51
|
Mulrooney DA, Hyun G, Ness KK, Bhakta N, Pui CH, Ehrhardt MJ, Krull KR, Crom DB, Chemaitilly W, Srivastava DK, Relling MV, Jeha S, Green DM, Yasui Y, Robison LL, Hudson MM. The changing burden of long-term health outcomes in survivors of childhood acute lymphoblastic leukaemia: a retrospective analysis of the St Jude Lifetime Cohort Study. LANCET HAEMATOLOGY 2019; 6:e306-e316. [PMID: 31078468 DOI: 10.1016/s2352-3026(19)30050-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/16/2019] [Accepted: 02/19/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Treatment for childhood acute lymphoblastic leukemia has evolved over the past five decades, with moderation of traditional chemotherapy and radiotherapy and the introduction of targeted immune-based and cellular-based therapies. The affect of these changes on late health outcomes has not been assessed. Using data from the The St Jude Lifetime (SJLIFE) Cohort, we aimed to characterise the magnitude of morbidity and patterns of late health outcomes among survivors of childhood acute lymphoblastic leukaemia treated over time. METHODS The St Jude Lifetime (SJLIFE) Cohort is a retrospective cohort study with prospective follow-up and ongoing data accrual designed to facilitate longitudinal, clinically-based assessment of health outcomes among survivors of paediatric malignancies. 980 survivors included in this analysis were diagnosed with paediatric acute lymphoblastic leukaemia at St Jude Children's Research Hospital (SJCRH) between Aug 28, 1963, and July 19, 2003, were aged 18 years old and older at enrolment, had a minimum follow-up of 10 years after diagnosis, and completed an initial on-campus SJLIFE assessment as of data cutoff (June 30, 2015). 272 community control participants, matched to survivors on 5-year age blocks in each sex, were recruited for comparison. Cumulative chemotherapy and radiation dose exposures and major medical events during and after therapy were retrieved from the medical records of the survivors. History or physical examination, laboratory analysis, physical fitness, and neurocognitive testing were done. Health conditions were graded according to a modified version of the Common Terminology Criteria for Adverse Events. Neurocognitive domains of attention (Trial Making Test Part A and Conner's Continuous Performance Test-II) and executive function (Trail Making Test Part B, Controlled Oral Word Association Test, and Wechsler Adult Intelligence Scale-III Digit Span Test Backward) were measured and age-adjusted Z scores were calculated. Mean cumulative count was used to calculate the age-standardised cumulative burden of health conditions over time. This cohort study is registered at ClinicalTrials.gov, number NCT00760656. FINDINGS 980 survivors of acute lymphoblastic leukaemia (50% women, median age at diagnosis 5 years [IQR 3·1-9·1 years], and median time from diagnosis of 30·0 years [22·7-36·3]) had a median age of 35·8 years (29·4-42·9) at assessment compared with 35·1 years (28·7-42·6) for 272 controls. Survivors had significantly more growth hormone deficiency, hypogonadism, and neuropathy than controls. By age 30 years, survivors of acute lymphoblastic leukaemia had, on average, 5·4 (95% CI 5·1-5·8) grade 1-4 health conditions, including 3·2 (2·9-3·4) grade 2-4 health conditions, compared with 2·0 (CI 1·7-2·2) grade 1-4 and 1·2 (1·03-1·4) grade 2-4 health conditions among controls. The cumulative burden of grade 2-4 health conditions involved multiple organ systems for survivors treated on protocols between 1962-91, but after elimination of cranial radiotherapy for children with acute lymphoblastic leukaemia, conditions now predominately include musculoskeletal and endocrine disorders for survivors on protocols between 1991-2007. INTERPRETATION Although changes in paediatric acute lymphoblastic leukaemia treatment protocols have improved overall survival, the burden of late morbidity remains high for these patients. We show that the pattern of late toxic effects has markedly changed over time, with survivors having a reduction in health conditions that are immediately life-threatening, however, maintaining health status and quality of life for survivors of paediatric acute lymphoblastic leukaemia requires continued medical surveillance, counselling, and lifestyle modifications. FUNDING US National Cancer Institute and the American Lebanese Syrian Associated Charities.
Collapse
Affiliation(s)
- Daniel A Mulrooney
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA; Departments of Pediatrics and Medicine, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA.
| | - Geehong Hyun
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Nickhill Bhakta
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Matthew J Ehrhardt
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Kevin R Krull
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Psychology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Deborah B Crom
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Wassim Chemaitilly
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Mary V Relling
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sima Jeha
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Daniel M Green
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA; Departments of Pediatrics and Medicine, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| |
Collapse
|
52
|
Karol SE, Janke LJ, Panetta JC, Ramsey LB, Cai X, Payton MA, Jenkins DA, Evans WE, Relling MV. Asparaginase combined with discontinuous dexamethasone improves antileukemic efficacy without increasing osteonecrosis in preclinical models. PLoS One 2019; 14:e0216328. [PMID: 31059548 PMCID: PMC6502315 DOI: 10.1371/journal.pone.0216328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/19/2019] [Indexed: 11/18/2022] Open
Abstract
Introduction Combination therapy for acute lymphoblastic leukemia (ALL) is highly effective but results in significant toxicity including osteonecrosis. Asparaginase is known to potentiate both the antileukemic and osteonecrosis-inducing effects of dexamethasone. The schedule of dexamethasone alters osteonecrosis risk. However, the effects of the interaction with asparaginase are unknown when dexamethasone is given on a discontinuous schedule. Methods Using the murine model of osteonecrosis, we compared the frequency of osteonecrosis in mice receiving discontinuous dexamethasone (3.5 days/ week) with mice receiving asparaginase and discontinuous dexamethasone. We then tested the effect on antileukemic efficacy using six pediatric ALL xenografts. Results The addition of asparaginase to discontinuous dexamethasone did not alter the rate of osteonecrosis compared to dexamethasone alone (7/35 in dexamethasone with asparaginase combination vs. 10/36 in dexamethasone alone, p = 0.62) despite increasing steady-state plasma dexamethasone levels (103.9 nM vs. 33.4 nM, p = 9.2x10-7). Combination therapy with asparaginase and dexamethasone demonstrated synergistic antileukemic effects across all six xenografts studied. Conclusions When discontinuous dexamethasone was given, its anti-leukemic activity synergized with asparaginase but the osteonecrosis-worsening effects of asparaginase (above dexamethasone alone) were not observed. Thus, there is a favorable drug interaction (unchanged toxicity, synergistic efficacy) between discontinuous dexamethasone and asparaginase.
Collapse
Affiliation(s)
- Seth E. Karol
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| | - Laura J. Janke
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - John C. Panetta
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Laura B. Ramsey
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Xiangjun Cai
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Monique A. Payton
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - David A. Jenkins
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - William E. Evans
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
53
|
Lauschke VM, Zhou Y, Ingelman-Sundberg M. Novel genetic and epigenetic factors of importance for inter-individual differences in drug disposition, response and toxicity. Pharmacol Ther 2019; 197:122-152. [PMID: 30677473 PMCID: PMC6527860 DOI: 10.1016/j.pharmthera.2019.01.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Individuals differ substantially in their response to pharmacological treatment. Personalized medicine aspires to embrace these inter-individual differences and customize therapy by taking a wealth of patient-specific data into account. Pharmacogenomic constitutes a cornerstone of personalized medicine that provides therapeutic guidance based on the genomic profile of a given patient. Pharmacogenomics already has applications in the clinics, particularly in oncology, whereas future development in this area is needed in order to establish pharmacogenomic biomarkers as useful clinical tools. In this review we present an updated overview of current and emerging pharmacogenomic biomarkers in different therapeutic areas and critically discuss their potential to transform clinical care. Furthermore, we discuss opportunities of technological, methodological and institutional advances to improve biomarker discovery. We also summarize recent progress in our understanding of epigenetic effects on drug disposition and response, including a discussion of the only few pharmacogenomic biomarkers implemented into routine care. We anticipate, in part due to exciting rapid developments in Next Generation Sequencing technologies, machine learning methods and national biobanks, that the field will make great advances in the upcoming years towards unlocking the full potential of genomic data.
Collapse
Affiliation(s)
- Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
54
|
Nadeau G, Ouimet-Grennan E, Aaron M, Drouin S, Bertout L, Shalmiev A, Beaulieu P, St-Onge P, Veilleux LN, Rauch F, Petrykey K, Laverdière C, Sinnett D, Alos N, Krajinovic M. Identification of genetic variants associated with skeletal muscle function deficit in childhood acute lymphoblastic leukemia survivors. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:33-45. [PMID: 31114288 PMCID: PMC6489684 DOI: 10.2147/pgpm.s192924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022]
Abstract
Background: Although 80% of childhood acute lymphoblastic leukemia (ALL) cases are cured with current treatment protocols, exposure to chemotherapeutics or radiation therapy during a vulnerable period of child development has been associated with a high frequency of late adverse effects (LAE). Previous observations suggest important skeletal muscle size, density and function deficits in ALL survivors. Purpose: Given that only a fraction of all patients will suffer from this particular complication, we investigated whether it could be predicted by genetic markers. Patients and methods: We analysed associations between skeletal muscle force (Fmax) and power (Pmax) and germline genetic variants from 1039 genes derived through whole-exome sequencing. Top-ranking association signals retained after correction for multiple testing were confirmed through genotyping, and further analysed through stratified analyses and multivariate models. Results: Our results show that skeletal muscle function deficit is associated with two common single nucleotide polymorphisms (SNPs) (rs2001616DUOX2, P=0.0002 (Pmax) and rs41270041ADAMTS4, P=0.02 (Fmax)) and two rare ones located in the ALOX15 gene (P=0.001 (Pmax)). These associations were further modulated by sex, body mass index and risk groups, which reflected glucocorticoid dose and radiation therapy (P≤0.02). Conclusion: Occurrence of muscle function deficit in childhood ALL is thus strongly modulated by variations in the DUOX2, ADAMTS4 and ALOX15 genes, which could lead to personalized prevention strategies in childhood ALL survivors.
Collapse
Affiliation(s)
- Geneviève Nadeau
- Department of Medicine, University of Montreal, Montreal, QC, Canada
| | | | - Michelle Aaron
- Department of Medicine, University of Montreal, Montreal, QC, Canada
| | - Simon Drouin
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Laurence Bertout
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Albert Shalmiev
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Patrick Beaulieu
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Pascal St-Onge
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | | | - Frank Rauch
- Division of paediatrics, Montreal Shriners Hospital for Children, Montreal, QC, Canada
| | - Kateryna Petrykey
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Caroline Laverdière
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Hemato-Oncology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| | - Daniel Sinnett
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Hemato-Oncology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| | - Nathalie Alos
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Endocrinology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| | - Maja Krajinovic
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Hemato-Oncology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| |
Collapse
|
55
|
Pavlovic S, Kotur N, Stankovic B, Zukic B, Gasic V, Dokmanovic L. Pharmacogenomic and Pharmacotranscriptomic Profiling of Childhood Acute Lymphoblastic Leukemia: Paving the Way to Personalized Treatment. Genes (Basel) 2019; 10:E191. [PMID: 30832275 PMCID: PMC6471971 DOI: 10.3390/genes10030191] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Personalized medicine is focused on research disciplines which contribute to the individualization of therapy, like pharmacogenomics and pharmacotranscriptomics. Acute lymphoblastic leukemia (ALL) is the most common malignancy of childhood. It is one of the pediatric malignancies with the highest cure rate, but still a lethal outcome due to therapy accounts for 1%⁻3% of deaths. Further improvement of treatment protocols is needed through the implementation of pharmacogenomics and pharmacotranscriptomics. Emerging high-throughput technologies, including microarrays and next-generation sequencing, have provided an enormous amount of molecular data with the potential to be implemented in childhood ALL treatment protocols. In the current review, we summarized the contribution of these novel technologies to the pharmacogenomics and pharmacotranscriptomics of childhood ALL. We have presented data on molecular markers responsible for the efficacy, side effects, and toxicity of the drugs commonly used for childhood ALL treatment, i.e., glucocorticoids, vincristine, asparaginase, anthracyclines, thiopurines, and methotrexate. Big data was generated using high-throughput technologies, but their implementation in clinical practice is poor. Research efforts should be focused on data analysis and designing prediction models using machine learning algorithms. Bioinformatics tools and the implementation of artificial i Lack of association of the CEP72 rs924607 TT genotype with intelligence are expected to open the door wide for personalized medicine in the clinical practice of childhood ALL.
Collapse
Affiliation(s)
- Sonja Pavlovic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Nikola Kotur
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Biljana Stankovic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Branka Zukic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Vladimir Gasic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Lidija Dokmanovic
- University Children's Hospital, 11000 Belgrade, Serbia.
- University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia.
| |
Collapse
|
56
|
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
|
57
|
Chen X, Zhang L, Liang D, Li J, Liu F, Ma H. Lipid Transporter Activity-Related Genetic Polymorphisms Are Associated With Steroid-Induced Osteonecrosis of the Femoral Head: An Updated Meta-Analysis Based on the GRADE Guidelines. Front Physiol 2018; 9:1684. [PMID: 30559675 PMCID: PMC6287043 DOI: 10.3389/fphys.2018.01684] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/08/2018] [Indexed: 01/11/2023] Open
Abstract
Aims: The purpose of this study was to assess the relationship between genetic variants and steroid-induced osteonecrosis of the femoral head (SONFH) in steroid use populations. Methods: We searched the public databases up to April 15, 2018. This study analyzed only the single-nucleotide polymorphisms (SNPs) that have appeared in more than three studies and assessed the level of evidence by classifying the outcomes according to the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. Results: The ABCB1 rs1045642 C>T mutation had a protective effect against SONFH in the allelic model (I 2 = 50.2%; OR: 0.74; 95% CI: 0.55-1.00; p = 0.046). The rs2032582 mutation in the ABCB1 gene showed no relationship to SONFH (allelic model: I 2 = 63.4%; OR: 0.85; 95% CI: 0.58-1.23; p = 0.382). In ApoB rs693, four models showed that mutations can increase SONFH risk, but the allelic model did not. The ApoB rs1042031 mutation increased SONFH risk in the dominant model (I 2 = 50.3%; OR: 2.90; 95% CI: 1.49-5.66; p = 0.002). Conclusion: An allelic model of ABCB1 rs1045642 showed that mutations have a protective effect against SONFH at a very low level of evidence. The mutations in ApoB rs693 and rs1042031 increase the SONFH risk with moderate levels of evidence.
Collapse
Affiliation(s)
- Xiantao Chen
- Department of Osteonecrosis of the Femoral Head, Luoyang Orthopedic Hospital of Henan Province, Luoyang, China
| | - Leilei Zhang
- Department of Osteonecrosis of the Femoral Head, Luoyang Orthopedic Hospital of Henan Province, Luoyang, China
| | - Dawei Liang
- Department of Osteonecrosis of the Femoral Head, Luoyang Orthopedic Hospital of Henan Province, Luoyang, China
| | - Jing Li
- Department of Osteoarthritis, Luoyang Orthopedic Hospital of Henan Province, Luoyang, China
| | - Fenzhi Liu
- Department of Osteoarthritis, Luoyang Orthopedic Hospital of Henan Province, Luoyang, China
| | - Hongxia Ma
- Department of Osteonecrosis of the Femoral Head, Luoyang Orthopedic Hospital of Henan Province, Luoyang, China
| |
Collapse
|
58
|
ElHarouni D, Yassin D, Ali N, Gohar S, Zaky I, Adwan H, Sidhom I. A Pharmacogenetic Study of VDR fok1 and TYMS Polymorphisms and Their Association With Glucocorticoid-Induced Osteonecrosis in Egyptian Children With Acute Lymphoblastic Leukemia. Front Oncol 2018; 8:541. [PMID: 30533396 PMCID: PMC6266501 DOI: 10.3389/fonc.2018.00541] [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: 07/23/2018] [Accepted: 11/05/2018] [Indexed: 12/02/2022] Open
Abstract
Purpose: Osteonecrosis is a significant toxicity resulting from the treatment of pediatric Acute Lymphoblastic Leukemia (ALL). This study aimed to investigate the relationship between vitamin D receptor fok1 (VDR fok1) and thymidylate synthase (TYMS) gene polymorphisms with the glucocorticoid (GC) induced osteonecrosis (ON) in Egyptian pediatric ALL patients. In addition, to identify the possible association of genetic polymorphisms with other factors such as gender and ALL subtypes. Patients and Methods: A retrospective case-control study was conducted on 102 pediatric ALL patients under the age of 18 who were treated at Children Cancer Hospital Egypt according to St Jude SR/HR total XV protocol. The recruited patients were composed of 51 cases who developed GC-induced osteonecrosis and 51 age- and gender-matched patients who received glucocorticoids but remained osteonecrosis-free (controls). Genotyping of the VDR fok1 and TYMS genes was performed using restriction fragment length polymorphism (RFLP) and conventional PCR, respectively. Results: For the total 102 studied patients, the VDR fok1 single nucleotide polymorphisms (SNPs) frequency distribution were TT (8.8%), CT (34.3%), and CC (56.9%), while the TYMS tandem repeat gene variations were reported as 2R2R (20.6%), 2R3R (45.1%), and 3R3R (34.3%). VDR fok1 and TYMS polymorphic variants showed no association neither with gender; P-values 0.3808 and 0.1503, respectively, nor with ALL subtypes; P-values 0.9396 and 0.6596, respectively. The VDR fok1 polymorphisms showed a significant association with the development of ON; P-value = 0.003, on the other hand, TYMS tandem repeats did not show significant impact on osteonecrosis development; P-value = 0.411. Conclusion: This study showed a significant association between the VDR fok1 polymorphism and osteonecrosis. Such clinical pharmacogenetics results would be promising to discuss the possibility of dose adjustments aiming a regimen with the highest efficacy and least toxicity.
Collapse
Affiliation(s)
- Dina ElHarouni
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology German University in Cairo, New Cairo, Egypt
| | - Dina Yassin
- Clinical Pathology Department, Children Cancer Hospital Egypt and National Cancer Institute Cairo University, Cairo, Egypt
| | - Nesreen Ali
- Pediatric Oncology Department, Children Cancer Hospital Egypt and National Cancer Institute Cairo University, Cairo, Egypt
| | - Seham Gohar
- Pediatric Oncology Department Children Cancer Hospital Egypt, Cairo, Egypt
| | - Iman Zaky
- Radiology Department, Children Cancer Hospital Egypt and National Cancer Institute Cairo University, Cairo, Egypt
| | - Hassan Adwan
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology German University in Cairo, New Cairo, Egypt
| | - Iman Sidhom
- Pediatric Oncology Department, Children Cancer Hospital Egypt and National Cancer Institute Cairo University, Cairo, Egypt
| |
Collapse
|
59
|
Tanoshima R, Carleton BC. The incidence of symptomatic osteonecrosis after allogeneic haematopoietic stem cell transplantation in children with acute lymphoblastic leukaemia – controversy on dexamethasone as a risk factor. Br J Haematol 2018; 185:958-959. [DOI: 10.1111/bjh.15657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Reo Tanoshima
- Division of Translational Therapeutics Department of Pediatrics University of British Columbia Vancouver BC Canada
| | - Bruce C. Carleton
- Division of Translational Therapeutics Department of Pediatrics University of British Columbia Vancouver BC Canada
| |
Collapse
|
60
|
Wang T, Azeddine B, Mah W, Harvey EJ, Rosenblatt D, Séguin C. Osteonecrosis of the femoral head: genetic basis. INTERNATIONAL ORTHOPAEDICS 2018; 43:519-530. [PMID: 30328481 DOI: 10.1007/s00264-018-4172-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Genetic factors and hereditary forms of osteonecrosis of the femoral head (ONFH) have been elucidated through genetic association studies. The significance of these cases is that they suggest an alternative hypothesis to the development of the disease. This review presents a summary of single nucleotide polymorphisms (SNPs) and other genetic mutation variations found in association with ONFH, including our recent identification of a novel mutation in the transient receptor potential vanilloid 4 (TRPV4) gene in association with inherited ONFH. The purpose of this review is to consolidate and categorize genetic linkages according to physiological pathways. METHODS A systematic review of literature from PubMed and Google Scholar was undertaken with a focus on genetic linkages and hereditary case studies of the disease. Recent genetic analysis studies published after 2007 were the focus of genetic linkages in non-hereditary cases. RESULTS The summary of these genetic findings identifies biological processes believed to be involved in the development of ONFH, which include circulation, steroid metabolism, immunity, and the regulation of bone formation. CONCLUSION Taken together, these associations may lead to new pathways of bone repair and remodeling while opening new avenues for therapeutic targets. Knowledge of genetic variations could help identify individuals considered to be at higher risk of developing ONFH and prevent the multiple hit effect.
Collapse
Affiliation(s)
- Tracy Wang
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
| | - Bouziane Azeddine
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Wayne Mah
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Edward J Harvey
- Department Surgery, Division Orthopaedic Surgery, McGill University Health Centre, B5 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - David Rosenblatt
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Chantal Séguin
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada. .,Department of Medicine, Division of Hematology and Oncology, McGill University Health Centre, Montreal, Quebec, H4A 3J1, Canada. .,Glen Site, Cedars Cancer Centre, McGill University Health Centre, 1001 Décarie Blvd., room D02.7519, Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
61
|
Mogensen SS, Harila-Saari A, Mäkitie O, Myrberg IH, Niinimäki R, Vestli A, Hafsteinsdottir S, Griškevicius L, Saks K, Hallböök H, Retpen J, Helt LR, Toft N, Schmiegelow K, Frandsen TL. Comparing osteonecrosis clinical phenotype, timing, and risk factors in children and young adults treated for acute lymphoblastic leukemia. Pediatr Blood Cancer 2018; 65:e27300. [PMID: 29943905 DOI: 10.1002/pbc.27300] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Treatment-related osteonecrosis (ON) is a serious complication of treatment of acute lymphoblastic leukemia (ALL). PROCEDURE This study included 1,489 patients with ALL, aged 1-45 years, treated according to the Nordic Society of Paediatric Haematology and Oncology ALL2008 protocol, using alternate-week dexamethasone during delayed intensification, with prospective registration of symptomatic ON. We aimed at comparing risk factors, timing, and clinical characteristics of ON in children and young adults. RESULTS ON was diagnosed in 67 patients, yielding a 5-year cumulative incidence of 6.3%, but 28% in female adolescents. Median age at ALL diagnosis was 12.1 years and 14.9 years for females and males, respectively. At ON diagnosis, 59 patients had bone pain (91%) and 30 (46%) had multiple-joint involvement. The median interval between ALL and ON diagnosis was significantly shorter in children aged 1.0-9.9 years (0.7 years [range: 0.2-2.1]) compared with adolescents (1.8 years [range: 0.3-3.7, P < 0.001]) and adults (2.1 years [range: 0.4-5.3, P = 0.001]). Female sex was a risk factor in adolescent patients (hazard ratio [HR] = 2.1, 95% confidence interval [CI]: 1.1-4.2) but not in children aged 1.1-9.9 years (HR = 2.4, 95% CI: 0.9-6.2, P = 0.08) or adults aged 19-45 years (HR = 1.1, 95% CI: 0.3-4.0). Age above 10 years at ALL diagnosis (odds ratio [OR] = 3.7, P = 0.026) and multiple joints affected at ON diagnosis (OR = 3.4, P = 0.027) were risk factors for developing severe ON. CONCLUSION We provide a detailed phenotype of patients with ALL with symptomatic ON, including description of risk factors and timing of ON across age groups. This awareness is essential in exploring measures to prevent development of ON.
Collapse
Affiliation(s)
- Signe Sloth Mogensen
- Department of Pediatrics and Adolescent Medicine, the Juliane Marie Center, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Arja Harila-Saari
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Outi Mäkitie
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Ida Hed Myrberg
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Riitta Niinimäki
- Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
| | - Anne Vestli
- Department of Pediatric Oncology and Hematology, Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Laimonas Griškevicius
- Department of Hematology, Oncology, and Transfusion Medicine Center, Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Kadri Saks
- Department of Hematology Oncology, Tallinn Children´s Hospital, Tallinn, Estonia
| | - Helene Hallböök
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jens Retpen
- Department of Orthopedic Surgery, University Hospital Herlev Gentofte, Copenhagen, Denmark
| | - Louise Rold Helt
- Department of Pediatrics and Adolescent Medicine, the Juliane Marie Center, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Nina Toft
- Department of Hematology, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, the Juliane Marie Center, University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Leth Frandsen
- Department of Pediatrics and Adolescent Medicine, the Juliane Marie Center, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
62
|
Abstract
OBJECTIVES Glucocorticoids such as dexamethasone have pleiotropic effects, including desired antileukemic, anti-inflammatory, or immunosuppressive effects, and undesired metabolic or toxic effects. The most serious adverse effects of dexamethasone among patients with acute lymphoblastic leukemia are osteonecrosis and thrombosis. To identify inherited genomic variation involved in these severe adverse effects, we carried out genome-wide association studies (GWAS) by analyzing 14 pleiotropic glucocorticoid phenotypes in 391 patients with acute lymphoblastic leukemia. PATIENTS AND METHODS We used the Projection Onto the Most Interesting Statistical Evidence integrative analysis technique to identify genetic variants associated with pleiotropic dexamethasone phenotypes, stratifying for age, sex, race, and treatment, and compared the results with conventional single-phenotype GWAS. The phenotypes were osteonecrosis, central nervous system toxicity, hyperglycemia, hypokalemia, thrombosis, dexamethasone exposure, BMI, growth trajectory, and levels of cortisol, albumin, and asparaginase antibodies, and changes in cholesterol, triglycerides, and low-density lipoproteins after dexamethasone. RESULTS The integrative analysis identified more pleiotropic single nucleotide polymorphism variants (P=1.46×10(-215), and these variants were more likely to be in gene-regulatory regions (P=1.22×10(-6)) than traditional single-phenotype GWAS. The integrative analysis yielded genomic variants (rs2243057 and rs6453253) in F2RL1, a receptor that functions in hemostasis, thrombosis, and inflammation, which were associated with pleiotropic effects, including osteonecrosis and thrombosis, and were in regulatory gene regions. CONCLUSION The integrative pleiotropic analysis identified risk variants for osteonecrosis and thrombosis not identified by single-phenotype analysis that may have importance for patients with underlying sensitivity to multiple dexamethasone adverse effects.
Collapse
|
63
|
Liu LH, Zhang QY, Sun W, Li ZR, Gao FQ. Corticosteroid-induced Osteonecrosis of the Femoral Head: Detection, Diagnosis, and Treatment in Earlier Stages. Chin Med J (Engl) 2018; 130:2601-2607. [PMID: 29067959 PMCID: PMC5678261 DOI: 10.4103/0366-6999.217094] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective: This review aimed to provide a current recommendation to multidisciplinary physicians for early detection, diagnosis, and treatment of corticosteroid-induced osteonecrosis of the femoral head (ONFH) based on a comprehensive analysis of the clinical literature. Data Sources: For the purpose of collecting potentially eligible articles, we searched for articles in the PubMed, Cochrane Library, Embase, and CNKI databases up to February 2017, using the following key words: “corticosteroid”, “osteonecrosis of the femoral head”, “risk factors”, “diagnosis”, “prognosis”, and “treatment”. Study Selection: Articles on relationships between corticosteroid and ONFH were selected for this review. Articles on the diagnosis, prognosis, and intervention of earlier-stage ONFH were also reviewed. Results: The incidence of corticosteroid-induced ONFH was associated with high doses of corticosteroids, and underlying diseases in certain predisposed individuals mainly occurred in the first 3 months of corticosteroid prescription. The enhanced awareness and minimized exposure to the established risk factors and earlier definitive diagnosis are essential for the success of joint preservation. When following up patients with ONFH, treatment should be started if necessary. Surgical treatment yielded better results than conservative therapy in earlier-stage ONFH. The ideal purpose of earlier intervention and treatment is permanent preservation of the femoral head without physical restrictions in daily living. Conclusions: Clinicians should enhance their precaution awareness of corticosteroid-induced ONFH. For high-risk patients, regular follow-up is very important in the 1st year after high-dose prescription of corticosteroids. Patients with suspected ONFH should be referred to orthopedists for diagnosis and treatment in its earlier stage to preserve the joint.
Collapse
Affiliation(s)
- Li-Hua Liu
- Peking Union Medical College, China-Japan Friendship Institute of Clinical Medicine, Beijing 100029, China
| | - Qing-Yu Zhang
- Peking Union Medical College, China-Japan Friendship Institute of Clinical Medicine, Beijing 100029, China
| | - Wei Sun
- Department of Orthopaedic Surgery, Centre for Osteonecrosis and Joint Preserving and Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China
| | - Zi-Rong Li
- Department of Orthopaedic Surgery, Centre for Osteonecrosis and Joint Preserving and Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China
| | - Fu-Qiang Gao
- Department of Orthopaedic Surgery, Centre for Osteonecrosis and Joint Preserving and Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
64
|
Pui CH, Yang JJ, Bhakta N, Rodriguez-Galindo C. Global efforts toward the cure of childhood acute lymphoblastic leukaemia. THE LANCET. CHILD & ADOLESCENT HEALTH 2018; 2:440-454. [PMID: 30169285 PMCID: PMC6467529 DOI: 10.1016/s2352-4642(18)30066-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 01/31/2018] [Accepted: 02/13/2018] [Indexed: 12/18/2022]
Abstract
Improvements in risk-directed treatment and supportive care, together with increased reliance on both national and international collaborative studies, have made childhood acute lymphoblastic leukaemia (ALL) one of the most curable human cancers. Next-generation sequencing studies of leukaemia cells and the host germline provide new opportunities for precision medicine and thus potential improvements in the cure rate and quality of life of patients. Efforts are underway to assess the global impact of childhood ALL and develop initiatives that can meet the long-term challenge of providing quality care to children with this disease worldwide and improving cure rates globally. This ambitious task will rely on increased collaborative research and international networking so that the therapeutic gains in high-income countries can be translated to patients in low-income and middle-income countries. Ultimately, the greatest obstacle to overcome will be to fully understand leukaemogenesis, enabling measures to decrease the risk of leukaemia development and thus close the last major gap in offering a cure to any child who might have the disease.
Collapse
Affiliation(s)
- Ching-Hon Pui
- Department of Oncology, St Jude Children's Research Hospital, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Jun J Yang
- Department of Pharmaceutical Science, St Jude Children's Research Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nickhill Bhakta
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Carlos Rodriguez-Galindo
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
65
|
Krull K, Kunstreich M, Bronsema A, Bleckmann K, Classen CF, Erdlenbruch B, Jorch N, Kolb R, Leipold A, Moser O, Prokop A, Scheurlen W, Steinbach D, Klasen-Sansone J, Klee D, Escherich G, Moericke A, Schrappe M, Borkhardt A, Kuhlen M. Osteonecrosis in children with acute lymphoblastic leukemia at initial diagnosis and prior to any chemotherapy. Leuk Lymphoma 2018; 60:78-84. [DOI: 10.1080/10428194.2018.1466292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Kathinka Krull
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, University of Duesseldorf, Duesseldorf, Germany
| | - Marina Kunstreich
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, University of Duesseldorf, Duesseldorf, Germany
| | - Annika Bronsema
- Clinic of Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kirsten Bleckmann
- Department of Pediatrics, University Medical Centre Schleswig-Holstein Campus Kiel, Kiel, Germany
| | | | - Bernhard Erdlenbruch
- University Hospital for Children and Adolescents, Johannes Wesling Klinikum Minden, Ruhr University Hospital, Bochum, Germany
| | - Norbert Jorch
- Department of Pediatrics, Evangelical Hospital, Bielefeld, Germany
| | - Reinhard Kolb
- Pediatric Hematology and Oncology, Elisabeth-Hospital Oldenburg, Oldenburg, Germany
| | | | - Olga Moser
- Department of Pediatric Hematology and Oncology, University Hospital Bonn, Germany
| | - Aram Prokop
- Department of Pediatric Oncology/Hematology, Children's Hospital Cologne, Cologne, Germany
| | - Wolfram Scheurlen
- Cnopf'sche Kinderklinik, Nürnberg Children's Hospital, Nürnberg, Germany
| | - Daniel Steinbach
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Janina Klasen-Sansone
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| | - Dirk Klee
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| | - Gabriele Escherich
- Clinic of Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Anja Moericke
- Department of Pediatrics, University Medical Centre Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Martin Schrappe
- Department of Pediatrics, University Medical Centre Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, University of Duesseldorf, Duesseldorf, Germany
| | - Michaela Kuhlen
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, University of Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
66
|
Conyers R, Devaraja S, Elliott D. Systematic review of pharmacogenomics and adverse drug reactions in paediatric oncology patients. Pediatr Blood Cancer 2018; 65. [PMID: 29286579 DOI: 10.1002/pbc.26937] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022]
Abstract
Many paediatric patients with cancer experience significant chemotherapy side effects. Predisposition to drug reactions is governed by single nucleotide polymorphisms (SNPs). We performed a systematic review of the literature from 2006 through 2016. Outcomes of interest included patient characteristics, cancer type drug of interest, genes investigated, toxicity identified and genetic polymorphisms implicated. The primary toxicities studied were neurotoxicity cardiotoxicity, osteonecrosis, and thromboembolism and hypersensitivity reactions. The retrieved studies were grouped according to toxicity reported and SNP associations. This review highlights the discoveries to date in pharmacogenomics and paediatric oncology along with highlighting some of the important limitations in the area.
Collapse
Affiliation(s)
- Rachel Conyers
- Murdoch Children's Research Institute, Melbourne, Australia.,Children's Cancer Centre, The Royal Children's Hospital, Melbourne, Australia
| | - Subalatha Devaraja
- Department of Medicine, Melbourne University, Melbourne, Australia.,Children's Cancer Centre, The Royal Children's Hospital, Melbourne, Australia
| | - David Elliott
- Murdoch Children's Research Institute, Melbourne, Australia
| |
Collapse
|
67
|
Kahn JM, Cole PD, Blonquist TM, Stevenson K, Jin Z, Barrera S, Davila R, Roberts E, Neuberg DS, Athale UH, Clavell LA, Laverdiere C, Leclerc JM, Michon B, Schorin MA, Welch JJ, Sallan SE, Silverman LB, Kelly KM. An investigation of toxicities and survival in Hispanic children and adolescents with ALL: Results from the Dana-Farber Cancer Institute ALL Consortium protocol 05-001. Pediatr Blood Cancer 2018; 65:10.1002/pbc.26871. [PMID: 29090520 PMCID: PMC5766393 DOI: 10.1002/pbc.26871] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/15/2022]
Abstract
PURPOSE This study compared the relative incidence of treatment-related toxicities and the event-free and overall survival between Hispanic and non-Hispanic children undergoing therapy for acute lymphoblastic leukemia (ALL) on Dana-Farber Cancer Institute ALL Consortium protocol 05-001. PATIENTS AND METHODS Secondary analysis of prospectively collected data from a phase III multicenter study in children and adolescents of 1-18 years with previously untreated ALL. RESULTS Between 2005 and 2011, 794 eligible patients enrolled on DFCI 05-001, 730 of whom were included in this analysis (19% [N = 150] Hispanic, 73% [N = 580] non-Hispanic). Hispanic patients were more likely to be ≥10 years of age (32% vs. 24%, P = 0.045) at diagnosis. Toxicity analyses revealed that Hispanic patients had significantly lower cumulative incidence of bone fracture (P < 0.001) and osteonecrosis (ON; P = 0.047). In multivariable risk regression, the risk of ON was significantly lower in Hispanic patients ≥10 years (HR 0.23; P = 0.006). Hispanic patients had significantly lower 5-year event-free survival (EFS) (79.4%; 95% CI: 71.6-85.2) and overall survival (OS) (89.2%; 95% CI: 82.7-93.4) than non-Hispanic patients (EFS: 87.5%; 95% CI: 84.5-90.0, P = 0.004; OS: 92.7%; 95% CI: 90.2-94.6, P = 0.006). Exploratory analyses revealed differences between Hispanic and non-Hispanic patients in the frequency of common variants in genes related to toxicity or ALL outcome. CONCLUSION Hispanic children treated for ALL on DFCI 05-001 had fewer bone-related toxicities and inferior survival than non-Hispanic patients. While disease biology is one explanatory variable for outcome disparities, these findings suggest that biologic and non-biologic mechanisms affecting drug delivery and exposure in this population may be important contributing factors as well.
Collapse
Affiliation(s)
- Justine M. Kahn
- Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Columbia University Medical Center, NewYork-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA
| | - Peter D. Cole
- Division of Pediatric Hematology/Oncology, Children’s Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Traci M. Blonquist
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kristen Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Zhezhen Jin
- Department of Biostatistics, Columbia University Medical Center, New York, New York, USA
| | - Sergio Barrera
- Department of Economics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Randy Davila
- Department of Psychology, University of California, Davis, Davis, California, USA
| | - Emily Roberts
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Donna S. Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Uma H. Athale
- Division of Pediatric Hematology/Oncology, McMaster University, Hamilton, ON, Canada
| | - Luis A. Clavell
- Division of Pediatric Oncology, San Jorge Children’s Hospital, San Juan, Puerto Rico
| | - Caroline Laverdiere
- Division of Hematology and Oncology, Hospital Sainte-Justine, University of Montreal, Montreal, Canada
| | - Jean-Marie Leclerc
- Division of Hematology and Oncology, Hospital Sainte-Justine, University of Montreal, Montreal, Canada
| | - Bruno Michon
- Division of Hematology-Oncology, Centre Hospitalier de l’Université Laval, Quebec City, Canada
| | | | - Jennifer J.G. Welch
- Division of Pediatric Hematology-Oncology, Hasbro Children’s Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Stephen E. Sallan
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lewis B. Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kara M. Kelly
- Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Columbia University Medical Center, NewYork-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA,Roswell Park Cancer Institute and Women and Children’s Hospital, University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
68
|
Sakamoto K, Imamura T, Kihira K, Suzuki K, Ishida H, Morita H, Kanno M, Mori T, Hiramatsu H, Matsubara K, Terui K, Takahashi Y, Suenobu SI, Hasegawa D, Kosaka Y, Kato K, Moriya-Saito A, Sato A, Kawasaki H, Yumura-Yagi K, Hara J, Hori H, Horibe K. Low Incidence of Osteonecrosis in Childhood Acute Lymphoblastic Leukemia Treated With ALL-97 and ALL-02 Study of Japan Association of Childhood Leukemia Study Group. J Clin Oncol 2018; 36:900-907. [PMID: 29360413 DOI: 10.1200/jco.2017.75.5066] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Osteonecrosis (ON) is a serious complication of the treatment of childhood acute lymphoblastic leukemia (ALL); however, data relating to ON in Asian pediatric patients with ALL are scarce. Therefore, we performed a retrospective analysis of cohorts of Japanese patients with ALL to clarify the incidence, clinical characteristics, and risk factors of ON. Patients and Methods The incidence and characteristics of ON were determined in patients with ALL (n = 1,662) enrolled in two studies from the Japan Association of Childhood Leukemia Study (JACLS) group (n = 635 and n = 1,027 patients treated with the ALL-97 and ALL-02 protocols, respectively). Results In total, 24 of 1,662 patients suffered from ON, of which 12 of 635 and 12 of 1,027 patients were treated with the ALL-97 and the ALL-02 protocol, respectively. Of the 24 patients, 23 were older than 10 years. In multivariate analysis, age (≥ 10 years) was the sole significant risk factor for ON ( P < .001). Separate evaluation of patients ≥ 10 years of age indicated a 5-year cumulative incidence of ON of 7.2% (95% CI, 4.0% to 12.6%) and 5.9% (95% CI, 3.3% to 10.4%) in the ALL-97 and the ALL-02 protocol, respectively, which was lower than reported previously, despite an administration of dexamethasone (DEX) similar to that in comparable studies; however, concomitant administration of DEX and l-asparaginase was reduced in the JACLS protocols. Conclusion We identified a low frequency of ON in the JACLS ALL-97 and ALL-02 studies. Although the sole risk factor for ON was age (≥ 10 years), even among high-risk patients, ON incidence was significantly lower than that reported in previous studies. These results suggest that, not only the total amount of DEX, but also how DEX and l-asparaginase are administered, which affects the clearance of DEX, may be associated with ON incidence in patients with ALL.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Toshihiko Imamura
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Kentaro Kihira
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Koji Suzuki
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Hisashi Ishida
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Hiromi Morita
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Miyako Kanno
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Takeshi Mori
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Hidefumi Hiramatsu
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Kousaku Matsubara
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Kiminori Terui
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Yoshihiro Takahashi
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - So-Ichi Suenobu
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Daiichiro Hasegawa
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Yoshiyuki Kosaka
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Koji Kato
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Akiko Moriya-Saito
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Atsushi Sato
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Hirohide Kawasaki
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Keiko Yumura-Yagi
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Junichi Hara
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Hiroki Hori
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| | - Keizo Horibe
- Kenichi Sakamoto and Toshihiko Imamura, Kyoto Prefectural University of Medicine; Hidefumi Hiramatsu, Kyoto University, Kyoto; Kenichi Sakamoto, Toshihiko Imamura, Akiko Moriya-Saito, and Keizo Horibe, National Hospital Organization Nagoya Medical Center; Koji Kato, Japanese Red Cross Nagoya First Hospital, Nagoya; Kentaro Kihira and Hiroki Hori, Mie University, Mie; Koji Suzuki, University of Fukui Faculty of Medical Sciences, Fukui; Hisashi Ishida, Okayama University, Okayama; Hiromi Morita, University of Occupational and Environmental Health, Iseigaoka; Miyako Kanno, Yamagata University, Yamagata; Takeshi Mori, Kobe University; Kousaku Matsubara, Kobe City Nishi-Kobe Medical Center; Daiichiro Hasegawa and Yoshiyuki Kosaka, Hyogo Prefectural Children's Hospital, Kobe; Kiminori Terui, Hirosaki University, Hirosaki; Yoshihiro Takahashi, Aomori Prefectural Central Hospital, Aomori; So-ichi Suenobu, Oita University, Oita; Atsushi Sato, Miyagi Children's Hospital, Sendai; Hirohide Kawasaki, Kansai Medical University; Keiko Yumura-Yagi, Yumura Clinic; and Junichi Hara, Osaka City General Hospital, Osaka, Japan
| |
Collapse
|
69
|
Plesa M, Gagné V, Glisovic S, Younan M, Sharif-Askari B, Laverdière C, Alos N, Leclerc JM, Sallan SE, Neuberg D, Kutok JL, Silverman LB, Sinnett D, Krajinovic M. Influence of BCL2L11 polymorphism on osteonecrosis during treatment of childhood acute lymphoblastic leukemia. THE PHARMACOGENOMICS JOURNAL 2017; 19:33-41. [PMID: 29282361 DOI: 10.1038/s41397-017-0002-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/06/2017] [Accepted: 09/18/2017] [Indexed: 11/09/2022]
Abstract
Osteonecrosis (ON) is corticosteroid-related complication, reported in children with acute lymphoblastic leukemia (ALL). We have previously found that polymorphisms in BCL2L11 gene coding for pro-apoptotic Bim protein influence reduction of overall survival (OS) in a corticosteroid (CS) dose-dependent manner in childhood ALL patients. The same set of SNPs was here investigated for an association with CS-related ON assessed retrospectively in 304 children with ALL from Quebec (QcALL cohort) who received Dana-Farber Cancer Institute (DFCI) ALL treatment protocols. Two-year cumulative incidence of symptomatic ON was 10.6%. Two BCL2L11 polymorphisms, the 891T>G (rs2241843) in all QcALL patients and 29201C>T (rs724710) in high-risk group were significantly associated with ON, P = 0.009 and P = 0.003, respectively. The association remained significant in multivariate model (HR891TT = 2.4, 95% CI 1.2-4.8, P = 0.01 and HR29201CC = 5.7, 95% CI 1.6-20.9, P = 0.008). Both polymorphisms influenced viability of dexamethasone treated lymphoblastoid cell lines (P ≤ 0.03). The 891T>G influenced Bim gamma isoform levels (0.03) and its association with ON was also confirmed in replication DFCI cohort (N = 168, P = 0.03). QcALL children had a high incidence of ON during therapy, which was highly associated with BCL2L11 polymorphisms.
Collapse
Affiliation(s)
- Maria Plesa
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Pharmacology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Vincent Gagné
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Sanja Glisovic
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Melissa Younan
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Bahram Sharif-Askari
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Caroline Laverdière
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Nathalie Alos
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Jean-Marie Leclerc
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Stephen E Sallan
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Hematology/Oncology, Children's Hospital, Boston, MA, USA
| | - Donna Neuberg
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jeffery L Kutok
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Lewis B Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Hematology/Oncology, Children's Hospital, Boston, MA, USA
| | - Daniel Sinnett
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Maja Krajinovic
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada. .,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada. .,Department of Pharmacology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
70
|
Genome-wide Association Study of Idiopathic Osteonecrosis of the Femoral Head. Sci Rep 2017; 7:15035. [PMID: 29118346 PMCID: PMC5678103 DOI: 10.1038/s41598-017-14778-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/12/2017] [Indexed: 11/13/2022] Open
Abstract
Idiopathic osteonecrosis of the femoral head (IONFH) is an ischemic disorder that causes bone necrosis of the femoral head, resulting in hip joint dysfunction. IONFH is a polygenic disease and steroid and alcohol have already known to increase its risk; however, the mechanism of IONFH remains to be elucidated. We performed a genome-wide association study using ~60,000 subjects and found two novel loci on chromosome 20q12 and 12q24. Big data analyses identified LINC01370 as a candidate susceptibility gene in the 20q12 locus. Stratified analysis by IONFH risk factors suggested that the 12q24 locus was associated with IONFH through drinking capacity. Our findings would shed new light on pathophysiology of IONFH.
Collapse
|
71
|
de Rojas T, Martínez-Álvarez S, Lerma-Lara S, Díaz MÁ, Madero L, Ramírez M. Outcome of childhood leukaemia survivors and necrosis of the femoral head treated with autologous mesenchymal stem cells. Clin Transl Oncol 2017; 20:584-590. [DOI: 10.1007/s12094-017-1752-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 09/02/2017] [Indexed: 12/20/2022]
|
72
|
Hardy KK, Embry L, Kairalla JA, Helian S, Devidas M, Armstrong D, Hunger S, Carroll WL, Larsen E, Raetz EA, Loh ML, Yang W, Relling MV, Noll RB, Winick N. Neurocognitive Functioning of Children Treated for High-Risk B-Acute Lymphoblastic Leukemia Randomly Assigned to Different Methotrexate and Corticosteroid Treatment Strategies: A Report From the Children's Oncology Group. J Clin Oncol 2017; 35:2700-2707. [PMID: 28671857 DOI: 10.1200/jco.2016.71.7587] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Survivors of childhood acute lymphoblastic leukemia (ALL) are at risk for neurocognitive deficits that are associated with treatment, individual, and environmental factors. This study examined the impact of different methotrexate (MTX) and corticosteroid treatment strategies on neurocognitive functioning in children with high-risk B-lineage ALL. Methods Participants were randomly assigned to receive high-dose MTX with leucovorin rescue or escalating dose MTX with PEG asparaginase without leucovorin rescue. Patients were also randomly assigned to corticosteroid therapy that included either dexamethasone or prednisone. A neurocognitive evaluation of intellectual functioning (IQ), working memory, and processing speed (PS) was conducted 8 to 24 months after treatment completion (n = 192). Results The method of MTX delivery and corticosteroid assignment were unrelated to differences in neurocognitive outcomes after controlling for ethnicity, race, age, gender, insurance status, and time off treatment; however, survivors who were age < 10 years at diagnosis (n = 89) had significantly lower estimated IQ ( P < .001) and PS scores ( P = .02) compared with participants age ≥ 10 years. In addition, participants who were covered by US public health insurance had estimated IQs that were significantly lower ( P < .001) than those with US private or military insurance. Conclusion Children with high-risk B-lineage ALL who were age < 10 years at diagnosis are at risk for deficits in IQ and PS in the absence of cranial radiation, regardless of MTX delivery or corticosteroid type. These data may serve as a basis for developing screening protocols to identify children who are at high risk for deficits so that early intervention can be initiated to mitigate the impact of therapy on neurocognitive outcomes.
Collapse
Affiliation(s)
- Kristina K Hardy
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Leanne Embry
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - John A Kairalla
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Shanjun Helian
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Meenakshi Devidas
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Daniel Armstrong
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Stephen Hunger
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - William L Carroll
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Eric Larsen
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Elizabeth A Raetz
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Mignon L Loh
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Wenjian Yang
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Mary V Relling
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Robert B Noll
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| | - Naomi Winick
- Kristina K. Hardy, Children's National Medical Center, Washington, DC; Leanne Embry, The University of Texas Health Science Center at San Antonio, San Antonio; Naomi Winick, University of Texas Southwestern Medical Center, Dallas, TX; John A. Kairalla and Meenakshi Devidas, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville; Daniel Armstrong, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL; Shanjun Helian, Merck, Upper Gwynedd; Stephen Hunger, Children's Hospital of Philadelphia, Philadelphia; Robert B. Noll, University of Pittsburgh School of Medicine, Pittsburgh, PA; William L. Carroll, New York University Medical Center, New York, NY; Eric Larsen, Maine Children's Cancer Program, Scarborough, ME; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Mignon L. Loh, University of California, San Francisco, San Francisco, CA; and Wenjian Yang and Mary V. Relling, St Jude Children's Research Hospital; University of Tennessee Health Science Center, Memphis, TN
| |
Collapse
|
73
|
Finkelstein Y, Blonquist TM, Vijayanathan V, Stevenson KE, Neuberg DS, Silverman LB, Vrooman LM, Sallan SE, Cole PD. A thymidylate synthase polymorphism is associated with increased risk for bone toxicity among children treated for acute lymphoblastic leukemia. Pediatr Blood Cancer 2017; 64. [PMID: 27957785 DOI: 10.1002/pbc.26393] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/03/2016] [Accepted: 11/15/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bone fractures and osteonecrosis frequently complicate therapy for childhood acute lymphoblastic leukemia (ALL). Bone toxicity has been associated with exposure to corticosteroids and methotrexate (MTX) and age greater than 10 years. We tested whether common genetic polymorphisms were associated with bone toxicity during treatment for ALL. PROCEDURE A total of 615 of 794 children enrolled on Dana Farber Cancer Institute ALL Consortium protocol 05-001 (NCT00400946) met eligibility criteria for inclusion in this analysis. Nineteen candidate polymorphisms were selected a priori, targeting genes related to glucocorticoid metabolism, oxidative damage, and folate physiology. Polymorphisms were genotyped using either PCR-based allelic discrimination or PCR product length analysis. RESULTS Twenty percent of subjects were homozygous for two 28 bp repeats (2R/2R, where 2R is two 28-nucleotide repeats within the 5' untranslated region [UTR] of the thymidylate synthase [TS] gene) within the 5' UTR of the gene for TS. This 2R/2R genotype was associated with increased risk of osteonecrosis among children younger than 10 years at diagnosis (multivariable hazard ratio [HR] 2.71; 95% confidence interval [CI] 1.23-5.95; P = 0.013), and with bone fracture among children ≥ 10 years (multivariable HR 2.10; 95% CI 1.11-3.96; P = 0.022). No significant association was observed between TS genotype and red blood cell (RBC) folate, RBC MTX, or relapse risk. CONCLUSIONS A common genetic variant is associated with increased risk of osteonecrosis among children younger than 10 years at diagnosis and with bone fractures among older children. These findings suggest that children and adolescents with the 2R/2R TS genotype should be closely monitored for the development of bone toxicity during therapy for ALL, and support a clinical trial testing the efficacy of protective interventions specifically in this vulnerable population.
Collapse
Affiliation(s)
| | | | - Veena Vijayanathan
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Lewis B Silverman
- Dana Farber Cancer Institute, Boston, Massachusetts.,Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Lynda M Vrooman
- Dana Farber Cancer Institute, Boston, Massachusetts.,Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Stephen E Sallan
- Dana Farber Cancer Institute, Boston, Massachusetts.,Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Peter D Cole
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
74
|
Portera MV, Karol SE, Smith C, Yang W, Cheng C, Neel MD, Pui CH, Relling MV, Kaste SC. Osteonecrosis is unrelated to hip anatomy in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2017; 64:10.1002/pbc.26407. [PMID: 28035753 PMCID: PMC5596390 DOI: 10.1002/pbc.26407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/31/2022]
Abstract
Osteonecrosis is a debilitating toxicity associated with acute lymphoblastic leukemia (ALL) treatment. A recent report associated interindividual differences in hip anatomy with the development of idiopathic osteonecrosis in adults. To evaluate the impact of hip anatomy on the development of therapy-related osteonecrosis, we retrospectively evaluated the femoral neck-shaft angle, femoral neck offset, and lateral center-edge angle using x-rays of 18 osteonecrosis cases and 46 control children treated for newly diagnosed ALL on a single protocol. Despite adequate statistical power, we found no association between hip anatomy and osteonecrosis. Investigation of other factors contributing to ALL-associated osteonecrosis is warranted.
Collapse
Affiliation(s)
- Mary V. Portera
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Seth E. Karol
- Department of Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, TN, USA,Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Colton Smith
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Cheng Cheng
- Department of Biostatics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Michael D. Neel
- Department of Orthopaedics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sue C. Kaste
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
75
|
Kuhlen M, Kunstreich M, Krull K, Meisel R, Borkhardt A. Osteonecrosis in children and adolescents with acute lymphoblastic leukemia: a therapeutic challenge. Blood Adv 2017; 1:981-994. [PMID: 29296741 PMCID: PMC5737600 DOI: 10.1182/bloodadvances.2017007286] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/18/2017] [Indexed: 01/19/2023] Open
Abstract
Osteonecrosis (ON) represents one of the most common and debilitating sequelae of antileukemic treatment in children and adolescents with acute lymphoblastic leukemia (ALL). Systematic screening strategies can focus on early detection and intervention to prevent ON from progressing to stages associated with pain and functional impairment. These strategies hold promise for reducing ON-associated morbidity without the risk of impairing leukemia control. Herein, we critically reviewed clinical data on pharmacological, nonpharmacological/nonsurgical, and surgical (including cellular) treatment options for ON, which are covered in the literature and/or are conceivable based on the supposed underlying ON pathophysiology. Prevention of ON progression is of paramount importance, and attempts seem to be more effective in early (precollapse) disease status than in late-stage (collapse) ON. Based on the results of ongoing prospective magnetic resonance imaging screening studies, which will hopefully identify those patients with a high risk of ON progression and debilitating sequelae, prospective interventional studies are urgently needed. Although there is still a lack of high-quality studies, based on currently available data, core decompression surgery combined with cellular therapies (eg, employing mesenchymal stem cells) appears most promising for preventing joint infraction in children at high risk of developing late-stage ON.
Collapse
Affiliation(s)
- Michaela Kuhlen
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| | - Marina Kunstreich
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| | - Kathinka Krull
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| | - Roland Meisel
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
76
|
Al-Mahayri ZN, Patrinos GP, Ali BR. Pharmacogenomics in pediatric acute lymphoblastic leukemia: promises and limitations. Pharmacogenomics 2017; 18:687-699. [PMID: 28468529 DOI: 10.2217/pgs-2017-0005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/17/2017] [Indexed: 02/07/2023] Open
Abstract
Despite the significant advances achieved in pediatric acute lymphocytic leukemia (ALL) treatment, adverse side effects of drugs remain a challenging issue. Numerous ALL pharmacogenomic studies have been conducted to elucidate the predisposing genetic factors for their development. Plausible pharmacogenomic data are available for the osteonecrosis associated with glucocorticoids, the neurotoxicity associated with vincristine and the cardiotoxicity related to anthracyclines. However, these data have not been fully translated into the clinic due to several limitations, most importantly the lack of reliable evidence. The most robust pharmacogenomics data are those for thiopurines and methotrexate use, with evidence-based preemptive testing recommendations for the former. Pharmacogenomics has a significant potential utility in pediatric ALL treatment regimens. In this review, gaps and limitations in this field are emphasized, which may provide a useful guide for future research design.
Collapse
Affiliation(s)
- Zeina N Al-Mahayri
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, United Arab Emirates
| | - George P Patrinos
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, United Arab Emirates
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, Patras, Greece
| | - Bassam R Ali
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, United Arab Emirates
| |
Collapse
|
77
|
Alabdulwahab AS, Elsayed HG, Sherisher MA, Elbjeirami WM, Alieldin N. The Dana Farber Consortium Protocol for the Treatment of Adolescents and Young Adults With Acute Lymphoblastic Leukemia: A Single Institution Experience in Saudi Arabia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:320-325. [DOI: 10.1016/j.clml.2017.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 01/05/2017] [Accepted: 02/07/2017] [Indexed: 01/01/2023]
|
78
|
Mapes B, El Charif O, Al-Sawwaf S, Dolan ME. Genome-Wide Association Studies of Chemotherapeutic Toxicities: Genomics of Inequality. Clin Cancer Res 2017; 23:4010-4019. [PMID: 28442506 DOI: 10.1158/1078-0432.ccr-17-0429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/15/2017] [Accepted: 04/18/2017] [Indexed: 12/21/2022]
Abstract
With an estimated global population of cancer survivors exceeding 32 million and growing, there is a heightened awareness of the long-term toxicities resulting from cancer treatments and their impact on quality of life. Unexplained heterogeneity in the persistence and development of toxicities, as well as an incomplete understanding of their mechanisms, have generated a growing need for the identification of predictive pharmacogenomic markers. Early studies addressing this need used a candidate gene approach; however, over the last decade, unbiased and comprehensive genome-wide association studies (GWAS) have provided markers of phenotypic risk and potential targets to explore the mechanistic and regulatory pathways of biological functions associated with chemotherapeutic toxicity. In this review, we provide the current status of GWAS of chemotherapeutic toxicities with an emphasis on examining the ancestral diversity of the representative cohorts within these studies. Persistent calls to incorporate both ancestrally diverse and/or admixed populations into genomic efforts resulted in a recent rise in the number of studies utilizing cohorts of East Asian descent; however, few pharmacogenomic studies to date include cohorts of African, Indigenous American, Southwest Asian, and admixed populations. Through comprehensively evaluating sample size, composition by ancestry, genome-wide significant variants, and population-specific minor allele frequencies as reported by HapMap/dbSNP using NCBI PubMed and the NHGRI-EBI GWAS Catalog, we illustrate how allele frequencies and effect sizes tend to vary among individuals of differing ancestries. In an era of personalized medicine, the lack of diversity in genome-wide studies of anticancer agent toxicity may contribute to the health disparity gap. Clin Cancer Res; 23(15); 4010-9. ©2017 AACR.
Collapse
Affiliation(s)
- Brandon Mapes
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Omar El Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois.
| |
Collapse
|
79
|
Schmiegelow K, Müller K, Mogensen SS, Mogensen PR, Wolthers BO, Stoltze UK, Tuckuviene R, Frandsen T. Non-infectious chemotherapy-associated acute toxicities during childhood acute lymphoblastic leukemia therapy. F1000Res 2017; 6:444. [PMID: 28413626 PMCID: PMC5389408 DOI: 10.12688/f1000research.10768.1] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2017] [Indexed: 01/19/2023] Open
Abstract
During chemotherapy for childhood acute lymphoblastic leukemia, all organs can be affected by severe acute side effects, the most common being opportunistic infections, mucositis, central or peripheral neuropathy (or both), bone toxicities (including osteonecrosis), thromboembolism, sinusoidal obstruction syndrome, endocrinopathies (especially steroid-induced adrenal insufficiency and hyperglycemia), high-dose methotrexate-induced nephrotoxicity, asparaginase-associated hypersensitivity, pancreatitis, and hyperlipidemia. Few of the non-infectious acute toxicities are associated with clinically useful risk factors, and across study groups there has been wide diversity in toxicity definitions, capture strategies, and reporting, thus hampering meaningful comparisons of toxicity incidences for different leukemia protocols. Since treatment of acute lymphoblastic leukemia now yields 5-year overall survival rates above 90%, there is a need for strategies for assessing the burden of toxicities in the overall evaluation of anti-leukemic therapy programs.
Collapse
Affiliation(s)
- Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Signe Sloth Mogensen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Pernille Rudebeck Mogensen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Diabetes and Metabolism, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Benjamin Ole Wolthers
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Kristoffer Stoltze
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Ruta Tuckuviene
- Department of Pediatrics, Aalborg University Hospital, Aalborg, Denmark
| | - Thomas Frandsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
80
|
Liu Y, Fernandez CA, Smith C, Yang W, Cheng C, Panetta JC, Kornegay N, Liu C, Ramsey LB, Karol SE, Janke LJ, Larsen EC, Winick N, Carroll WL, Loh ML, Raetz EA, Hunger SP, Devidas M, Yang JJ, Mullighan CG, Zhang J, Evans WE, Jeha S, Pui CH, Relling MV. Genome-Wide Study Links PNPLA3 Variant With Elevated Hepatic Transaminase After Acute Lymphoblastic Leukemia Therapy. Clin Pharmacol Ther 2017; 102:131-140. [PMID: 28090653 DOI: 10.1002/cpt.629] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/04/2017] [Accepted: 01/11/2017] [Indexed: 12/12/2022]
Abstract
Remission induction therapy for acute lymphoblastic leukemia (ALL) includes medications that may cause hepatotoxicity, including asparaginase. We used a genome-wide association study to identify loci associated with elevated alanine transaminase (ALT) levels after induction therapy in children with ALL enrolled on St. Jude Children's Research Hospital (SJCRH) protocols. Germline DNA was genotyped using arrays and exome sequencing. Adjusting for age, body mass index, ancestry, asparaginase preparation, and dosage, the PNPLA3 rs738409 (C>G) I148M variant, previously associated with fatty liver disease risk, had the strongest genetic association with ALT (P = 2.5 × 10-8 ). The PNPLA3 rs738409 variant explained 3.8% of the variability in ALT, and partly explained race-related differences in ALT. The PNPLA3 rs738409 association was replicated in an independent cohort of 2,285 patients treated on Children's Oncology Group protocol AALL0232 (P = 0.024). This is an example of a pharmacogenetic variant overlapping with a disease risk variant.
Collapse
Affiliation(s)
- Y Liu
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - C A Fernandez
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - C Smith
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - W Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - C Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - J C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - N Kornegay
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - C Liu
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - L B Ramsey
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - S E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - L J Janke
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - E C Larsen
- Maine Children's Cancer Program, Scarborough, Maine, USA
| | - N Winick
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - W L Carroll
- Department of Pediatrics, New York University Langone Medical Center, New York, New York, USA
| | - M L Loh
- Department of Pediatrics, University of California School of Medicine, San Francisco, California, USA
| | - E A Raetz
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - S 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, USA
| | - M Devidas
- Department of Biostatistics, Colleges of Medicine, Public Health & Health Professions, University of Florida, Gainesville, Florida, USA
| | - J J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - C G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - J Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - W E Evans
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - S Jeha
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - C-H Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - M V Relling
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
81
|
Maxwell RR, Cole PD. Pharmacogenetic Predictors of Treatment-Related Toxicity Among Children With Acute Lymphoblastic Leukemia. Curr Hematol Malig Rep 2017; 12:176-186. [DOI: 10.1007/s11899-017-0376-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
82
|
Amin NL, Feltbower R, Kinsey S, Vora A, James B. Osteonecrosis in patients with acute lymphoblastic leukaemia: a national questionnaire study. BMJ Paediatr Open 2017; 1:e000122. [PMID: 29637145 PMCID: PMC5862222 DOI: 10.1136/bmjpo-2017-000122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVES To establish prevalence, management and long-term outcomes of osteonecrosis (ON) in young people diagnosed with acute lymphoblastic leukaemia (ALL) between 2003 and 2011. DESIGN SETTING PARTICIPANTS This study assessed ON in 3113 patients aged 1-24 years who participated in the UK national leukaemia study UKALL 2003. UKALL 2003 recruited patients in 40 UK hospitals between 2003 and 2011 and included patients between ages 1 and 25 diagnosed with ALL. RESULTS 170 patients were diagnosed with ON, giving a prevalence of 5.5%. The multivariable analysis showed that the risk of ON was highest for children aged between 10 and 20 years (ages 10-15 years, OR 23.7, 95% CI 14.8 to 38.0; ages 16-20 years, OR 22.5, 95% CI 12.7 to 39.8, compared with age <10 years). Among ethnic groups, Asian patients had the highest risk of ON (OR 1.92, 95% CI 1.1 to 3.6, compared with White patients). Eighty-five per cent of patients with ON had multifocal ON. Thirty-eight per cent of patients with ON required surgery and 19% of patients with ON required a hip replacement. Fifteen per cent of patients who had surgery still describe significant disability or use of a wheelchair. CONCLUSIONS ON has considerable morbidity for patients being treated for ALL, with a high burden of surgery. Age and ethnicity were found to be the most significant risk factors for development of ON, with Asian patients and patients aged 10-20 years at diagnosis of ALL at greatest risk. These results will help risk stratify patients at diagnosis of ALL, and help tailor future prospective studies in this area.
Collapse
Affiliation(s)
- Nadia Laila Amin
- Paediatric haematology, Leeds Children's Hospital, Leeds, UK.,University of Leeds, Leeds, UK
| | | | - Sally Kinsey
- Paediatric haematology, Leeds Children's Hospital, Leeds, UK.,University of Leeds, Leeds, UK
| | - Ajay Vora
- Paediatric haematology, Great Ormond Street Hospital, London, UK
| | - Beki James
- Paediatric haematology, Leeds Children's Hospital, Leeds, UK
| |
Collapse
|
83
|
Kunstreich M, Kummer S, Laws HJ, Borkhardt A, Kuhlen M. Osteonecrosis in children with acute lymphoblastic leukemia. Haematologica 2016; 101:1295-1305. [PMID: 27742768 DOI: 10.3324/haematol.2016.147595] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/23/2016] [Indexed: 11/09/2022] Open
Abstract
The morbidity and toxicity associated with current intensive treatment protocols for acute lymphoblastic leukemia in childhood become even more important as the vast majority of children can be cured and become long-term survivors. Osteonecrosis is one of the most common therapy-related and debilitating side effects of anti-leukemic treatment and can adversely affect long-term quality of life. Incidence and risk factors vary substantially between study groups and therapeutic regimens. We therefore analyzed 22 clinical trials of childhood acute lymphoblastic leukemia in terms of osteonecrosis incidence and risk factors. Adolescent age is the most significant risk factor, with patients >10 years old at the highest risk. Uncritical modification or even significant reduction of glucocorticoid dosage cannot be recommended at this stage. A novel and innovative approach to reduce osteonecrosis-associated morbidity might be systematic early screening for osteonecrosis by serial magnetic resonance images. However, discriminating patients at risk of functional impairment and debilitating progressive joint disease from asymptomatic patients still remains challenging.
Collapse
Affiliation(s)
- Marina Kunstreich
- University of Duesseldorf, Medical Faculty, Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Germany
| | - Sebastian Kummer
- University of Duesseldorf, Medical Faculty, Department of General Pediatrics, Neonatology and Pediatric Cardiology, Center for Child and Adolescent Health, Germany
| | - Hans-Juergen Laws
- University of Duesseldorf, Medical Faculty, Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Germany
| | - Arndt Borkhardt
- University of Duesseldorf, Medical Faculty, Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Germany
| | - Michaela Kuhlen
- University of Duesseldorf, Medical Faculty, Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Germany
| |
Collapse
|
84
|
Denny JC, Bastarache L, Roden DM. Phenome-Wide Association Studies as a Tool to Advance Precision Medicine. Annu Rev Genomics Hum Genet 2016; 17:353-73. [PMID: 27147087 PMCID: PMC5480096 DOI: 10.1146/annurev-genom-090314-024956] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Beginning in the early 2000s, the accumulation of biospecimens linked to electronic health records (EHRs) made possible genome-phenome studies (i.e., comparative analyses of genetic variants and phenotypes) using only data collected as a by-product of typical health care. In addition to disease and trait genetics, EHRs proved a valuable resource for analyzing pharmacogenetic traits and developing reverse genetics approaches such as phenome-wide association studies (PheWASs). PheWASs are designed to survey which of many phenotypes may be associated with a given genetic variant. PheWAS methods have been validated through replication of hundreds of known genotype-phenotype associations, and their use has differentiated between true pleiotropy and clinical comorbidity, added context to genetic discoveries, and helped define disease subtypes, and may also help repurpose medications. PheWAS methods have also proven to be useful with research-collected data. Future efforts that integrate broad, robust collection of phenotype data (e.g., EHR data) with purpose-collected research data in combination with a greater understanding of EHR data will create a rich resource for increasingly more efficient and detailed genome-phenome analysis to usher in new discoveries in precision medicine.
Collapse
Affiliation(s)
- Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37203;
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37203;
| | - Dan M Roden
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37203;
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
85
|
Genetic Biomarkers to Identify the Risk of Osteonecrosis in Children with Acute Lymphoblastic Leukemia. Mol Diagn Ther 2016; 20:519-522. [PMID: 27365083 DOI: 10.1007/s40291-016-0226-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Osteonecrosis is a disabling complication of treatment for pediatric acute lymphoblastic leukemia, and much effort has been made to predict which patients are prone to develop this disease. Multiple clinical and genetic factors have already been identified as being associated with osteonecrosis; however, a prediction model that combines pretreatment genetic biomarkers and clinical factors has not yet been designed. Such a prediction model can only be developed with continuing international collaborations and research efforts, including large genome-wide association studies.
Collapse
|
86
|
Richesson RL, Sun J, Pathak J, Kho AN, Denny JC. Clinical phenotyping in selected national networks: demonstrating the need for high-throughput, portable, and computational methods. Artif Intell Med 2016; 71:57-61. [PMID: 27506131 PMCID: PMC5480212 DOI: 10.1016/j.artmed.2016.05.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/30/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The combination of phenomic data from electronic health records (EHR) and clinical data repositories with dense biological data has enabled genomic and pharmacogenomic discovery, a first step toward precision medicine. Computational methods for the identification of clinical phenotypes from EHR data will advance our understanding of disease risk and drug response, and support the practice of precision medicine on a national scale. METHODS Based on our experience within three national research networks, we summarize the broad approaches to clinical phenotyping and highlight the important role of these networks in the progression of high-throughput phenotyping and precision medicine. We provide supporting literature in the form of a non-systematic review. RESULTS The practice of clinical phenotyping is evolving to meet the growing demand for scalable, portable, and data driven methods and tools. The resources required for traditional phenotyping algorithms from expert defined rules are significant. In contrast, machine learning approaches that rely on data patterns will require fewer clinical domain experts and resources. CONCLUSIONS Machine learning approaches that generate phenotype definitions from patient features and clinical profiles will result in truly computational phenotypes, derived from data rather than experts. Research networks and phenotype developers should cooperate to develop methods, collaboration platforms, and data standards that will enable computational phenotyping and truly modernize biomedical research and precision medicine.
Collapse
Affiliation(s)
- Rachel L Richesson
- Duke University School of Nursing, 311 Trent Drive, Durham, NC 27710 USA.
| | - Jimeng Sun
- School of Computational Science and Engineering, Georgia Institute of Technology, 266 Ferst Drive, Atlanta, GA 30313, USA.
| | - Jyotishman Pathak
- Department of Health Sciences Research, 200 1st Street SW, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Abel N Kho
- Departments of Medicine and Preventive Medicine, Northwestern University, 633 N St. Clair St. 20th floor. Chicago IL 60611, USA.
| | - Joshua C Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, 2525 West End Ave, Suite 672, Nashville, TN 37203, USA.
| |
Collapse
|
87
|
Mah W, Sonkusare SK, Wang T, Azeddine B, Pupavac M, Carrot-Zhang J, Hong K, Majewski J, Harvey EJ, Russell L, Chalk C, Rosenblatt DS, Nelson MT, Séguin C. Gain-of-function mutation in TRPV4 identified in patients with osteonecrosis of the femoral head. J Med Genet 2016; 53:705-9. [PMID: 27330106 PMCID: PMC5035228 DOI: 10.1136/jmedgenet-2016-103829] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/25/2016] [Indexed: 12/11/2022]
Abstract
Background Osteonecrosis of the femoral head is a debilitating disease that involves impaired blood supply to the femoral head and leads to femoral head collapse. Methods We use whole-exome sequencing and Sanger sequencing to analyse a family with inherited osteonecrosis of the femoral head and fluorescent Ca2+ imaging to functionally characterise the variant protein. Results We report a family with four siblings affected with inherited osteonecrosis of the femoral head and the identification of a c.2480_2483delCCCG frameshift deletion followed by a c.2486T>A substitution in one allele of the transient receptor potential vanilloid 4 (TRPV4) gene. TRPV4 encodes a Ca2+-permeable cation channel known to play a role in vasoregulation and osteoclast differentiation. While pathogenic TRPV4 mutations affect the skeletal or nervous systems, association with osteonecrosis of the femoral head is novel. Functional measurements of Ca2+ influx through mutant TRPV4 channels in HEK293 cells and patient-derived dermal fibroblasts identified a TRPV4 gain of function. Analysis of channel open times, determined indirectly from measurement of TRPV4 activity within a cluster of TRPV4 channels, revealed that the TRPV4 gain of function was caused by longer channel openings. Conclusions These findings identify a novel TRPV4 mutation implicating TRPV4 and altered calcium homeostasis in the pathogenesis of osteonecrosis while reinforcing the importance of TRPV4 in bone diseases and vascular endothelium.
Collapse
Affiliation(s)
- Wayne Mah
- Division of Hematology and Oncology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Swapnil K Sonkusare
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Tracy Wang
- Division of Hematology and Oncology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Bouziane Azeddine
- Division of Hematology and Oncology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Mihaela Pupavac
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Jian Carrot-Zhang
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada Department of Human Genetics, McGill University and Genome Québec Innovation Centre, Montreal, Quebec, Canada
| | - Kwangseok Hong
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada Department of Human Genetics, McGill University and Genome Québec Innovation Centre, Montreal, Quebec, Canada
| | - Edward J Harvey
- Department of Surgery, Division of Orthopaedic Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Laura Russell
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Colin Chalk
- Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - David S Rosenblatt
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Mark T Nelson
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Chantal Séguin
- Division of Hematology and Oncology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
88
|
Hao C, Yang S, Xu W, Shen JK, Ye S, Liu X, Dong Z, Xiao B, Feng Y. MiR-708 promotes steroid-induced osteonecrosis of femoral head, suppresses osteogenic differentiation by targeting SMAD3. Sci Rep 2016; 6:22599. [PMID: 26932538 PMCID: PMC4773864 DOI: 10.1038/srep22599] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/17/2016] [Indexed: 01/03/2023] Open
Abstract
Steroid-induced osteonecrosis of femoral head (ONFH) is a serious complication of glucocorticoid (GC) use. We investigated the differential expression of miRs in the mesenchymal stem cells (MSCs) of patients with ONFH, and aimed to explain the relationship between GC use and the development of MSC dysfunction in ONFH. Cells were collected from bone marrow of patients with ONFH. Samples were assigned to either GCs Group or Control Group at 1:1 matched with control. We then used miRNA microarray analysis and real-time PCR to identify the differentially expressed miRs. We also induced normal MSCs with GCs to verify the differential expression above. Subsequently, we selected some of the miRs for further studies, including miRNA target and pathway prediction, and functional analysis. We discovered that miR-708 was upregulated in ONFH patients and GC-treated MSCs. SMAD3 was identified as a direct target gene of miR-708, and functional analysis demonstrated that miR-708 could markedly suppress osteogenic differentiation and adipogenesis differentiation of MSCs. Inhibition of miR-708 rescued the suppressive effect of GC on osteonecrosis. Therefore, we determined that GC use resulted in overexpression of miR-708 in MSCs, and thus, targeting miR-708 may serve as a novel therapeutic biomarker for the prevention and treatment of ONFH.
Collapse
Affiliation(s)
- Cheng Hao
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Shuhua Yang
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Weihua Xu
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jacson K Shen
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| | - Shunan Ye
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xianzhe Liu
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Zhe Dong
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Baojun Xiao
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yong Feng
- Orthopedic Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.,Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| |
Collapse
|
89
|
Roden DM, Denny JC. Integrating electronic health record genotype and phenotype datasets to transform patient care. Clin Pharmacol Ther 2016; 99:298-305. [PMID: 26667791 PMCID: PMC4760864 DOI: 10.1002/cpt.321] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/11/2015] [Accepted: 12/11/2015] [Indexed: 12/16/2022]
Abstract
The Health Information Technology for Economic and Clinical Health (HITECH) Act of 2009 mandates the development and implementation of electronic health record (EHR) systems across the country. While a primary goal is to improve the care of individual patients, EHRs are also key enabling resources for a vision of individualized (or personalized or precision) medicine: the aggregation of multiple EHRs within or across healthcare systems should allow discovery of patient subsets that have unusual and definable clinical trajectories that deviate importantly from the expected response in a "typical" patient. The spectrum of such personalized care can then extend from prevention to choice of medication to intensity or nature of follow-up.
Collapse
Affiliation(s)
- D M Roden
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J C Denny
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
90
|
Kaul S, Korgenski EK, Ying J, Ng CF, Smits‐Seemann RR, Nelson RE, Andrews S, Raetz E, Fluchel M, Lemons R, Kirchhoff AC. A retrospective analysis of treatment-related hospitalization costs of pediatric, adolescent, and young adult acute lymphoblastic leukemia. Cancer Med 2016; 5:221-9. [PMID: 26714675 PMCID: PMC4735779 DOI: 10.1002/cam4.583] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 12/14/2022] Open
Abstract
This retrospective study examined the longitudinal hospital outcomes (costs adjusted for inflation, hospital days, and admissions) associated with the treatment of pediatric, adolescent, and young adult acute lymphoblastic leukemia (ALL). Patients between one and 26 years of age with newly diagnosed ALL, who were treated at Primary Children's Hospital (PCH) in Salt Lake City, Utah were included. Treatment and hospitalization data were retrieved from system-wide cancer registry and enterprise data warehouse. PCH is a member of the Children's Oncology Group (COG) and patients were treated on, or according to, active COG protocols. Treatment-related hospital costs of ALL were examined by computing the average annual growth rates (AAGR). Longitudinal regressions identified patient characteristics associated with costs. A total of 505 patients (46.9% female) were included. The majority of patients had B-cell lineage ALL, 6.7% had T-ALL, and the median age at diagnosis was 4 years. Per-patient, first-year ALL hospitalization costs at PCH rose from $24,197 in 1998 to $37,924 in 2012. The AAGRs were 6.1, 13.0, and 7.6% for total, pharmacy, and room and care costs, respectively. Average days (AAGR = 5.2%) and admissions (AAGR = 3.8%) also demonstrated an increasing trend. High-risk patients had 47% higher costs per 6-month period in the first 5 years from diagnosis than standard-risk patients (P < 0.001). Similarly, relapsed ALL and stem cell transplantations were associated with significantly higher costs than nonrelapsed and no transplantations, respectively (P < 0.001). Increasing treatment-related costs of ALL demonstrate an area for further investigation. Value-based interventions such as identifying low-risk fever and neutropenia patients and managing them in outpatient settings should be evaluated for reducing the hospital burden of ALL.
Collapse
Affiliation(s)
| | | | - Jian Ying
- University of UtahSalt Lake CityUtah
| | - Christi F. Ng
- Tufts University School of MedicineBostonMassachusetts
| | | | | | | | - Elizabeth Raetz
- University of UtahSalt Lake CityUtah
- Primary Children's HospitalSalt Lake CityUtah
| | - Mark Fluchel
- University of UtahSalt Lake CityUtah
- Primary Children's HospitalSalt Lake CityUtah
| | - Richard Lemons
- University of UtahSalt Lake CityUtah
- Primary Children's HospitalSalt Lake CityUtah
| | | |
Collapse
|
91
|
Padhye B, Dalla-Pozza L, Little D, Munns C. Incidence and outcome of osteonecrosis in children and adolescents after intensive therapy for acute lymphoblastic leukemia (ALL). Cancer Med 2016; 5:960-7. [PMID: 26792372 PMCID: PMC4864825 DOI: 10.1002/cam4.645] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 12/14/2015] [Accepted: 12/27/2015] [Indexed: 12/22/2022] Open
Abstract
Osteonecrosis (ON), a significant complication following treatment of acute lymphoblastic leukemia (ALL), has a profound impact on quality of life of ALL survivors. We studied incidence and outcome of ON in patients treated on or according to Australian and New Zealand Children's Haematology/ Oncology Group (ANZCHOG) study 8 at The Children's Hospital at Westmead. The study involved retrospective chart review of the patients. ON was defined by development of symptoms and confirmed by magnetic resonance imaging. From 2002-2011, 251 patients (143M, 108F, 59 Standard Risk (SR), 159 Medium Risk (MR) 5 High Risk (HR), and 28 Very high risk (VHR)) were treated according to study 8. Eighteen (7M, 11F, 2 SR, 12 MR, 4 VHR) patients developed ON (7.2%). Median age at diagnosis was 13.05 years(4.3-16.7). Incidence of ON in patients > 10 years at diagnosis was 29%. Six out of 18 patients developed ON after allogeneic stem cell transplantation. Median time from diagnosis to the development of ON following chemotherapy for ALL was 1.15 years (range 0.25-2.12). Most patients were treated with intravenous Zoledronic acid. At last follow-up, three patients had undergone arthroplasty, two patients were symptom free, and the remaining 13 patients reported persistent pain with activity. A majority of patients with ON of the hips had radiological progression. Overall, 7% of patients with ALL developed ON. Age >10 years was the most important risk factor. At last follow-up, 70% of patients had persistent symptoms. Although Zoledronic acid improved pain, most patients with ON of the hips had radiological progression.
Collapse
Affiliation(s)
- Bhavna Padhye
- Department of Oncology, The Children's Hospital at Westmead, Sydney, Australia
| | - Luciano Dalla-Pozza
- Department of Oncology, The Children's Hospital at Westmead, Sydney, Australia
| | - David Little
- Department Orthopaedics, The Children's Hospital at Westmead, Sydney, Australia
| | - Craig Munns
- Department of Endocrinology, The Children's Hospital at Westmead, Sydney, Australia
| |
Collapse
|
92
|
Abstract
In this issue of Blood, Karol et al establish a link between glutamate receptor polymorphisms and osteonecrosis, using a large discovery cohort and 2 validation cohorts. One validation cohort included patients under treatment of acute lymphoblastic leukemia (ALL) and the second is patients receiving glucocorticoid treatment of various other indications.
Collapse
|
93
|
Jackson RK, Irving JAE, Veal GJ. Personalization of dexamethasone therapy in childhood acute lymphoblastic leukaemia. Br J Haematol 2016; 173:13-24. [DOI: 10.1111/bjh.13924] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Rosanna K. Jackson
- Northern Institute for Cancer Research; Newcastle University; Newcastle upon Tyne UK
| | - Julie A. E. Irving
- Northern Institute for Cancer Research; Newcastle University; Newcastle upon Tyne UK
| | - Gareth J. Veal
- Northern Institute for Cancer Research; Newcastle University; Newcastle upon Tyne UK
| |
Collapse
|
94
|
Genetic risk factors for the development of osteonecrosis in children under age 10 treated for acute lymphoblastic leukemia. Blood 2015; 127:558-64. [PMID: 26590194 DOI: 10.1182/blood-2015-10-673848] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/10/2015] [Indexed: 11/20/2022] Open
Abstract
Osteonecrosis is a dose-limiting toxicity in the treatment of pediatric acute lymphoblastic leukemia (ALL). Prior studies on the genetics of osteonecrosis have focused on patients ≥10 years of age, leaving the genetic risk factors for the larger group of children <10 years incompletely understood. Here, we perform the first evaluation of genetic risk factors for osteonecrosis in children <10 years. The discovery cohort comprised 82 cases of osteonecrosis and 287 controls treated on Children's Oncology Group (COG) standard-risk ALL protocol AALL0331 (NCT00103285, https://clinicaltrials.gov/ct2/show/NCT00103285), with results tested for replication in 817 children <10 years treated on COG protocol AALL0232 (NCT00075725, https://clinicaltrials.gov/ct2/show/NCT00075725). The top replicated single nucleotide polymorphisms (SNPs) were near bone morphogenic protein 7 [BMP7: rs75161997, P = 5.34 × 10(-8) (odds ratio [OR] 15.0) and P = .0498 (OR 8.44) in the discovery and replication cohorts, respectively] and PROX1-antisense RNA1 (PROX1-AS1: rs1891059, P = 2.28 × 10(-7) [OR 6.48] and P = .0077 [OR 3.78] for the discovery and replication cohorts, respectively). The top replicated nonsynonymous SNP, rs34144324, was in a glutamate receptor gene (GRID2, P = 8.65 × 10(-6) [OR 3.46] and P = .0136 [OR 10.8] in the discovery and replication cohorts, respectively). In a meta-analysis, the BMP7 and PROX1-AS1 variants (rs75161997 and rs1891059, respectively) met the significance threshold of <5 × 10(-8). Top replicated SNPs were enriched in enhancers active in mesenchymal stem cells, and analysis of annotated genes demonstrated enrichment in glutamate receptor and adipogenesis pathways. These data may provide new insights into the pathophysiology of osteonecrosis.
Collapse
|