1
|
Larkin T, Kashif R, Elsayed AH, Greer B, Mangrola K, Raffiee R, Nguyen N, Shastri V, Horn B, Lamba JK. Polygenic Pharmacogenomic Markers as Predictors of Toxicity Phenotypes in the Treatment of Acute Lymphoblastic Leukemia: A Single-Center Study. JCO Precis Oncol 2023; 7:e2200580. [PMID: 36952646 PMCID: PMC10309546 DOI: 10.1200/po.22.00580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/31/2023] [Indexed: 03/25/2023] Open
Abstract
PURPOSE Acute lymphoblastic leukemia (ALL) is the most prevalent cause of childhood cancer and requires a long course of therapy consisting of three primary phases with interval intensification blocks. Although these phases are necessary to achieve remission, the primary chemotherapeutic agents have potentially serious toxicities, which may lead to delays or discontinuations of therapy. The purpose of this study was to perform a comprehensive pharmacogenomic evaluation of common antileukemic agents and develop a polygenic toxicity risk score predictive of the most common toxicities observed during ALL treatment. METHODS This cross-sectional study included 75 patients with pediatric ALL treated between 2012 and 2020 at the University of Florida. Toxicity data were collected within 100 days of initiation of therapy using CTCAE v4.0 for toxicity grading. For pharmacogenomic evaluation, single-nucleotide polymorphisms (SNPs) and genes were selected from previous reports or PharmGKB database. 116 unique SNPs were evaluated for incidence of various toxicities. A multivariable multi-SNP modeling for up to 3-SNP combination was performed to develop a polygenic toxicity risk score of prognostic value. RESULTS We identified several SNPs predictive of toxicity phenotypes in univariate analysis. Further multivariable SNP-SNP combination analysis suggest that susceptibility to chemotherapy-induced toxicities is likely multigenic in nature. For 3-SNPscore models, patients with high scores experienced increased risk of GI (P = 2.07E-05, 3 SNPs: TYMS-rs151264360/FPGS-rs1544105/GSTM1-GSTM5-rs3754446), neurologic (P = .0005, 3 SNPs: DCTD-rs6829021/SLC28A3-rs17343066/CTPS1-rs12067645), endocrine (P = 4.77E-08, 3 SNPs: AKR1C3-rs1937840/TYMS-rs2853539/CTH-rs648743), and heme toxicities (P = .053, 3 SNPs: CYP3A5-rs776746/ABCB1-rs4148737/CTPS1-rs12067645). CONCLUSION Our results imply that instead of a single-SNP approach, SNP-SNP combinations in multiple genes in drug pathways increases the robustness of prediction of toxicity. These results further provide promising SNP models that can help establish clinically relevant biomarkers allowing for greater individualization of cancer therapy to maximize efficacy and minimize toxicity for each patient.
Collapse
Affiliation(s)
- Trisha Larkin
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL
- St Joseph's Children's Hospital/BayCare Medical Group, Tampa, FL
| | - Reema Kashif
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL
| | - Abdelrahman H. Elsayed
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL
| | - Beate Greer
- Pediatrics Division, UF Health Cancer Center, University of Florida, Gainesville, FL
| | - Karna Mangrola
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL
| | - Roya Raffiee
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL
| | - Nam Nguyen
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL
| | - Vivek Shastri
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL
| | - Biljana Horn
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL
| | - Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL
| |
Collapse
|
2
|
Renatino Canevarolo R, Pereira de Souza Melo C, Moreno Cury N, Luiz Artico L, Ronchi Corrêa J, Tonhasca Lau Y, Sousa Mariano S, Reddy Sudalagunta P, Regina Brandalise S, Carolina de Mattos Zeri A, Andrés Yunes J. Glutathione levels are associated with methotrexate resistance in acute lymphoblastic leukemia cell lines. Front Oncol 2022; 12:1032336. [PMID: 36531023 PMCID: PMC9751399 DOI: 10.3389/fonc.2022.1032336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Methotrexate (MTX), a folic acid antagonist and nucleotide synthesis inhibitor, is a cornerstone drug used against acute lymphoblastic leukemia (ALL), but its mechanism of action and resistance continues to be unraveled even after decades of clinical use. Methods To better understand the mechanisms of this drug, we accessed the intracellular metabolic content of 13 ALL cell lines treated with MTX by 1H-NMR, and correlated metabolome data with cell proliferation and gene expression. Further, we validated these findings by inhibiting the cellular antioxidant system of the cells in vitro and in vivo in the presence of MTX. Results MTX altered the concentration of 31 out of 70 metabolites analyzed, suggesting inhibition of the glycine cleavage system, the pentose phosphate pathway, purine and pyrimidine synthesis, phospholipid metabolism, and bile acid uptake. We found that glutathione (GSH) levels were associated with MTX resistance in both treated and untreated cells, suggesting a new constitutive metabolic-based mechanism of resistance to the drug. Gene expression analyses showed that eight genes involved in GSH metabolism were correlated to GSH concentrations, 2 of which (gamma-glutamyltransferase 1 [GGT1] and thioredoxin reductase 3 [TXNRD3]) were also correlated to MTX resistance. Gene set enrichment analysis (GSEA) confirmed the association between GSH metabolism and MTX resistance. Pharmacological inhibition or stimulation of the main antioxidant systems of the cell, GSH and thioredoxin, confirmed their importance in MTX resistance. Arsenic trioxide (ATO), a thioredoxin inhibitor used against acute promyelocytic leukemia, potentiated MTX cytotoxicity in vitro in some of the ALL cell lines tested. Likewise, the ATO+MTX combination decreased tumor burden and extended the survival of NOD scid gamma (NSG) mice transplanted with patient-derived ALL xenograft, but only in one of four ALLs tested. Conclusion Altogether, our results show that the cellular antioxidant defense systems contribute to leukemia resistance to MTX, and targeting these pathways, especially the thioredoxin antioxidant system, may be a promising strategy for resensitizing ALL to MTX.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanca Tonhasca Lau
- Centro de Pesquisa Boldrini, Centro Infantil Boldrini, Campinas, SP, Brazil
| | | | - Praneeth Reddy Sudalagunta
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | | | - Ana Carolina de Mattos Zeri
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - José Andrés Yunes
- Centro de Pesquisa Boldrini, Centro Infantil Boldrini, Campinas, SP, Brazil,Medical Genetics Department, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil,*Correspondence: José Andrés Yunes,
| |
Collapse
|
3
|
Maternal Haplotypes in DHFR Promoter and MTHFR Gene in Tuning Childhood Acute Lymphoblastic Leukemia Onset-Latency: Genetic/Epigenetic Mother/Child Dyad Study (GEMCDS). Genes (Basel) 2019; 10:genes10090634. [PMID: 31443485 PMCID: PMC6770441 DOI: 10.3390/genes10090634] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
Childhood acute lymphoblastic leukemia (ALL) peaks around age 2–4, and in utero genetic epigenetic mother-fetus crosstalk might tune ALL onset during childhood life. Folate genes variably interact with vitamin status on ALL risk and prognosis. We investigated DHFR and MTHFR gene variants in 235 ALL children and their mothers to disclose their role in determining ALL onset age and survival. Pyrosequence of DHFR 19bp ins/del (rs70991108; W/D), MTHFR C677T (rs1801133; C>T), and MTHFR A1298C (rs1801131; A>C) was assessed in children and in 72% of mothers for dyad-analysis comparison. DHFR DD-children had delayed ALL onset compared to WW-children (7.5 ± 4.8 vs. 5.2 ± 3.7 years; P = 0.002) as well as MTHFR 1298 CC-children compared to AA-children (8.03 ± 4.8 vs. 5.78 ± 4.1 years; P = 0.006), and according to the strong linkage disequilibrium between MTHFR 677 T-allele and 1298C-allele, MTHFR TT-children showed early mean age of onset though not significant. Offspring of MTHFR 677 TT-mothers had earlier ALL onset compared to offspring of 677 CC-mothers (5.4 ± 3.3 vs. 7 ± 5.3 years; P = 0.017). DHFR/MTHFR 677 polymorphism combination influenced onset age by comparing DD/CC vs. WW/TT children (8.1 ± 5.7 vs. 4.7 ± 2.1 years; P = 0.017). Moreover, mother-child genotype combination gave 5.5-years delayed onset age in favor of DD-offspring of 677 CC-mothers vs. WW-offspring of 677 TT-mothers, and it was further confirmed including any D-carrier children and any 677 T-carrier mothers (P = 0.00052). Correction for multiple comparisons maintained statistical significance for DHFR ins/del and MTHFR A1298C polymorphisms. Unexpectedly, among the very-early onset group (<2.89 years; 25th), DD-genotype inversely clustered in children and mothers (4.8% vs. 23.8% respectively), and accordingly ALL offspring of homozygous DD-mothers had increased risk to have early-onset (adjusted OR (odds ratio) = 3.08; 1.1–8.6; P = 0.03). The opposite effect DHFR promoter variant has in tuning ALL onset-time depending on who is the carrier (i.e., mother or child) might suggest a parent-origin-effect of the D-allele or a two-faced epigenetic role driven by unbalanced folate isoform availability during the in-utero leukemogenesis responsible for the wide postnatal childhood ALL latency.
Collapse
|
4
|
Fesenko DO, Avdonina MA, Gukasyan LG, Surzhikov SA, Chudinov AV, Zasedatelev AS, Nasedkina TV. Multiplex Genotyping of Allelic Variants of Genes Involved in Metabolizing Antileukemic Drugs. Mol Biol 2018. [DOI: 10.1134/s0026893318020036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5
|
Joob B, Wiwanitkit V. SLC19A1 Polymorphism and Serum Methotrexate in Patients with Acute Lymphoblastic Leukemia. Indian J Med Paediatr Oncol 2018. [DOI: 10.4103/ijmpo.ijmpo_6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
AbstractAcute lymphoblastic leukemia (ALL) is a common pediatric malignancy. Methotrexate is the widely used chemotherapy for ALL. The polymorphism (rs1051296) of SLC19A1 is proposed for its effect on serum methotrexate. To explain this observation, the authors hereby studied the interrelationship between SLC19A1 polymorphism and blood methotrexate level in the patients with ALL. Here, the authors use a quantum chemistry analysis for explaining of this observation.
Collapse
Affiliation(s)
- Beuy Joob
- Sanitation 1 Medical Academic Center, Bangkok, Thailand
| | - Viroj Wiwanitkit
- Department of Tropical Medicine, Hainan Medical University, Haikou, China; Department of Medicine, Faculty of Medicine, University of Nis, Nis, Serbia; Department of Biological Science, Joseph Ayobabalola University, Ilara-Mokin, Nigeria; Department of Community Medicine, Dr. DY Patil Medical University, Pune, Maharashtra, India
| |
Collapse
|
6
|
Lopez-Santillan M, Iparraguirre L, Martin-Guerrero I, Gutierrez-Camino A, Garcia-Orad A. Review of pharmacogenetics studies of L-asparaginase hypersensitivity in acute lymphoblastic leukemia points to variants in the GRIA1 gene. Drug Metab Pers Ther 2017; 32:1-9. [PMID: 28259867 DOI: 10.1515/dmpt-2016-0033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/07/2017] [Indexed: 12/17/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is a major pediatric cancer in developed countries. Although treatment outcome has improved owing to advances in chemotherapy, there is still a group of patients who experience severe adverse events. L-Asparaginase is an effective antineoplastic agent used in chemotherapy of ALL. Despite its indisputable indication, hypersensitivity reactions are common. In those cases, discontinuation of treatment is usually needed and anti-asparaginase antibody production may also attenuate asparaginase activity, compromising its antileukemic effect. Till now, six pharmacogenetic studies have been performed in order to elucidate possible genetic predisposition for inter-individual differences in asparaginase hypersensitivity. In this review we have summarized the results of those studies which describe the involvement of four different genes, being polymorphisms in the glutamate receptor, ionotropic, AMPA 1 (GRIA1) the most frequently associated with asparaginase hypersensitivity. We also point to new approaches focusing on epigenetics that could be interesting for consideration in the near future.
Collapse
|
7
|
Gervasini G, de Murillo SG, Jiménez M, de la Maya MD, Vagace JM. Effect of polymorphisms in transporter genes on dosing, efficacy and toxicity of maintenance therapy in children with acute lymphoblastic leukemia. Gene 2017; 628:72-77. [DOI: 10.1016/j.gene.2017.07.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/21/2017] [Accepted: 07/10/2017] [Indexed: 12/17/2022]
|
8
|
Marcoux S, Drouin S, Laverdière C, Alos N, Andelfinger GU, Bertout L, Curnier D, Friedrich MG, Kritikou EA, Lefebvre G, Levy E, Lippé S, Marcil V, Raboisson MJ, Rauch F, Robaey P, Samoilenko M, Séguin C, Sultan S, Krajinovic M, Sinnett D. The PETALE study: Late adverse effects and biomarkers in childhood acute lymphoblastic leukemia survivors. Pediatr Blood Cancer 2017; 64. [PMID: 27917589 DOI: 10.1002/pbc.26361] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/27/2016] [Accepted: 10/17/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND Childhood cancer survivorship issues represent an established public health challenge. Most late adverse effects (LAEs) have been demonstrated to be time and treatment dependent. The PETALE study is a multidisciplinary research project aiming to comprehensively characterize LAEs and identify associated predictive biomarkers in childhood acute lymphoblastic leukemia (cALL) survivors. METHODS cALL survivors treated at Sainte-Justine University Health Center with Dana-Farber Cancer Institution-ALL protocols 87-01 through 2005-01 were eligible. During Phase I of the study, the participants underwent comprehensive clinical, biologic, and psychosocial investigation targeting metabolic syndrome, cardiotoxicity, bone morbidity, neurocognitive problems, and quality of life issues. Whole-exome sequencing was performed for all participants. Subjects identified with an extreme phenotype during Phase I were recalled for additional testing (Phase II). RESULTS Phase I included 246 survivors (recall rate 71.9%). Of those, 85 participants completed Phase II (recall rate 88.5%). Survivors agreeing to participate in Phase I (n = 251) were similar to those who refused (n = 31) in terms of relapse risk profile, radiotherapy exposure, and age at the time of study. Participants, however, tended to be slightly older at diagnosis (6.1 vs. 4.7 years old, P = 0.08), with a higher proportion of female agreeing to participate compared with males (93.2 vs. 86.5%, P = 0.07). CONCLUSION The PETALE study will contribute to comprehensively characterize clinical, psychosocial, biologic, and genomic features of cALL survivors using an integrated approach. Expected outcomes include LAE early detection biomarkers, long-term follow-up guidelines, and recommendations for physicians and health professionals.
Collapse
Affiliation(s)
- Sophie Marcoux
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Simon Drouin
- Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Caroline Laverdière
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Nathalie Alos
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Gregor U Andelfinger
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Laurence Bertout
- Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Daniel Curnier
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | | | | | | | - Emile Levy
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Sarah Lippé
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Valérie Marcil
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Marie-Josée Raboisson
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Frank Rauch
- Shriners Hospitals for Children, Montréal, Québec, Canada
| | - Philippe Robaey
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada.,Children's Hospital of Eastern Ontario (CHEO), Ottawa, Ontario, Canada
| | | | - Chantal Séguin
- McGill University Health Center Research Institute, Montréal, Québec, Canada
| | - Serge Sultan
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Maja Krajinovic
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Daniel Sinnett
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| |
Collapse
|
9
|
Krull KR, Cheung YT, Liu W, Fellah S, Reddick WE, Brinkman TM, Kimberg C, Ogg R, Srivastava D, Pui CH, Robison LL, Hudson MM. Chemotherapy Pharmacodynamics and Neuroimaging and Neurocognitive Outcomes in Long-Term Survivors of Childhood Acute Lymphoblastic Leukemia. J Clin Oncol 2016; 34:2644-53. [PMID: 27269941 DOI: 10.1200/jco.2015.65.4574] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To examine associations among methotrexate pharmacodynamics, neuroimaging, and neurocognitive outcomes in long-term survivors of childhood acute lymphoblastic leukemia treated on a contemporary chemotherapy-only protocol. PATIENTS AND METHODS This longitudinal study linked pharmacokinetic assays collected during therapy to neurocognitive and brain imaging outcomes during long-term follow-up. A total of 218 (72.2%) of 302 eligible long-term survivors were recruited for outcome studies when they were more than 5 years post-diagnosis and older than 8 years of age. At long-term follow-up, survivors were an average of 13.8 years old and 7.7 years from diagnosis, and 51% were male. Neurocognitive testing, functional magnetic resonance imaging (MRI) during an executive function task, and structural MRI with diffusion tensor imaging were conducted. Generalized linear models were developed to identify predictors, and models were adjusted for age at diagnosis, sex, and parent education. RESULTS Intelligence was within normal limits (mean, 98; standard deviation, 14) compared with population expectations (mean, 100; standard deviation, 15), though measures of executive function, processing speed, and memory were less than population means (all P < .02 after correction for false discovery rates). Higher plasma concentration of methotrexate was associated with a poorer executive function score (P < .02). Higher plasma methotrexate was also associated with higher functional MRI activity, with thicker cortices in dorsolateral prefrontal brain regions, and with white matter microstructure in the frontostriatal tact. Neurocognitive impairment was associated with these imaging findings as well. Associations did not change after adjustment for age or dose of leucovorin rescue. CONCLUSION Survivors of childhood acute lymphoblastic leukemia treated on contemporary chemotherapy-only protocols demonstrate executive dysfunction. A higher plasma concentration of methotrexate was associated with executive dysfunction as well as with a thicker cortex and higher activity in frontal brain regions, regions often associated with executive function.
Collapse
Affiliation(s)
- Kevin R Krull
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | - Yin Ting Cheung
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | - Wei Liu
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | - Slim Fellah
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | | | - Tara M Brinkman
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | - Cara Kimberg
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | - Robert Ogg
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | | | - Ching-Hon Pui
- All authors: St Jude Children's Research Hospital, Memphis, TN
| | | | | |
Collapse
|
10
|
Cole PD. Does genetic susceptibility increase risk for neurocognitive decline among patients with acute lymphoblastic leukemia? Future Oncol 2015; 11:1855-8. [PMID: 26161922 DOI: 10.2217/fon.15.117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Peter D Cole
- Albert Einstein College of Medicine, 1225 Morris Park Avenue, Van Etten Building, Room VE 6A03, Bronx, NY 10461, USA.,The Children's Hospital at Montefiore, 3415 Bainbridge Avenue, Bronx, NY 10467, USA
| |
Collapse
|
11
|
Ceppi F, Cazzaniga G, Colombini A, Biondi A, Conter V. Risk factors for relapse in childhood acute lymphoblastic leukemia: prediction and prevention. Expert Rev Hematol 2014; 8:57-70. [PMID: 25367188 DOI: 10.1586/17474086.2015.978281] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
With current treatment regimens, survival rates for acute lymphoblastic leukemia (ALL) have improved dramatically since the 1980s, with current 5-year overall survival rates estimated at greater than 85%. This success was achieved, in part, through the implementation of risk-stratified therapy. Nevertheless, for a subgroup of patients (15-20%) with newly diagnosed ALL who will ultimately relapse, traditional risk assessment remains inadequate. The risk of relapse may be estimated on the basis of diagnostic features or early treatment response findings. Further progress in this field may thus come from refinement of predictive factors for relapse and treatment adaptation and from the identification of biological subsets of ALL patients who could benefit from specific target therapies. This article summarizes the aspects associated with the identification of predictive factors for relapse in childhood ALL and options available for prevention of disease recurrence.
Collapse
Affiliation(s)
- Francesco Ceppi
- Division of Haematology/Oncology, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|