cancer 2018; 65:10.1002/pbc.26873. [PMID: 29090524 PMCID: PMC5839116 DOI: 10.1002/pbc.26873] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Erwinia asparaginase is a Food and Drug Administration approved agent for the treatment of acute lymphoblastic leukemia (ALL) for patients who develop hypersensitivity to Escherichia coli derived asparaginases. Erwinia asparaginase is efficacious, but has a short half-life, requiring six doses to replace one dose of the most commonly used first-line asparaginase, pegaspargase, a polyethylene glycol (PEG) conjugated E. coli asparaginase. Pegcristantaspase, a recombinant PEGylated Erwinia asparaginase with improved pharmacokinetics, was developed for patients with hypersensitivity to pegaspargase. Here, we report a series of patients treated on a pediatric phase 2 trial of pegcrisantaspase. PROCEDURE Pediatric patients with ALL or lymphoblastic lymphoma and hypersensitivity to pegaspargase enrolled on Children's Oncology Group trial AALL1421 (Jazz 13-011) and received intravenous pegcrisantaspase. Serum asparaginase activity (SAA) was monitored before and after dosing; immunogenicity assays were performed for antiasparaginase and anti-PEG antibodies and complement activation was evaluated. RESULTS Three of the four treated patients experienced hypersensitivity to pegcrisantaspase manifested as clinical hypersensitivity reactions or rapid clearance of SAA. Immunogenicity assays demonstrated the presence of anti-PEG immunoglobulin G antibodies in all three hypersensitive patients, indicating a PEG-mediated immune response. CONCLUSIONS This small series of patients, nonetheless, provides data, suggesting preexisting immunogenicity against the PEG moiety of pegaspargase and poses the question as to whether PEGylation may be an effective strategy to optimize Erwinia asparaginase administration. Further study of larger cohorts is needed to determine the incidence of preexisting antibodies against PEG-mediated hypersensitivity to pegaspargase.
Collapse
Affiliation(s)
- Rachel E. Rau
- Division of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - ZoAnn Dreyer
- Division of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | - Wei Liang
- Jazz Pharmaceuticals, Palo Alto, California
| | | | | | - Meenakshi Devidas
- Department of Biostatistics, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville, Florida
| | | | - Peter C. Adamson
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan M. Blaney
- Division of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Mignon L Loh
- Department of Pediatrics, University of California School of Medicine, San Francisco, California
| | - Stephen P. Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
81
|
|
82
|
Kloos RQH, Uyl-de Groot CA, van Litsenburg RRL, Kaspers GJL, Pieters R, van der Sluis IM. A cost analysis of individualized asparaginase treatment in pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 2017; 64. [PMID: 28766887 DOI: 10.1002/pbc.26651] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Therapeutic drug monitoring (TDM) of asparaginase is necessary to respond to variability in asparaginase activity levels, detect silent inactivation, and distinguish between real allergies and allergic-like reactions with and without asparaginase neutralization, respectively. In this study, the costs of an individualized and fixed asparaginase dosing schedule were compared. PROCEDURE Patients, treated according to the Dutch Childhood Oncology Group ALL-11 protocol (individualized PEGasparaginase treatment, starting dose: 1,500 IU/m2 ) or ALL-10 protocol (native Escherichia coli asparaginase followed by 2,500 IU/m2 PEGasparaginase), were included. To focus on TDM of PEGasparaginase, the costs were also calculated excluding patients treated with Erwinia asparaginase and compared to a hypothetical protocol with a fixed dose of 1,500 IU/m2 PEGasparaginase. Direct asparaginase-related medical costs, including costs for asparaginase use (calculated with the absolute dose), TDM, laboratory tests, daycare treatment, and outpatient clinic visits, were calculated. RESULTS Eighty-three ALL-10 patients and 51 ALL-11 patients were included. The asparaginase-related costs were 30.8% lower in ALL-11 than in ALL-10 ($29,048 vs. $41,960). The ALL-11 costs of nonallergic patients were 20.4% lower, when using TDM, than the hypothetical protocol with a fixed dose of 1,500 IU/m2 ($13,178 vs. $16,551). TDM accounted for 12.4% of the costs. Including asparaginase waste, TDM in ALL-11 will be cost saving if three doses can be prepared out of one vial compared to a fixed dose of 1,500 IU/m2 . CONCLUSIONS TDM of asparaginase is cost saving if calculated with the absolute asparaginase dose and will be if the waste is minimalized by preparing multiple doses out of one vial.
Collapse
Affiliation(s)
- Robin Q H Kloos
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Carin A Uyl-de Groot
- Institute of Health Care Policy and Management/Institute for Medical Technology Assessment, Erasmus University, Rotterdam, The Netherlands
| | - Raphaële R L van Litsenburg
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Gertjan J L Kaspers
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Inge M van der Sluis
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
83
|
Liang J, Shi P, Guo X, Li J, He L, Wang Y, Wei Q, Huang F, Fan Z, Xu B. A retrospective comparison of Escherichia coli and polyethylene glycol-conjugated asparaginase for the treatment of adolescents and adults with newly diagnosed acute lymphoblastic leukemia. Oncol Lett 2017; 15:75-82. [PMID: 29387211 PMCID: PMC5769219 DOI: 10.3892/ol.2017.7271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/16/2017] [Indexed: 11/10/2022] Open
Abstract
Data from clinical trials suggest that polyethylene glycol-conjugated asparaginase (PEG asparaginase) should be recommended as a replacement for Escherichia coli (E. coli) asparaginase in the treatment of pediatric acute lymphoblastic leukemia (ALL) due to its prolonged effect, similar safety profile and convenience. The present study investigated the efficacy and safety of PEG asparaginase in adolescents and adults with newly diagnosed ALL. The clinical data of 122 patients, ≥14 years old with de novo ALL, who received either PEG asparaginase or E. coli asparaginase as part of an induction regimen, were retrospectively analyzed. The results revealed that PEG asparaginase had a comparable complete remission rate (95.65 vs. 90.79%), median overall survival time (14.07 vs. 16.29 months) and median relapse-free survival time (10.00 vs. 8.57 months) with E. coli asparaginase. In addition, patients <35 years old receiving PEG asparaginase obtained a higher median RFS time compared with those receiving E. coli asparaginase (10.93 vs. 8.97 months; P=0.037). Patients treated with E. coli asparaginase exhibited a significantly higher incidence of central nervous system leukemia (CNSL) compared with those treated with PEG asparaginase (27.63 vs. 10.87%; P=0.028) during the consolidation phase. Toxic events, including allergy, grade III–IV liver dysfunction, renal function damage and pancreatic lesions were similar between the two groups. A longer duration of coagulation dysfunction (9.80±5.51 vs. 6.80±4.21 days; P=0.002) and agranulocytosis (18.89±8.79 vs. 12.03±8.34 days; P<0.01), and a higher incidence of grade IV–V infections (22.73 vs. 7.25%; P=0.018) were observed in the PEG asparaginase group. However, these did not increase bleeding events or infection-associated mortalities. When taking the convenience and superior efficacy in preventing CNSL into consideration, PEG asparaginase is a candidate for first-line treatment of adolescent and adult ALL. A larger prospective clinical trial is required to further confirm this point of view.
Collapse
Affiliation(s)
- Jiabao Liang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Hematology, The First People Hospital of Foshan, Affiliated Hospital of Sun Yat-Sen University, Foshan, Guangdong 528000, P.R. China
| | - Pengcheng Shi
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xutao Guo
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jie Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Lingli He
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yan Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qi Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhiping Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| |
Collapse
|
84
|
Salzer W, Bostrom B, Messinger Y, Perissinotti AJ, Marini B. Asparaginase activity levels and monitoring in patients with acute lymphoblastic leukemia. Leuk Lymphoma 2017; 59:1797-1806. [DOI: 10.1080/10428194.2017.1386305] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Wanda Salzer
- U.S. Army, Medical Research and Materiel Command, Fort Detrick, MD, USA
| | | | | | | | - Bernard Marini
- University of Michigan, College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
85
|
Landmann E, Burkhardt B, Zimmermann M, Meyer U, Woessmann W, Klapper W, Wrobel G, Rosolen A, Pillon M, Escherich G, Attarbaschi A, Beishuizen A, Mellgren K, Wynn R, Ratei R, Plesa A, Schrappe M, Reiter A, Bergeron C, Patte C, Bertrand Y. Results and conclusions of the European Intergroup EURO-LB02 trial in children and adolescents with lymphoblastic lymphoma. Haematologica 2017; 102:2086-2096. [PMID: 28983060 PMCID: PMC5709108 DOI: 10.3324/haematol.2015.139162] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 09/28/2017] [Indexed: 11/09/2022] Open
Abstract
In the European Intergroup EURO-LB02 trial, children and adolescents with lymphoblastic lymphoma underwent the non-Hodgkin lymphoma Berlin-Frankfurt-Münster protocol without prophylactic cranial radiotherapy. The primary aims of this trial were to test whether replacing prednisone with dexamethasone during induction increases event-free survival in the subgroups with T-cell lymphoblastic lymphoma and whether therapy duration could be reduced from 24 to 18 months (factorial design, randomizations). These questions could not be answered due to premature closure of the trial. Here we report on the secondary aims of the trial: whether the results of the NHL-BFM90 study could be reproduced and evaluation of disease features and prognostic factors. Three hundred and nineteen patients (66 with precursor B-cell lymphoblastic lymphoma, 233 with T-cell lymphoblastic lymphoma, 12 with mixed phenotype, 8 not classifiable) were enrolled. In induction, 215 patients received prednisone and 104 patients received dexamethasone. The median follow-up was 6.8 years (range, 3.0–10.3). The 5-year event-free survival was 82±2% [12 toxic deaths, 5 secondary malignancies, 43 non-response/relapse (central nervous system n=9; all received prednisone during induction)]. The event-free survival rate was 80±5% for patients with precursor B-cell lymphoblastic lymphoma, 82±3% for those with T-cell lymphoblastic lymphoma, and 100% for patients with a mixed phenotype. During induction, significantly more grade III/IV toxicities were observed in patients receiving dexamethasone, resulting in significant treatment delays. The number of toxic deaths did not differ significantly. The only variable associated with outcome was performance status at diagnosis. The 90% event-free survival rate for patients with T-cell lymphoblastic lymphoma shown in study NHL-BFM90 was not replicated, mainly due to more toxic deaths and central nervous system relapses. Dexamethasone in induction may prevent central nervous system relapse more effectively than prednisone but produces a higher burden of toxicity. (#NCT00275106).
Collapse
Affiliation(s)
- Eva Landmann
- Department of Pediatric Hematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | - Birgit Burkhardt
- Department of Pediatric Hematology and Oncology, Children's University Hospital, Münster, Germany
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Germany
| | - Ulrike Meyer
- Department of Pediatric Hematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | - Wilhelm Woessmann
- Department of Pediatric Hematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | - Wolfram Klapper
- Department of Hematopathology and Lymph Node Registry, University Hospital, Kiel, Germany
| | - Grazyna Wrobel
- Department of Bone Marrow Transplantation, Children's Oncology and Hematology, Wroclaw Medical University, Poland
| | - Angelo Rosolen
- Clinica di Oncoematologia Pediatrica, Università di Padova, Italy
| | - Marta Pillon
- Clinica di Oncoematologia Pediatrica, Università di Padova, Italy
| | - Gabriele Escherich
- Clinic for Pediatric Hematology and Oncology, University Medical Center, Hamburg, Germany
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Austria
| | - Auke Beishuizen
- Department of Pediatric Hematology/Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands and the Dutch Childhood Oncology Group, the Hague, the Netherlands
| | - Karin Mellgren
- Department of Pediatric Oncology and Hematology, The Queen Silvia Children's Hospital, Göteborg, Sweden
| | - Robert Wynn
- Central Manchester University Hospitals, Great Britain
| | - Richard Ratei
- Department of Hematology, Oncology and Tumor Immunology, Helios Klinikum, Berlin-Buch, Germany
| | - Adriana Plesa
- Department of Hematopathology and Flow Cytometry, CHU, Lyon-HCL, France
| | - Martin Schrappe
- Department of Pediatrics, Christian-Albrechts-University, Kiel, Germany
| | - Alfred Reiter
- Department of Pediatric Hematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | - Christophe Bergeron
- Institut d'Hematologie et d'Oncologie Pediatrique, Centre Léon Bérard and HCL, Claude Bernard University, Lyon, France
| | | | - Yves Bertrand
- Institut d'Hematologie et d'Oncologie Pediatrique, Centre Léon Bérard and HCL, Claude Bernard University, Lyon, France
| |
Collapse
|
86
|
Mondelaers V, Suciu S, De Moerloose B, Ferster A, Mazingue F, Plat G, Yakouben K, Uyttebroeck A, Lutz P, Costa V, Sirvent N, Plouvier E, Munzer M, Poirée M, Minckes O, Millot F, Plantaz D, Maes P, Hoyoux C, Cavé H, Rohrlich P, Bertrand Y, Benoit Y. Prolonged versus standard native E. coli asparaginase therapy in childhood acute lymphoblastic leukemia and non-Hodgkin lymphoma: final results of the EORTC-CLG randomized phase III trial 58951. Haematologica 2017; 102:1727-1738. [PMID: 28751566 PMCID: PMC5622857 DOI: 10.3324/haematol.2017.165845] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/21/2017] [Indexed: 12/05/2022] Open
Abstract
Asparaginase is an essential component of combination chemotherapy for childhood acute lymphoblastic leukemia and non-Hodgkin lymphoma. The value of asparaginase was further addressed in a group of non-very high-risk patients by comparing prolonged (long-asparaginase) versus standard (short-asparaginase) native E. coli asparaginase treatment in a randomized part of the phase III 58951 trial of the European Organization for Research and Treatment of Cancer Children's Leukemia Group. The main endpoint was disease-free survival. Overall, 1,552 patients were randomly assigned to long-asparaginase (775 patients) or short-asparaginase (777 patients). Patients with grade ≥2 allergy to native E. coli asparaginase were switched to equivalent doses of Erwinia or pegylated E. coli asparaginase. The 8-year disease-free survival rate (±standard error) was 87.0±1.3% in the long-asparaginase group and 84.4±1.4% in the short-asparaginase group (hazard ratio: 0.87; P=0.33) and the 8-year overall survival rate was 92.6±1.0% and 91.3±1.2% respectively (hazard ratio: 0.89; P=0.53). An exploratory analysis suggested that the impact of long-asparaginase was beneficial in the National Cancer Institute standard-risk group with regards to disease-free survival (hazard ratio: 0.70; P=0.057), but far less so with regards to overall survival (hazard ratio: 0.89). The incidences of grade 3-4 infection during consolidation (25.2% versus 14.4%) and late intensification (22.6% versus 15.9%) and the incidence of grade 2-4 allergy were higher in the long-asparaginase arm (30% versus 21%). Prolonged native E. coli asparaginase therapy in consolidation and late intensification for our non-very high-risk patients did not improve overall outcome but led to an increase in infections and allergy. This trial was registered at www.clinicaltrials.gov as #NCT00003728.
Collapse
Affiliation(s)
- Veerle Mondelaers
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent University, Belgium
| | | | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent University, Belgium
| | - Alina Ferster
- Department of Pediatric Hematology-Oncology, Children's University Hospital Queen Fabiola, Université Libre de Bruxelles (ULB), Belgium
| | | | - Geneviève Plat
- Department of Pediatric Hematology-Oncology, CHU-Hopital Purpan, Toulouse, France
| | - Karima Yakouben
- Department of Pediatric Hematology, Robert Debré Hospital, AP-HP, Paris, France
| | - Anne Uyttebroeck
- Department of Pediatric Hematology-Oncology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Patrick Lutz
- Department of Pediatric Hematology-Oncology, University Hospital Hautepierre, Strasbourg, France
| | - Vitor Costa
- Department of Pediatrics, Portuguese Oncology Institute, Porto, Portugal
| | - Nicolas Sirvent
- Department of Pediatric Hematology-Oncology, CHU, Montpellier, France
| | - Emmanuel Plouvier
- Pediatric Hematology Unit, CHU Jean Minjoz Hospital, Besançon, France
| | - Martine Munzer
- Department of Pediatric Hematology-Oncology, American Memorial Hospital, Reims, France
| | - Maryline Poirée
- Department of Pediatric Hematology-Oncology, CHU Lenval, Nice, France
| | - Odile Minckes
- Department of Pediatric Hematology-Oncology, CHU, Caen, France
| | - Frédéric Millot
- Pediatric Oncology Unit, University Hospital, Poitiers, France
| | - Dominique Plantaz
- Department of Pediatric Oncology, University Hospital, Grenoble, France
| | - Philip Maes
- Department of Pediatrics, University Hospital Antwerp, Belgium
| | - Claire Hoyoux
- Department of Pediatrics, CHR de la Citadelle, Liège, Belgium
| | - Hélène Cavé
- Department of Genetics, Assistance Publique des Hôpitaux de Paris (AP-HP), Robert Debré Hospital, Paris, France
- INSERM UMR 1131, University Institute of Hematology, University Paris Diderot, Paris Sorbonne Cité, France
| | - Pierre Rohrlich
- Department of Pediatric Hematology-Oncology, CHU Lenval, Nice, France
| | - Yves Bertrand
- Institute of Pediatric Hematology and Oncology (IHOP), Hospices Civils de Lyon, and University Lyon 1, France
| | - Yves Benoit
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent University, Belgium
| |
Collapse
|
87
|
Nielsen SN, Eriksson F, Rosthoej S, Andersen MK, Forestier E, Hasle H, Hjalgrim LL, Aasberg A, Abrahamsson J, Heyman M, Jónsson ÓG, Pruunsild K, Vaitkeviciené GE, Vettenranta K, Schmiegelow K. Children with low-risk acute lymphoblastic leukemia are at highest risk of second cancers. Pediatr Blood Cancer 2017; 64. [PMID: 28500740 DOI: 10.1002/pbc.26518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/02/2017] [Accepted: 02/09/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The improved survival rates for childhood acute lymphoblastic leukemia (ALL) may be jeopardized by the development of a second cancer, which has been associated with thiopurine therapy. PROCEDURE We retrospectively analyzed three sequential Nordic Society of Paediatric Haematology and Oncology's protocols characterized by increasing intensity of thiopurine-based maintenance therapy. We explored the risk of second cancer in relation to protocols, risk group, thiopurine methyltransferase (TPMT) activity, ALL high hyperdiploidy (HeH), and t(12;21)[ETV6/RUNX1]. RESULTS After median 9.5 years (interquartile range, 5.4-15.3 yrs) of follow-up, 40 of 3,591 patients had developed a second cancer, of whom 38 had non-high-risk B-cell precursor ALL. Patients with standard-risk ALL, who received the longest maintenance therapy, had the highest adjusted hazard of second cancer (hazard ratio [HR], intermediate vs. standard risk: 0.16, 95% CI: 0.06-0.43, P < 0.001; HR, high vs. standard risk: 0.09, 95% CI: 0.02-0.49, P = 0.006); no significant effects of protocol, age, or white blood cell count at diagnosis, ALL HeH, or t(12;21)[ETV6/RUNX1] were observed. A subset analysis on the patients with standard-risk ALL did not show an increased hazard of second cancer from either HeH or t(12;21) (adjusted HR 2.02, 95% CI: 0.69-5.96, P = 0.20). The effect of low TPMT low activity was explored in patients reaching maintenance therapy in clinical remission (n = 3,368); no association with second cancer was observed (adjusted HR 1.43, 95% CI: 0.54-3.76, P = 0.47). CONCLUSIONS The rate of second cancer was generally highest in patients with low-risk ALL, but we could not identify a subset at higher risk than others.
Collapse
Affiliation(s)
- Stine N Nielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Frank Eriksson
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Rosthoej
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Mette K Andersen
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
| | - Erik Forestier
- Department of Medical Biosciences, Clinical Genetics, Umeå University, Umeå, Sweden
| | - Henrik Hasle
- Department of Paediatrics, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Lisa L Hjalgrim
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ann Aasberg
- Department of Paediatrics, University Hospital of Trondheim, Trondheim, Norway
| | - Jonas Abrahamsson
- Department of Pediatrics, Institution for Clinical Sciences Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mats Heyman
- Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Ólafur G Jónsson
- Pediatric Hematology-Oncology, Children's Hospital, Barnaspitali Hringsins, Landspitali University Hospital, Reykjavik, Iceland
| | - Kaie Pruunsild
- Department of Oncology and Hematology, Tallinn Children's Hospital, Tallinn, Estonia
| | - Goda E Vaitkeviciené
- Centre for Paediatric Oncology and Haematology, University Children's Hospital, Vilnius, Lithuania
| | - Kim Vettenranta
- Department of Paediatrics, University of Tampere, Tampere, Finland
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
88
|
Wolthers BO, Frandsen TL, Baruchel A, Attarbaschi A, Barzilai S, Colombini A, Escherich G, Grell K, Inaba H, Kovacs G, Liang DC, Mateos M, Mondelaers V, Möricke A, Ociepa T, Samarasinghe S, Silverman LB, van der Sluis IM, Stanulla M, Vrooman LM, Yano M, Zapotocka E, Schmiegelow K. Asparaginase-associated pancreatitis in childhood acute lymphoblastic leukaemia: an observational Ponte di Legno Toxicity Working Group study. Lancet Oncol 2017; 18:1238-1248. [PMID: 28736188 DOI: 10.1016/s1470-2045(17)30424-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Survival for childhood acute lymphoblastic leukaemia surpasses 90% with contemporary therapy; however, patients remain burdened by the severe toxic effects of treatment, including asparaginase-associated pancreatitis. To investigate the risk of complications and risk of re-exposing patients with asparaginase-associated pancreatitis to asparaginase, 18 acute lymphoblastic leukaemia trial groups merged data for this observational study. METHODS Patient files from 26 trials run by 18 trial groups were reviewed on children (aged 1·0-17·9 years) diagnosed with t(9;22)-negative acute lymphoblastic leukaemia between June 1, 1996, and Jan 1, 2016, who within 50 days of asparaginase exposure developed asparaginase-associated pancreatitis. Asparaginase-associated pancreatitis was defined by at least two criteria: abdominal pain, pancreatic enzymes at least three times the upper limit of normal (ULN), and imaging compatible with pancreatitis. Patients without sufficient data for diagnostic criteria were excluded. Primary outcomes were defined as acute and persisting complications of asparaginase-associated pancreatitis and risk of re-exposing patients who suffered an episode of asparaginase-associated pancreatitis to asparaginase. Data were collected from Feb 2, 2015, to June 30, 2016, and analysed and stored in a common database at Rigshospitalet, Copenhagen, Denmark. FINDINGS Of 465 patients with asparaginase-associated pancreatitis, 33 (8%) of 424 with available data needed mechanical ventilation, 109 (26%) of 422 developed pseudocysts, acute insulin therapy was needed in 81 (21%) of 393, and seven (2%) of 458 patients died. Risk of assisted mechanical ventilation, need for insulin, pseudocysts, or death was associated with older age (median age for patients with complications 10·5 years [IQR 6·4-13·8] vs without complications 6·1 years [IQR 3·6-12·2], p<0·0001), and having one or more affected vital signs (fever, hypotension, tachycardia, or tachypnoea; 96 [44%] of 217 patients with affected vital signs vs 11 [24%] of 46 patients without affected vital signs, p=0·02). 1 year after diagnosis of asparaginase-associated pancreatitis, 31 (11%) of 275 patients still needed insulin or had recurrent abdominal pain or both. Both the risk of persisting need for insulin therapy and recurrent abdominal pain were associated with having had pseudocysts (odds ratio [OR] 9·48 [95% CI 3·01-35·49], p=0·0002 for insulin therapy; OR 11·79 [4·30-37·98], p<0·0001 for recurrent abdominal pain). Within 8 years of asparaginase-associated pancreatitis, risk of abdominal symptoms dropped from 8% (26 of 312) to 0% (0 of 35) but the need for insulin therapy remained constant (9%, three of 35). 96 patients were re-exposed to asparaginase, including 59 after a severe asparaginase-associated pancreatitis (abdominal pain or pancreatic enzymes at least three times the ULN or both lasting longer than 72 h). 44 (46%) patients developed a second asparaginase-associated pancreatitis, 22 (52%) of 43 being severe. Risk of persisting need for insulin or abdominal pain after having had two versus one asparaginase-associated pancreatitis did not differ (three [7%] of 42 vs 28 [12%] of 233, p=0·51). Risk of a second asparaginase-associated pancreatitis was not associated with any baseline patient characteristics. INTERPRETATION Since the risk of a second asparaginase-associated pancreatitis was not associated with severity of the first asparaginase-associated pancreatitis and a second asparaginase-associated pancreatitis did not involve an increased risk of complications, asparaginase re-exposure should be determined mainly by the anticipated need for asparaginase for antileukaemic efficacy. A study of the genetic risk factors identifying patients in whom asparaginase exposure should be restricted is needed. FUNDING The Danish Childhood Cancer Foundation and The Danish Cancer Society (R150-A10181).
Collapse
Affiliation(s)
- Benjamin O Wolthers
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Thomas L Frandsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - André Baruchel
- Pediatric Hematology-Immunology Department, University Hospital Robert Debré, Paris Diderot University, Paris, France
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St Anna Children's Hospital and Department of Pediatric and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Shlomit Barzilai
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Antonella Colombini
- Department of Pediatrics, Ospedale San Gerardo, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Gabriele Escherich
- University Medical Center Eppendorf, Clinic of Pediatric Hematology and Oncology, Hamburg, Germany
| | - Kathrine Grell
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Hiroto Inaba
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Gábor Kovacs
- Second Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Der-Cherng Liang
- Division of Pediatric Hematology-Oncology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Marion Mateos
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, and School of Women and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Veerle Mondelaers
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Anja Möricke
- Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Department of Pediatrics, Kiel, Germany
| | - Tomasz Ociepa
- Department of Pediatrics, Hematology and Oncology, Pomeranian Medical University, Szczecin, Poland
| | | | - Lewis B Silverman
- Department of Pediatric Oncology, Dana Farber Cancer Institute and Division of Pediatric Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Inge M van der Sluis
- Dutch Childhood Oncology Group, The Hague, Erasmus Medical Center, Sophia Children's Hospital, Department of Pediatric Hematology-Oncology, Rotterdam, Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Martin Stanulla
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Lynda M Vrooman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michihiro Yano
- Department of Pediatrics, Akita University Hospital, Akita, Japan
| | - Ester Zapotocka
- University Hospital Motol, Department of Pediatric Hematology/Oncology, Prague, Czech Republic
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
89
|
Management of adults with T-cell lymphoblastic leukemia. Blood 2017; 129:1134-1142. [DOI: 10.1182/blood-2016-07-692608] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/27/2016] [Indexed: 12/17/2022] Open
Abstract
Abstract
T-cell acute lymphoblastic leukemia (ALL) is a rare disease in adults with inferior survival outcomes compared with those seen in pediatric patients. Although potentially curable with ∼50% survival at 5 years, adult patients with relapsed disease have dismal outcomes with <10% of patients surviving long term. This review will discuss the diagnosis and management of adult patients with newly diagnosed T-cell ALL with an emphasis on the immunophenotypic and genetic analyses required to assign prognosis, risk stratify, and guide post-remission therapy. The evidence for the main components of complex T-cell ALL treatment regimens is described. The importance of monitoring minimal residual disease is emphasized, with a discussion of the different methods used. The results of hematopoietic cell transplantation are analyzed, and recommendations made about which patients should be considered for this intervention. The treatment of the adolescent and young adult group is delineated, and the role of using “pediatric-inspired” regimens in older adults considered. We also describe the current data and potential future options for the use of novel therapies, including nelarabine and γ-secretase inhibitors, in adult patients with T-cell ALL.
Collapse
|
90
|
Talamo L, Douvas M, Macik BG, Ornan D. Successful treatment with apixaban of sinus venous thrombosis due to pegylated asparaginase in a young adult with T cell acute lymphoblastic leukemia: case report and review of management. Ann Hematol 2017; 96:691-693. [PMID: 28132079 PMCID: PMC5334383 DOI: 10.1007/s00277-017-2930-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/17/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Laura Talamo
- Department of Internal Medicine, Division of Hematology and Oncology, University of Virginia School of Medicine, PO Box 800716, Charlottesville, VA, 22908-0716, USA.
| | - Michael Douvas
- Department of Internal Medicine, Division of Hematology and Oncology, University of Virginia School of Medicine, PO Box 800716, Charlottesville, VA, 22908-0716, USA
| | - B Gail Macik
- Department of Internal Medicine, Division of Hematology and Oncology, University of Virginia School of Medicine, PO Box 800716, Charlottesville, VA, 22908-0716, USA
| | - David Ornan
- Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
91
|
Do immunoglobulin G and immunoglobulin E anti-l-asparaginase antibodies have distinct implications in children with acute lymphoblastic leukemia? A cross-sectional study. Rev Bras Hematol Hemoter 2017; 39:202-209. [PMID: 28830598 PMCID: PMC5567415 DOI: 10.1016/j.bjhh.2016.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/18/2016] [Accepted: 11/28/2016] [Indexed: 11/29/2022] Open
Abstract
Background l-Asparaginase is essential in the treatment of childhood acute lymphoblastic leukemia. If immunoglobulin G anti-l-asparaginase antibodies develop, they can lead to faster plasma clearance and reduced efficiency as well as to hypersensitivity reactions, in which immunoglobulin E can also participate. This study investigated the presence of immunoglobulin G and immunoglobulin E anti-l-asparaginase antibodies and their clinical associations. Methods Under 16-year-old patients at diagnosis of B-cell acute lymphoblastic leukemia confirmed by flow cytometry and treated with a uniform l-asparaginase and chemotherapy protocol were studied. Immunoglobulin G anti-l-asparaginase antibodies were measured using an enzyme-linked immunosorbent assay. Intradermal and prick skin testing was performed to establish the presence of specific immunoglobulin E anti-l-asparaginase antibodies in vivo. Statistical analysis was used to investigate associations of these antibodies with relevant clinical events and outcomes. Results Fifty-one children were studied with 42 (82.35%) having anti-l-asparaginase antibodies. In this group immunoglobulin G antibodies alone were documented in 10 (23.8%) compared to immunoglobulin E alone in 18 (42.8%) patients. Immunoglobulin G together with immunoglobulin E were simultaneously present in 14 patients. Children who produced exclusively immunoglobulin G or no antibodies had a lower event-free survival (p-value = 0.024). Eighteen children (35.3%) relapsed with five of nine of this group who had negative skin tests suffering additional relapses (range: 2–4), compared to none of the nine children who relapsed who had positive skin tests (p-value < 0.001). Conclusion Children with acute lymphoblastic leukemia and isolated immunoglobulin G anti-l-asparaginase antibodies had a higher relapse rate, whereas no additional relapses developed in children with immunoglobulin E anti-l-asparaginase antibodies after the first relapse.
Collapse
|
92
|
McNeer JL, Bleyer A, Conter V, Stock W. Acute Lymphoblastic Leukemia. CANCER IN ADOLESCENTS AND YOUNG ADULTS 2017. [DOI: 10.1007/978-3-319-33679-4_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
93
|
Use of PEG-asparaginase in newly diagnosed adults with standard-risk acute lymphoblastic leukemia compared with E. coli-asparaginase: a retrospective single-center study. Sci Rep 2016; 6:39463. [PMID: 28000713 PMCID: PMC5175128 DOI: 10.1038/srep39463] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/22/2016] [Indexed: 02/04/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous disease, and the long-term survival varies with different ages. We performed a retrospective analysis of 122 newly diagnosed adults with standard-risk ALL treated with Escherichia coli asparaginase (E. coli-asparaginase, n = 50) and polyethylene glycol-conjugated asparaginase (PEG-asparaginase, n = 72). No treatment-related mortality (TRM) occurred in the E. coli-asparaginase group, and 3 TRM events occurred in the PEG-asparaginase group without relation to asparaginase. In addition, 22 (44.0%) and 48 (66.7%) patients achieved a complete response (CR) on day 14 in the E. coli-asparaginase and PEG-asparaginase groups, respectively (P = 0.032). No different 5-year event-free survival (EFS) or overall survival (OS) rate (P = 0.632 and 0.769) was observed. Multivariate analysis revealed later CR (P = 0.008) and older age (P = 0.049) as adverse prognostic factors for both EFS and OS. In addition, we specifically monitored the known adverse effects of asparaginase, and no asparaginase-related death was observed. Allergy occurred in 9 patients using E. coli-asparaginase, and no patient in the PEG-asparaginase group suffered from allergies (P < 0.001). The incidence of other asparaginase-related toxicities was similar. We conclude that PEG-asparaginase can be safely and effectively used as asparaginase in adults with newly diagnosed standard-risk ALL.
Collapse
|
94
|
Kloos RQH, Pieters R, Escherich G, van der Sluis IM. Allergic-like reactions to asparaginase: Atypical allergies without asparaginase inactivation. Pediatr Blood Cancer 2016; 63:1928-34. [PMID: 27376478 DOI: 10.1002/pbc.26123] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/19/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Asparaginase is an important component of pediatric acute lymphoblastic leukemia (ALL) therapy. Unfortunately, this treatment is hampered by hypersensitivity reactions. In general, allergies - regardless of severity - cause complete inactivation of the drug. However, we report atypical allergic reactions without inactivation of asparaginase, here called allergic-like reactions. PROCEDURE Patients with an allergic-like reaction, who were treated according to the Dutch Childhood Oncology Group ALL-11 or the CoALL 08-09 protocol, were described. The reactions were identified by continual measurement of asparaginase activity levels. Characteristics, including timing of occurrence, symptoms, grade, and the presence of antiasparaginase antibodies, were compared to those of real allergies. RESULTS Fourteen allergic-like reactions occurred in nine patients. Five reactions were to PEGasparaginase and nine to Erwinia asparaginase. Allergic-like reactions occurred relatively late after the start of infusion compared to real allergies. Antibodies were absent in all but one patient with an allergic-like reaction, while they were detected in all patients with a real allergy. Symptoms and grade did not differ between the groups. Asparaginase was continued with the same formulation in six patients of whom four finished treatment with adequate activity levels. CONCLUSIONS In conclusion, allergic-like reactions occur relatively late after the start of infusion and without antibodies. Despite these clinical differences, allergic-like reactions can only be distinguished from real allergies by continually measuring asparaginase activity levels. If clinically tolerated, formulations should not be switched in case of allergic-like reactions. Moreover, failure to recognize these reactions may lead to a less favorable prognosis if asparaginase therapy is terminated unnecessarily.
Collapse
Affiliation(s)
- Robin Q H Kloos
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Gabriele Escherich
- Clinic of Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Inge M van der Sluis
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
95
|
Yu Q, Wang X, Wang L, Zheng J, Wang J, Wang B. Knockdown of asparagine synthetase (ASNS) suppresses cell proliferation and inhibits tumor growth in gastric cancer cells. Scand J Gastroenterol 2016; 51:1220-6. [PMID: 27251594 DOI: 10.1080/00365521.2016.1190399] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Asparagine synthetase (ASNS) gene encodes an enzyme that catalyzes the glutamine- and ATP-dependent conversion of aspartic acid to asparagine. ASNS is deemed as a promising therapeutic target and its expression is associated with the chemotherapy resistance in several human cancers. However, its role in gastric cancer tumorigenesis has not been investigated. METHODS In this study, we employed small interfering RNA (siRNA) to transiently knockdown ASNS in two gastric cancer cell lines, AGS and MKN-45, followed by growth rate assay and colony formation assay. Dose response curve analysis was performed in AGS and MKN-45 cells with stable ASNS knockdown to assess sensitivity to cisplatin. Xenograft experiment was performed to examine in vivo synergistic effects of ASNS depletion and cisplatin on tumor growth. Expression level of ASNS was evaluated in human patient samples using quantitative PCR. Kaplan-Meier curve analysis was performed to evaluate association between ASNS expression and patient survival. RESULTS Transient knockdown of ASNS inhibited cell proliferation and colony formation in AGS and MKN-45 cells. Stable knockdown of ASNS conferred sensitivity to cisplatin in these cells. Depletion of ASNS and cisplatin treatment exerted synergistic effects on tumor growth in AGS xenografts. Moreover, ASNS was found to be up-regulated in human gastric cancer tissues compared with matched normal colon tissues. Low expression of ASNS was significantly associated with better survival in gastric cancer patients. CONCLUSION ASNS may contribute to gastric cancer tumorigenesis and may represent a novel therapeutic target for prevention or intervention of gastric cancer.
Collapse
Affiliation(s)
- Qingxiang Yu
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Xiaoyu Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Li Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Jia Zheng
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Jiang Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Bangmao Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| |
Collapse
|
96
|
Horvat TZ, Pecoraro JJ, Daley RJ, Buie LW, King AC, Rampal RK, Tallman MS, Park JH, Douer D. The use of Erwinia asparaginase for adult patients with acute lymphoblastic leukemia after pegaspargase intolerance. Leuk Res 2016; 50:17-20. [PMID: 27631159 DOI: 10.1016/j.leukres.2016.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/01/2016] [Accepted: 08/26/2016] [Indexed: 11/28/2022]
Abstract
Asparaginase administration has become a crucial component of front-line pediatric and pediatric-insipired multi-agent regimens for the treatment of acute lymphoblastic leukemia (ALL). The aim of this retrospective study was to assess the safety and feasibility of switching to Erwinia asparaginase after pegaspargase intolerance in adult ALL patients treated at Memorial Sloan Kettering Cancer Center. Our analysis included 10 patients, with a median age of 39 years (range 20-72), male predominance (90%), and a typical B-cell to T-cell ratio (70:30%) for ALL. Nine patients were switched to Erwinia asparaginase after pegaspargase hypersensitivity and one patient after grade 4 hyperbilirubinemia secondary to pegaspargase. With Erwinia asparaginase, no hypersensitivity reactions occurred and no patient developed other known clinical asparaginase-related toxicities. Laboratory adverse effects consisted of mostly mild elevation in liver enzymes. No morphologic relapses have occurred in any patient switched to Erwinia asparaginase in first remission at a follow up of 0.4-34.6 months. These findings are unique in that all of our patients received Erwinia asparaginase after hypersensitivity or intolerance to pegaspargase and 50% of them were older than 40 years of age, a population with very limited Erwinia asparaginase data. Our observations provide preliminary information that treatment with Erwinia asparaginase can proceed as scheduled in adult patients, despite pegaspargase hypersensitivity and possibly liver intolerance.
Collapse
Affiliation(s)
- Troy Z Horvat
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Joshua J Pecoraro
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Ryan J Daley
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Larry W Buie
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Amber C King
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Raajit K Rampal
- Leukemia Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| | - Martin S Tallman
- Leukemia Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| | - Jae H Park
- Leukemia Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| | - Dan Douer
- Leukemia Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
97
|
Rodriguez V, Kairalla J, Salzer WL, Raetz EA, Loh ML, Carroll AJ, Heerema NA, Wood BL, Borowitz MJ, Burke MJ, Asselin BL, Devidas M, Winick NJ, Carroll WL, Hunger SP, Dreyer ZE. A Pilot Study of Intensified PEG-Asparaginase in High-risk Acute Lymphoblastic Leukemia: Children's Oncology Group Study AALL08P1. J Pediatr Hematol Oncol 2016; 38:409-17. [PMID: 27299599 PMCID: PMC4955695 DOI: 10.1097/mph.0000000000000589] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
AALL08P1 was designed to determine whether biweekly intensified pegaspargase (I-PEG) was feasible and safe in pediatric patients with newly diagnosed high-risk B-precursor lymphoblastic leukemia when given with Children's Oncology Group hemiaugmented BFM therapy. High-risk average (HR-Avg) patients received standard pegaspargase dosing (6 doses), whereas high-risk high (HR-High) patients received I-PEG biweekly from the start of Consolidation until day 1 of Maintenance. Feasibility and safety were defined in advance as ≥65% of patients tolerating at least 8 doses of I-PEG and 90% requiring ≤49 weeks from day 1 of Consolidation to the initiation of Maintenance. Targeted toxicities included allergic reactions, anaphylaxis, pancreatitis, thrombosis, bleeding, central nervous system events, and sepsis. AALL08P1 enrolled 104 patients; 54 were classified as HR-Avg and 30 as HR-High after completion of induction therapy. Only 53% (16/30) of the HR-High patients received ≥8 total doses of I-PEG and 50% (15/30) took ≤49 weeks from start of Consolidation to the initiation of Maintenance. I-PEG did not significantly increase grade 2 to 5 targeted toxicities. I-PEG was not feasible or safe as defined in AALL08P1. Complete assessment of this regimen was limited due to removal of patients from I-PEG regimen and early closure of the study.
Collapse
Affiliation(s)
- Vilmarie Rodriguez
- *Mayo Clinic Children's Center, Division of Pediatric Hematology Oncology, Rochester, MN †Children's Oncology Group Data Center Biostatistics, University of Florida, Gainesville, FL ‡Walter Reed National Military Medical Center, Congressionally Directed Medical Research Program, US Army Medical Research and Materiel Command, Fort Detrick, MD ††Department of Pathology, Johns Hopkins University Sidney Kimmel Cancer Center, John Hopkins University, Baltimore, MD §Division Pediatric Hematology-Oncology, Primary Children's Hospital, University of Utah, Salt Lake City, UT ∥Division of Pediatric Hematology Oncology, UCSF Medical Center-Mssion Bay, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA ¶Department of Genetics, Children's Hospital of Alabama, University of Alabama at Birmingham, Birmingham, AL #Nationwide Children's Hospital, Cytogenetics, The Ohio State University, Columbus, OH **Seattle Children's Hospital, Laboratory Medicine, Seattle, WA ‡‡Midwest Children's Cancer Center, MACC Fund Research Center, Milwaukee, WI §§Division of Pediatric Hematology Oncology, University of Rochester Medical Center, Rochester ¶¶Laura and Isaac Perlmutter Cancer Center at NYU Langone, Hassendfeld Children's Center, New York, NY ∥∥Division Pediatric Hematology-Oncology, UT Southwestern/Simmons Cancer Center, Dallas, TX †††Baylor College of Medicine, Comprehensive Cancer Center, Pediatric Oncology, Houston, TX ##Division of Oncology, Children's Hospital of Philadelphia ***Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Wolthers BO, Frandsen TL, Abrahamsson J, Albertsen BK, Helt LR, Heyman M, Jónsson ÓG, Kõrgvee LT, Lund B, Raja RA, Rasmussen KK, Taskinen M, Tulstrup M, Vaitkevičienė GE, Yadav R, Gupta R, Schmiegelow K. Asparaginase-associated pancreatitis: a study on phenotype and genotype in the NOPHO ALL2008 protocol. Leukemia 2016; 31:325-332. [PMID: 27451978 DOI: 10.1038/leu.2016.203] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022]
Abstract
Asparaginase (ASP)-associated pancreatitis (AAP) occurs during acute lymphoblastic leukemia treatment. Among 1285 children (1.0-17.9 years) diagnosed during July 2008-December 2014 and treated according to the Nordic/Baltic ALL2008 protocol, 86 (cumulative incidence=6.8%) developed AAP. Seventy-three cases were severe (diagnostic AAP criteria persisting >72 h) and 13 mild. Cases were older than controls (median: 6.5 vs 4.5 years; P=0.001). Pseudocysts developed in 28%. Of the 20 re-exposed to ASP, 9 (45%) developed a second AAP. After a median follow-up of 2.3 years, 8% needed permanent insulin therapy, and 7% had recurrent abdominal pain. Germline DNA on 62 cases and 638 controls was genotyped on Omni2.5exome-8-v1.2 BeadChip arrays. Overall, the ULK2 variant rs281366 showed the strongest association with AAP (P=5.8 × 10-7; odds ratio (OR)=6.7). Cases with the rs281366 variant were younger (4.3 vs 8 years; P=0.015) and had lower risk of AAP-related complications (15% vs 43%; P=0.13) compared with cases without this variant. Among 45 cases and 517 controls <10 years, the strongest associations with AAP were found for RGS6 variant rs17179470 (P=9.8 × 10-9; OR=7.3). Rs281366 is located in the ULK2 gene involved in autophagy, and RGS6 regulates G-protein signaling regulating cell dynamics. More than 50% of AAP cases <10 years carried one or both risk alleles.
Collapse
Affiliation(s)
- B O Wolthers
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - T L Frandsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - J Abrahamsson
- Department of Clinical Sciences, Queen Silvia's Children's Hospital, Gothenburg, Sweden
| | - B K Albertsen
- Department of Paediatrics, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - L R Helt
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - M Heyman
- Department of Pediatrics, Astrid Lindgrens Hospital, Stockholm, Sweden
| | - Ó G Jónsson
- Children's Hospital, Landspitali University Hospital, Reykjavík, Iceland
| | | | - B Lund
- Department of Paediatrics, St Olavs University Hospital, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, Medical Faculty, The Norwegian University of Science and Technology, Trondheim, Norway
| | - R A Raja
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - K K Rasmussen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - M Taskinen
- Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - M Tulstrup
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - G E Vaitkevičienė
- Clinic of Children's Diseases, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - R Yadav
- Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark
| | - R Gupta
- Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark
| | - K Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
99
|
Egler RA, Ahuja SP, Matloub Y. L-asparaginase in the treatment of patients with acute lymphoblastic leukemia. J Pharmacol Pharmacother 2016; 7:62-71. [PMID: 27440950 PMCID: PMC4936081 DOI: 10.4103/0976-500x.184769] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a hematologic malignancy that predominantly occurs in children between 2 and 10 years of age. L-asparaginase is an integral component of treatment for patients with ALL and since its introduction into pediatric treatment protocols in the 1960s, survival rates in children have progressively risen to nearly 90%. Outcomes for adolescent and young adult (AYA) patients, aged 15-39 years and diagnosed with ALL, have historically been less favorable. However, recent reports suggest substantially increased survival in AYA patients treated on pediatric-inspired protocols that include a greater cumulative dose of asparaginase. All currently available asparaginases share the same mechanism of action - the deamination and depletion of serum asparagine levels - yet each displays a markedly different pharmacokinetic profile. Pegylated asparaginase derived from the bacterium Escherichia coli is used as first-line therapy; however, up to 30% of patients develop a treatment-limiting hypersensitivity reaction. Patients who experience a hypersensitivity reaction to an E. coli-derived asparaginase can continue treatment with Erwinia chrysanthemi asparaginase. Erwinia asparaginase is immunologically distinct from E. coli-derived asparaginases and exhibits no cross-reactivity. Studies have shown that with adequate dosing, therapeutic levels of Erwinia asparaginase activity can be achieved, and patients switched to Erwinia asparaginase due to hypersensitivity can obtain outcomes similar to patients who do not experience a hypersensitivity reaction. Therapeutic drug monitoring may be required to ensure that therapeutic levels of asparaginase activity are maintained.
Collapse
Affiliation(s)
- Rachel A Egler
- Department of Pediatric Hematology/Oncology, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Sanjay P Ahuja
- Department of Pediatric Hematology/Oncology, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Yousif Matloub
- Department of Pediatric Hematology/Oncology, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| |
Collapse
|
100
|
Michaux K, Bergeron C, Gandemer V, Mechinaud F, Uyttebroeck A, Bertrand Y. Relapsed or Refractory Lymphoblastic Lymphoma in Children: Results and Analysis of 23 Patients in the EORTC 58951 and the LMT96 Protocols. Pediatr Blood Cancer 2016; 63:1214-21. [PMID: 27037853 DOI: 10.1002/pbc.25990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/25/2016] [Accepted: 03/06/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND The treatment of children with T-cell lymphoblastic lymphoma (T-LBL) and precursor B-cell lymphoblastic lymphoma (pB-LBL) has improved during the last decades. However, patients with relapsed or refractory lymphomas still have a poor prognosis. METHODS We report the characteristics and evolution of T-LBL and pB-LBL relapses in two multicenter prospective studies (LMT 96, European Organization for Research and Treatment of Cancer 58951). RESULTS From 1997 to 2008, 194 patients were included in these studies (157 T-LBL; 37 pB-LBL); among them, 23 patients underwent relapse or progression (18 T-LBL and 5 pB-LBL). The median age was 7.7 years (range 1.4-16.3). The survival rate at 8 years was 8.7% (21 deaths). The median time from diagnosis to relapse was 9 months [1-69] and 11 months [1-45] for T-LBL and pB-LBL, respectively. Twenty-two patients received a second-line treatment but remission was achieved in only seven patients. In 10 patients, intensification with hematopoietic stem cell transplantation (HSCT) was performed and four of them had a second relapse. Two patients still alive had T-LBL, experienced relapses 15 and 69 months after diagnosis, and received HSCT. Relapse during the intensive phase and second-line treatment without HSCT were identified as risk factors for bad prognosis (P = 0.01). CONCLUSIONS The results of second-line treatment, including intensive chemotherapy and HSCT, show that salvage treatment is still disappointing in controlling refractory forms. Early identification of patients at high risk of relapse is mandatory, allowing earlier intensification. Valid prognostic parameters, such as biological markers, are needed. International cooperation is warranted to collect more data on these rare diagnoses.
Collapse
Affiliation(s)
- Katell Michaux
- Pediatric Hematology/Oncology Department (IHOP), Hospices Civils de Lyon and Centre Léon Bérard, Lyon, France
| | - Christophe Bergeron
- Pediatric Hematology/Oncology Department (IHOP), Hospices Civils de Lyon and Centre Léon Bérard, Lyon, France
| | - Virginie Gandemer
- Pediatric Hematology Department, University Hospital, Rennes, France
| | - Françoise Mechinaud
- Pediatric Hematology/Oncology Department, University Hospital, Nantes, France
| | - Anne Uyttebroeck
- Pediatric Hematology/Oncology Department, University Hospital Gasthuisberg, Leuven, Belgium
| | - Yves Bertrand
- Pediatric Hematology/Oncology Department (IHOP), Hospices Civils de Lyon and Centre Léon Bérard, Lyon, France.,Pediatric Hematology Department, University Hospital, Rennes, France.,Pediatric Hematology/Oncology Department, University Hospital, Nantes, France.,Pediatric Hematology/Oncology Department, University Hospital Gasthuisberg, Leuven, Belgium.,Claude Bernard Lyon 1 University, Lyon, France
| | | |
Collapse
|