Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Blood Adv ; 8(9): 2094-2103, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38295280

ABSTRACT

ABSTRACT: We sought to define the cooccurring mutational profile of FLT3-ITD-positive (ITDpos) acute myeloid leukemia (AML) in pediatric and young adult patients and to define the prognostic impact of cooperating mutations. We identified 464 patients with FLT3-ITD mutations treated on Children's Oncology Group trials with available sequencing and outcome data. Overall survival, event-free survival (EFS), and relapse risk were determined according to the presence of cooccurring risk stratifying mutations. Among the cohort, 79% of patients had cooccurring alterations across 239 different genes that were altered through mutations or fusions. Evaluation of the prognostic impact of the cooccurring mutations demonstrated that patients with ITDpos AML experienced significantly different outcomes according to the cooccurring mutational profile. Patients with ITDpos AML harboring a cooccurring favorable-risk mutation of NPM1, CEBPA, t(8;21), or inv(16) experienced a 5-year EFS of 64%, which was significantly superior to of 22.2% for patients with ITDpos AML and poor-risk mutations of WT1, UBTF, or NUP98::NSD1 as well to 40.9% for those who lacked either favorable-risk or poor-risk mutation (ITDpos intermediate; P < .001 for both). Multivariable analysis demonstrated that cooccurring mutations had significant prognostic impact, whereas allelic ratio had no impact. Therapy intensification, specifically consolidation transplant in remission, resulted in significant improvements in survival for ITDpos AML. However, patients with ITDpos/NUP98::NSD1 continued to have poor outcomes with intensified therapy, including sorafenib. Cooccurring mutational profile in ITDpos AML has significant prognostic impacts and is critical to determining risk stratification and therapeutic allocation. These clinical trials were registered at www.clinicaltrials.gov as NCT00002798, NCT00070174, NCT00372593, and NCT01371981.


Subject(s)
Leukemia, Myeloid, Acute , Mutation , Nucleophosmin , fms-Like Tyrosine Kinase 3 , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/diagnosis , fms-Like Tyrosine Kinase 3/genetics , Child , Prognosis , Adolescent , Female , Male , Child, Preschool , Infant , Young Adult , Adult
2.
Front Oncol ; 11: 744739, 2021.
Article in English | MEDLINE | ID: mdl-34540703

ABSTRACT

PURPOSE: Medulloblastoma is one of the most common malignant brain tumors in children. To date, the treatment of average-risk (non-metastatic, completely resected) medulloblastoma includes craniospinal radiation therapy and adjuvant chemotherapy. Modern treatment modalities and now risk stratification of subgroups have extended the survival of these patients, exposing the long-term morbidities associated with radiation therapy. Prior to advances in molecular subgrouping, we sought to reduce the late effects of radiation in patients with average-risk medulloblastoma. METHODS: We performed a single-arm, multi-institution study, reducing the dose of craniospinal irradiation by 25% to 18 Gray (Gy) with the goal of maintaining the therapeutic efficacy as described in CCG 9892 with maintenance chemotherapy. RESULTS: Twenty-eight (28) patients aged 3-30 years were enrolled across three institutions between April 2001 and December 2010. Median age at enrollment was 9 years with a median follow-up time of 11.7 years. The 3-year relapse-free (RFS) and overall survival (OS) were 79% (95% confidence interval [CI] 58% to 90%) and 93% (95% CI 74% to 98%), respectively. The 5-year RFS and OS were 71% (95% CI 50% to 85%) and 86% (95% CI 66% to 94%), respectively. Toxicities were similar to those seen in other studies; there were no grade 5 toxicities. CONCLUSIONS: Given the known neurocognitive adverse effects associated with cranial radiation therapy, studies to evaluate the feasibility of dose reduction are needed. In this study, we demonstrate that select patients with average-risk medulloblastoma may benefit from a reduced craniospinal radiation dose of 18 Gy without impacting relapse-free or overall survival. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00031590.

3.
Clin Cancer Res ; 26(12): 2891-2897, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32122921

ABSTRACT

PURPOSE: Cytarabine is an effective treatment for AML with associated toxicities including treatment related mortality (TRM). The purpose is to determine the clinical relevance of SNPs identified through the use of HapMap lymphoblastoid cell-based models, in predicting cytarabine response and toxicity in AML. EXPERIMENTAL DESIGN: We tested clinical significance of SNPs associated with cytarabine sensitivity in children with AML treated on Children's Oncology Group regimens (CCG 2941/2961). Endpoints included overall survival (OS), event-free survival (EFS), and TRM. Patients who received bone marrow transplant were excluded. We tested 124 SNPs associated with cytarabine sensitivity in HapMap cell lines in 348 children to determine whether any associated with treatment outcomes. In addition, we tested five SNPs previously associated with TRM in children with AML in our independent dataset of 385 children. RESULTS: Homozygous variant genotypes of rs2025501 and rs6661575 had increased in vitro cellular sensitivity to cytarabine and were associated with increased TRM. TRM was particularly increased in children with variant genotype randomized to high-dose cytarabine (rs2025501: P = 0.0024 and rs6661575 P = 0.0188). In analysis of previously reported SNPs, only the variant genotype rs17202778 C/C was significantly associated with TRM (P < 0.0001). CONCLUSIONS: We report clinical importance of two SNPs not previously associated with cytarabine toxicity. Moreover, we confirm that SNP rs17202778 significantly impacts TRM in pediatric AML. Cytarabine sensitivity genotypes may predict TRM and could be used to stratify to standard versus high-dose cytarabine regimens, warranting further study in prospective AML trials.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/genetics , Cytarabine/therapeutic use , Leukemia, Myeloid, Acute/mortality , Polymorphism, Single Nucleotide , Child , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Prognosis , Prospective Studies , Survival Rate
4.
Lancet Oncol ; 18(1): 63-74, 2017 01.
Article in English | MEDLINE | ID: mdl-27914822

ABSTRACT

BACKGROUND: Sodium thiosulfate is an antioxidant shown in preclinical studies in animals to prevent cisplatin-induced hearing loss with timed administration after cisplatin without compromising the antitumour efficacy of cisplatin. The primary aim of this study was to assess sodium thiosulfate for prevention of cisplatin-induced hearing loss in children and adolescents. METHODS: ACCL0431 was a multicentre, randomised, open-label, phase 3 trial that enrolled participants at 38 participating Children's Oncology Group hospitals in the USA and Canada. Eligible participants aged 1-18 years with newly diagnosed cancer and normal audiometry were randomly assigned (1:1) to receive sodium thiosulfate or observation (control group) in addition to their planned cisplatin-containing chemotherapy regimen, using permuted blocks of four. Randomisation was initially stratified by age and duration of cisplatin infusion. Stratification by previous cranial irradiation was added later as a protocol amendment. The allocation sequence was computer-generated centrally and concealed to all personnel. Participants received sodium thiosulfate 16 g/m2 intravenously 6 h after each cisplatin dose or observation. The primary endpoint was incidence of hearing loss 4 weeks after final cisplatin dose. Hearing was measured using standard audiometry and reviewed centrally by audiologists masked to allocation using American Speech-Language-Hearing Association criteria but treatment was not masked for participants or clinicians. Analysis of the primary endpoint was by modified intention to treat, which included all randomly assigned patients irrespective of treatment received but restricted to those assessable for hearing loss. Enrolment is complete and this report represents the final analysis. This trial is registered with ClinicalTrials.gov, number NCT00716976. FINDINGS: Between June 23, 2008, and Sept 28, 2012, 125 eligible participants were randomly assigned to either sodium thiosulfate (n=61) or observation (n=64). Of these, 104 participants were assessable for the primary endpoint (sodium thiosulfate, n=49; control, n=55). Hearing loss was identified in 14 (28·6%; 95% CI 16·6-43·3) participants in the sodium thiosulfate group compared with 31 (56·4%; 42·3-69·7) in the control group (p=0·00022). Adjusted for stratification variables, the likelihood of hearing loss was significantly lower in the sodium thiosulfate group compared with the control group (odds ratio 0·31, 95% CI 0·13-0·73; p=0·0036). The most common grade 3-4 haematological adverse events reported, irrespective of attribution, were neutropenia (117 [66%] of 177 participant cycles in the sodium thiosulfate group vs 145 [65%] of 223 in the control group), whereas the most common non-haematological adverse event was hypokalaemia (25 [17%] of 147 vs 22 [12%] of 187). Of 194 serious adverse events reported in 26 participants who had received sodium thiosulfate, none were deemed probably or definitely related to sodium thiosulfate; the most common serious adverse event was decreased neutrophil count: 26 episodes in 14 participants. INTERPRETATION: Sodium thiosulfate protects against cisplatin-induced hearing loss in children and is not associated with serious adverse events attributed to its use. Further research is needed to define the appropriate role for sodium thiosulfate among emerging otoprotection strategies. FUNDING: US National Cancer Institute.


Subject(s)
Antineoplastic Agents/adverse effects , Antioxidants/adverse effects , Cisplatin/adverse effects , Hearing Loss/chemically induced , Neoplasms/drug therapy , Thiosulfates/adverse effects , Adolescent , Child , Child, Preschool , Drug Therapy, Combination , Female , Follow-Up Studies , Hearing Loss/epidemiology , Humans , Incidence , Infant , Male , Neoplasm Staging , Neoplasms/pathology , Prognosis , Survival Rate , United States/epidemiology
6.
J Natl Cancer Inst ; 106(8)2014 Aug.
Article in English | MEDLINE | ID: mdl-25080574

ABSTRACT

Survivors of childhood cancer frequently experience cancer-related cognitive dysfunction, commonly months to years after treatment for pediatric brain tumors, acute lymphoblastic leukemia (ALL), or tumors involving the head and neck. Risk factors for cancer-related cognitive dysfunction include young age at diagnosis, treatment with cranial irradiation, use of parenteral or intrathecal methotrexate, female sex, and pre-existing comorbidities. Limiting use and reducing doses and volume of cranial irradiation while intensifying chemotherapy have improved survival and reduced the severity of cognitive dysfunction, especially in leukemia. Nonetheless, problems in core functional domains of attention, processing speed, working memory and visual-motor integration continue to compromise quality of life and performance. We review the epidemiology, pathophysiology and assessment of cancer-related cognitive dysfunction, the impact of treatment changes for prevention, and the broad strategies for educational and pharmacological interventions to remediate established cognitive dysfunction following childhood cancer. The increased years of life saved after childhood cancer warrants continued study toward the prevention and remediation of cancer-related cognitive dysfunction, using uniform assessments anchored in functional outcomes.


Subject(s)
Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Neoplasms/psychology , Psychomotor Performance , Quality of Life , Survivors/psychology , Adolescent , Adult , Age Factors , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Attention , Benzhydryl Compounds/therapeutic use , Brain Neoplasms/psychology , Central Nervous System Stimulants/therapeutic use , Child , Child, Preschool , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/physiopathology , Comorbidity , Cranial Irradiation/adverse effects , Donepezil , Early Intervention, Educational/methods , Education, Special , Head and Neck Neoplasms/psychology , Human Growth Hormone/therapeutic use , Humans , Indans/therapeutic use , Infant , Injections, Spinal , Memory, Short-Term , Methotrexate/administration & dosage , Methotrexate/adverse effects , Methylphenidate/therapeutic use , Modafinil , Neurosurgical Procedures/adverse effects , Nootropic Agents/therapeutic use , Patient Education as Topic/methods , Piperidines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/psychology , Radiotherapy Dosage , Risk Factors , Severity of Illness Index , Sex Factors , Thinking , Wakefulness-Promoting Agents/therapeutic use , Young Adult
7.
Blood ; 124(15): 2400-7, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25145343

ABSTRACT

NUP98/NSD1 has recently been reported in association with poor outcome in acute myeloid leukemia (AML). Previous studies also observed a high overlap between NUP98/NSD1 and FLT3/ITD, raising the question as to whether the reported poor outcome is due to NUP98/NSD1 or caused by the co-occurrence of these 2 genetic lesions. We aimed to determine the prognostic significance of NUP98/NSD1 in the context of FLT3/ITD AML. A total of 1421 patients enrolled in 5 consecutive Children's Oncology Group/Children's Cancer Group and SWOG trials were evaluated. NUP98/NSD1 was found in 15% of FLT3/ITD and 7% of cytogenetically normal (CN)-AML. Those with dual FLT3/ITD and NUP98/NSD1 (82% of NUP98/NSD1 patients) had a complete remission rate of 27% vs 69% in FLT3/ITD without NUP98/NSD1 (P < .001). The corresponding 3-year overall survival was 31% vs 48% (P = .011), respectively. In CN-AML, patients with concomitant NUP98/NSD1 and FLT3/ITD had a worse outcome than those harboring NUP98/NSD1 only. In multivariate analysis, the dual NUP98/NSD1 and FLT3/ITD remained an independent predictor of poor outcome, and NUP98/NSD1 without FLT3/ITD lost its prognostic significance. Our study demonstrates that it is the interaction between NUP98/NSD1 and FLT3/ITD that determines the poor outcome of patients with NUP98/NSD1 disease.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/therapy , Oncogene Proteins, Fusion/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Adolescent , Age Distribution , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , Multivariate Analysis , Neoplasm, Residual/pathology , Regression Analysis , Remission Induction , Treatment Failure , Young Adult
8.
Br J Haematol ; 166(2): 254-259, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24661089

ABSTRACT

Acute myeloid leukaemia (AML) with t(6;9)(p23;q34) is a rare subtype associated with FLT3-internal tandem duplication (ITD) and poor outcomes. The clinical outcomes of paediatric patients with t(6;9) with and without FLT3-ITD treated on six consecutive cooperative trails were evaluated. In contrast to patients without t(6;9), those with t(6;9) had a significantly lower complete remission rate, higher relapse rate (RR), and poor overall survival (OS). Within t(6;9) patients, those with and without FLT3-ITD had an OS of 40% and 27% respectively (P > 0·9), demonstrating that t(6;9) is a high-risk cytogenetic feature in paediatric AML and its clinical impact is independent of the presence of FLT3-ITD.


Subject(s)
Chromosomes, Human, Pair 6/genetics , Chromosomes, Human, Pair 9/genetics , Leukemia, Myeloid, Acute/genetics , Translocation, Genetic , fms-Like Tyrosine Kinase 3/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Gene Duplication , Genes, Neoplasm , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/therapy , Male , Neoplasm Proteins/genetics , Prognosis , Recurrence , Remission Induction , Survival Analysis , Tandem Repeat Sequences , Treatment Outcome
9.
Pediatr Blood Cancer ; 61(2): 281-5, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24019241

ABSTRACT

BACKGROUND: To determine feasibility and safety of proactive enteral tube feeding (ETF) in pediatric oncology patients. METHODS: Pediatric patients with newly diagnosed brain tumors, myeloid leukemia or high-risk solid tumors were eligible. Subjects agreeing to start ETF before cycle 2 chemotherapy were considered proactive participants (PPs). Those who declined could enroll as chart collection receiving nutritional standard of care. Nutritional status was assessed using standard anthropometric measurements. Episodes of infection and toxicity related to ETF were documented from diagnosis to end of therapy. A descriptive comparison between PPs and controls was conducted. RESULTS: One hundred four eligible patients were identified; 69 enrolled (20 PPs and 49 controls). At diagnosis, 17% of all subjects were underweight and 26% overweight. Barriers to enrollment included physician, subject and/or family refusal, and inability to initiate ETF prior to cycle 2 of chemotherapy. Toxicity of ETF was minimal, but higher percentage of subjects in the proactive group had episodes of infection than controls. Thirty-nine percent of controls eventually started ETF and were twice as likely to receive parenteral nutrition. PPs experienced less weight loss at ETF initiation than controls receiving ETF and were the only group to demonstrate improved nutritional status at end of study. CONCLUSIONS: Proactive ETF is feasible in children with cancer and results in improved nutritional status at end of therapy. Episodes of infection in this study are concerning; therefore, a larger randomized trial is required to further delineate infectious risks and toxicities that may be mitigated by improved nutritional status.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Enteral Nutrition , Intubation, Gastrointestinal , Neoplasms/drug therapy , Neoplasms/rehabilitation , Nutritional Status , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Feasibility Studies , Female , Follow-Up Studies , Humans , Infant , Male , Pilot Projects , Prognosis , Young Adult
10.
Pediatr Blood Cancer ; 61(2): 269-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23955900

ABSTRACT

BACKGROUND: We sought to better define the role of hematopoietic cell transplantation (HCT) in first remission (CR1) for high-risk pediatric acute myeloid leukemia (AML). PROCEDURES: Outcomes were compared among patients aged less than 21 years with cytogenetically defined poor-risk AML treated with chemotherapy, matched related (MRD), or unrelated donor (URD) transplantation in CR1. Poor-risk cytogenetics was defined as monosomy 7/del7q, monosomy 5/del 5q, abnormalities of 3q, t(6;9)(p23;q34), or complex karyotype. Included are patients treated on Children's Oncology Group trials or reported to the Center for International Blood and Marrow Transplant Research from 1989 to 2006. RESULTS: Of the 233 patients, 123 received chemotherapy, 55 received MRD HCT, and 55 received URD HCT. The 5-year overall survival from the time of consolidation chemotherapy or transplant conditioning was similar: chemotherapy (43% ± 9%), MRD (46% ± 14%), or URD (50% ± 14%), P = 0.99. Similarly, multivariate analysis demonstrated no significant differences in survival [(reference group = chemotherapy); MRD HR 1.08, P = 0.76; URD HR 1.13, P = 0.67] despite lower relapse risk with URD HCT (HR = 0.43, P = 0.01). CONCLUSIONS: Our findings do not provide support for the preferential use of HCT over chemotherapy alone for children with cytogenetically defined poor-risk AML in CR1.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosome Aberrations , Donor Selection , Graft vs Host Disease/diagnosis , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/mortality , Adolescent , Adult , Child , Child, Preschool , Combined Modality Therapy , Cytogenetic Analysis , Female , Follow-Up Studies , Graft vs Host Disease/etiology , Graft vs Host Disease/genetics , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/therapy , Living Donors , Male , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Prognosis , Remission Induction , Risk Factors , Survival Rate , Transplantation, Autologous , Unrelated Donors , Young Adult
11.
Pediatr Blood Cancer ; 61(4): 647-52, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24127439

ABSTRACT

BACKGROUND: The inhibitor-of-apoptosis protein survivin, encoded by BIRC5, regulates apoptosis, cell division and proliferation. Several survivin splice variants have been described however, the prognostic significance of their expression has not been well defined in pediatric acute myeloid leukemia (AML). PROCEDURE: Quantitative expression analyses of BIRC5 mRNA (n = 306) and survivin transcript splice variants (n = 90) were performed on diagnostic bone marrow samples from children with de novo AML treated on the clinical trials CCG-2961 and AAML03P1, then correlated with disease characteristics and clinical outcome. RESULTS: Total BIRC5 expression did not correlate with clinical outcome. Fragment length analysis and sequencing of the entire BIRC5 transcript demonstrated three splice variants. The most prominent product, wild-type survivin, was expressed in all samples tested. Two minor transcripts were present in 90 patients treated on CCG-2961; survivin-2B and a novel variant, survivin-ΔEx2, characterized by deletion of BIRC5 exon II. A high 2B/ΔEx2 expression ratio (≥1) correlated with increased diagnostic WBC count, monocytic phenotype, +8 cytogenetics, lower complete remission (45% [n = 10] vs. 88% [n = 59], P < 0.001) and higher induction failure rates (23% [n = 5] vs. 3% [n = 2], P = 0.009). Consistent with this poor induction response, patients with a 2B/ΔEx2 ratio ≥1 had inferior 5-year survival rates (OS 36% vs. 60%, P = 0.011; EFS 23% vs. 53% at 5 years, P = 0.001) and appear to have increased relapse risk (P = 0.056). Subset analyses suggest that relative over-expression of 2B, rather than under-expression of ΔEx2 determines clinical response. CONCLUSIONS: High survivin-2B/ΔEx2 ratios are associated with refractory disease and inferior survival in childhood AML. Survivin splice variant expression warrants prospective evaluation in clinical trials.


Subject(s)
Alternative Splicing/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/genetics , Inhibitor of Apoptosis Proteins/genetics , Leukemia, Myeloid, Acute/mortality , Neoplasm Recurrence, Local/mortality , Child , Combined Modality Therapy , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Survival Rate , Survivin
12.
Cancer ; 119(23): 4162-9, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24104395

ABSTRACT

BACKGROUND: Studies comparing survival of adolescent and young adult (AYA) patients to that of younger patients with newly diagnosed acute myeloid leukemia (AML) have yielded conflicting results. In order to more accurately characterize relative survival and other outcomes of AYA patients, a cross-study analysis was conducted using data from recent trials conducted by the Children's Cancer Group (CCG) and Children's Oncology Group (COG). METHODS: Data were combined from the CCG-2891, CCG-2941, CCG-2961, and AAML03P1 trials. The data set included 1840 patients, comprising 238 AYA and 1602 younger patients. RESULTS: Overall survival was not significantly different in the 2 groups (AYA, 49% ± 7% versus younger, 54% ± 3% (± 2 standard errors), P = .058). Relapse was lower in AYA patients (30% ± 7% versus 41% ± 3%, P = .002), but treatment-related mortality (TRM) was higher (25% ± 6% versus 12% ± 2%, P < .001). After adjustment for other factors, older age remained strongly associated with TRM (hazard ratio = 2.30, 95% CI = 1.59-3.33, P < .001). Infection accounted for the excess TRM in AYA patients. CONCLUSIONS: Survival in AYA and younger patients with newly diagnosed AML is similar; however, older patients are at higher risk for TRM. More effective strategies for preventing mortality from infection in AYA patients are needed.


Subject(s)
Leukemia, Myeloid, Acute/mortality , Adolescent , Age Factors , Child , Female , Humans , Leukemia, Myeloid, Acute/therapy , Male , Young Adult
13.
Pediatr Blood Cancer ; 60(12): 2073-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24039149

ABSTRACT

BACKGROUND: Abnormalities of chromosome 5q (-5/5q-) are associated with poor prognosis in adults with acute myeloid leukemia (AML). However, there are no large studies on outcomes of children with -5/5q- AML. To determine the disease correlates of this group, we retrospectively analyzed cytogenetic data from five studies of childhood AML. PROCEDURE: Data from patients whose cytogenetic clones included -5/5q-, with the exception of those with acute promyelocytic leukemia or Down syndrome, were included. RESULTS: Of the 2,240 patients with cytogenetic data available, 26 (1.2%) had -5 or 5q-. A significant number of these patients were age 11-21 (61.5%, P = 0.031) and had M0 morphology compared with patients without -5/5q- (24.0% vs. 2.8%, P < 0.001). Twenty-two of the 26 patients had a complete remission (CR) response to induction chemotherapy. The 5-year overall survival (OS) from the time of diagnosis for the -5/5q- patients was significantly lower than for patients without -5/5q- (27 ± 17% vs. 50 ± 2%, P = 0.027). Similarly, from induction CR, patients with -5/5q- had significantly worse disease free survival, OS and relapse risk than those without this abnormality (27 ± 19% vs. 46 ± 2%, P = 0.035, 32 ± 20% vs. 57 ± 2%, P = 0.025, 68 ± 21% vs. 45 ± 2%, P = 0.01, respectively). CONCLUSIONS: Pediatric patients with AML and -5/5q- had a very poor outcome. These findings support the need for new or novel therapies for these patients.


Subject(s)
Chromosomes, Human, Pair 5/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Disease-Free Survival , Female , Gene Deletion , Humans , Infant , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Male , Prognosis , Retrospective Studies , Treatment Outcome , Young Adult
14.
Blood ; 121(7): 1083-93, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23243289

ABSTRACT

Signal transducer and activator of transcription 3 (Stat3) and Stat5 are critical signaling intermediates that promote survival in myeloid leukemias. We examined Stat3 and Stat5 activation patterns in resting and ligand-stimulated primary samples from pediatric patients with acute myeloid leukemia. Phosphorylated Stats were measured by FACS before and after stimulation with increasing doses of granulocyte-colony stimulating factor or IL-6. We also measured positive and negative regulators of Stat signaling, and we compared the variation in multiple parameters to identify biologic relationships. Levels of constitutively phosphorylated Stats were variable and did not correlate with survival. In terms of induced phospho-Stats, 15 of 139 specimens (11%) phosphorylated Stat3 in response to moderate doses of both granulocyte-colony stimulating factor and IL-6. Compared with groups that were resistant to 1 or both ligands, this pattern of dual sensitivity was associated with a superior outcome, with a 5-year event-free survival of 79% (P = .049) and 5-year overall survival of 100% (P = .006). This study provides important and novel insights into the biology of Stat3 and Stat5 signaling in acute myeloid leukemia. Patterns of ligand sensitivity may be valuable for improving risk identification, and for developing new agents for individualized therapy.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Adolescent , Cell Separation , Child , Child, Preschool , Disease-Free Survival , Female , Filgrastim , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Infant , Interleukin-6/pharmacology , Male , Phosphorylation , Prognosis , Recombinant Proteins/pharmacology , Risk Factors , Signal Transduction/drug effects , Survival Analysis , Young Adult
15.
Pediatr Blood Cancer ; 59(5): 922-4, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22517793

ABSTRACT

Specific cytogenetic clones might distinguish patients with unrecognized Fanconi anemia (FA) who present with acute myeloid leukemia (AML) from those with sporadic AML. Cytogenetic reports in literature cases of FA and AML were compared with de novo cases enrolled on CCG-2961. Gain of 1q, gain of 3q, monosomy 7, deleted 7q, gain of 13q, and deleted 20q were more frequent in FA AML; t(8;21), trisomy 8, t(9;11), t(6;9), and inversion 16 were exclusive to de novo AML cases. Observation of the FA AML cytogenetic clonal patterns should raise suspicion of an underlying leukemia predisposition syndrome and influence management.


Subject(s)
Chromosome Aberrations , Chromosomes, Human/genetics , Fanconi Anemia/genetics , Fanconi Anemia/therapy , Genetic Predisposition to Disease , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Adolescent , Adult , Child , Child, Preschool , Fanconi Anemia/complications , Female , Humans , Infant , Leukemia, Myeloid, Acute/complications , Male
16.
Pediatr Blood Cancer ; 58(5): 695-700, 2012 May.
Article in English | MEDLINE | ID: mdl-21618417

ABSTRACT

BACKGROUND: Recent studies suggest that polymorphisms in genes encoding enzymes involved in drug detoxification and metabolism may influence disease outcome in pediatric acute lymphoblastic leukemia (ALL). We sought to extend current knowledge by using standard and novel statistical methodology to examine polymorphic variants of genes and relapse risk, toxicity, and drug dose delivery in standard risk ALL. PROCEDURE: We genotyped and abstracted chemotherapy drug dose data from treatment roadmaps on 557 patients on the Children's Cancer Group ALL study, CCG-1891. Fourteen common polymorphisms in genes involved in folate metabolism and/or phase I and II drug detoxification were evaluated individually and clique-finding methodology was employed for detection of significant gene-gene interactions. RESULTS: After controlling for known risk factors, polymorphisms in four genes: GSTP1*B (HR = 1.94, P = 0.047), MTHFR (HR = 1.61, P = 0.034), MTRR (HR = 1.95, P = 0.01), and TS (3R/4R, HR = 3.69, P = 0.007) were found to significantly increase relapse risk. One gene-gene pair, MTRR A/G and GSTM1 null genotype, significantly increased the risk of relapse after correction for multiple comparisons (P = 0.012). Multiple polymorphisms were associated with various toxicities and there was no significant difference in dose of chemotherapy delivered by genotypes. CONCLUSIONS: These data suggest that various polymorphisms play a role in relapse risk and toxicity during childhood ALL therapy and that genotype does not play a role in adjustment of drug dose administered. Additionally, gene-gene interactions may increase the risk of relapse in childhood ALL and the clique method may have utility in further exploring these interactions. childhood ALL therapy.


Subject(s)
Polymorphism, Genetic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child, Preschool , Female , Genetic Variation , Glutathione S-Transferase pi/genetics , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methyltransferases/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Randomized Controlled Trials as Topic
17.
Pediatr Blood Cancer ; 58(4): 519-24, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21618422

ABSTRACT

BACKGROUND: Extramedullary leukemia (EML) is common in pediatric acute myeloid leukemia (AML) and occurs as leukemia cells within the cerebrospinal fluid (CSF) or as a solid tumor (myeloid sarcoma-MS). The effect of MS on survival is unknown. METHODS: Patients on CCG protocols 2861, 2891, 2941, and 2961 being treated for AML with intensive-timing chemotherapy were classified for the presence of EML (CSF leukemia, CNS-MS, orbital-MS, or non-CNS MS). CSF leukemia was classified as CNS3 (>5 WBC in the CSF with blasts) and non-CSF leukemia as CNS1/2 (<5 WBC in the CSF with or without blasts). Characteristics and outcomes of these patients were compared. RESULTS: Of the 1,459 total patients, 1,206 (82%) had no EML, 154 (11%) had CSF leukemia, 19 (1%) had CNS-MS, 23 (2%) had orbital-MS, and 57 (4%) had non-CNS MS. The CR rate was significantly higher in patients with orbital-MS and CNS-MS than in those with non-MS and non-CNS MS (96% and 95% vs. 78% and 78%, P = 0.034). Patients with orbital-MS and CNS-MS had significantly higher overall survival than patients with non-CNS MS (92% and 73% vs. 38%, P < 0.001), CNS3 patients (92% and 73% vs. 51, P < 0.001), and CNS1/2 patients (92% and 73% vs. 50%, P < 0.001). Patients with orbital-MS had a significantly lower relapse rate. CONCLUSION: Patients with MS involving orbital and CNS sites had a significantly better survival than patients with non-CNS MS, with CSF leukemia, or with no EML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Central Nervous System Neoplasms , Leukemia, Myeloid, Acute , Neoplasms, Second Primary , Orbital Neoplasms , Sarcoma, Myeloid , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/mortality , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Neoplasms, Second Primary/classification , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/mortality , Orbital Neoplasms/classification , Orbital Neoplasms/drug therapy , Orbital Neoplasms/mortality , Sarcoma, Myeloid/classification , Sarcoma, Myeloid/drug therapy , Sarcoma, Myeloid/mortality , Survival Rate
18.
Leuk Res ; 36(1): 29-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21974856

ABSTRACT

We speculated that some individuals with de novo acute myelogenous leukemia (AML) may have undiagnosed Fanconi Anemia (FA). Data from patients enrolled on AML protocol CCG-2961, published FA cohort studies, SEER, and Bayes rule were used to estimate the probability of FA among all newly diagnosed AML cases, and among those who had no or delayed recovery of the absolute neutrophil count following initial chemotherapy. We determined that the probability of undiagnosed FA in patients in a treatment trial for newly diagnosed patients was around 0.18%, and around 0.83% in the subset who had poor marrow recovery. We suggest that FA or other inherited bone marrow failure syndromes be considered prior to treatment, or certainly among those with poor recovery.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fanconi Anemia/complications , Fanconi Anemia/epidemiology , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/epidemiology , Cohort Studies , Delayed Diagnosis , Fanconi Anemia/diagnosis , Humans , Leukemia, Myeloid, Acute/rehabilitation , Prevalence , Recovery of Function , Time Factors
19.
Pediatr Blood Cancer ; 57(3): 398-405, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21681921

ABSTRACT

PURPOSE: To assess associations of soluble IL-2 receptor alpha (sIL-2rα) concentration with outcomes in pediatric acute myeloid leukemia (AML) in a phase 3 trial of IL-2 therapy. PROCEDURES: We randomized 289 children with AML in first remission after intensive chemotherapy to receive IL-2 infused on days 0-3 and 8-17 (IL-2 group) or no further therapy (AML control group). We measured sequential serum sIL-2rα concentrations in both groups before, during and after therapy in both groups and in reference controls without AML. RESULTS: Before treatment, mean sIL-2rα concentrations were similar in the IL-2 group and AML controls, but significantly higher than in reference controls. Both AML groups experienced reduction in sIL-2rα concentration after chemotherapy. Thereafter in the IL-2 group, mean sIL-2rα concentration increased from 2,669 pg/ml before IL-2 to 15,534 pg/ml on day 4 (P < 0.001) and 10,585 pg/ml on day 18 (P < 0.001). In the control group sIL-2rα concentration did not change after 28 days of follow-up. Five-year disease-free survival (DFS) was 51% in the IL-2 group and 58% in the controls (P = 0.489) and overall survival was 70% and 73%, respectively (P = 0.727). CONCLUSION: SIL-2rα concentration was elevated in AML at diagnosis and tended to normalize after chemotherapy. IL-2 infusion significantly increased sIL-2rα concentration, but did not improve DFS or survival in pediatric AML. Furthermore, sIL-2rα concentration was not predictive of outcome before, during or after treatment for AML.


Subject(s)
Interleukin-2 Receptor alpha Subunit/blood , Interleukin-2/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Monitoring , Female , Humans , Infant , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute/mortality , Male , Prognosis , Solubility , Survival Analysis
20.
Blood ; 117(26): 7007-13, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21518931

ABSTRACT

IL-2 is a natural, T cell-derived cytokine that stimulates the cytotoxic functions of T and natural killer cells. IL-2 monotherapy has been evaluated in several randomized clinical trials (RCTs) for remission maintenance in patients with acute myeloid leukemia (AML) in first complete remission (CR1), and none demonstrated a significant benefit of IL-2 monotherapy. The objective of this meta-analysis was to reliably determine IL-2 efficacy by combining all available individual patient data (IPD) from 5 RCTs (N = 905) and summary data from a sixth RCT (N = 550). Hazard ratios (HRs) were estimated using Cox regression models stratified by trial, with HR < 1 indicating treatment benefit. Combined IPD showed no benefit of IL-2 over no treatment in terms of leukemia-free survival (HR = 0.97; P = .74) or overall survival (HR = 1.08; P = .39). Analyses including the sixth RCT yielded qualitatively identical results (leukemia-free survival HR = 0.96, P = .52; overall survival HR = 1.06; P = .46). No significant heterogeneity was found between the trials. Prespecified subset analyses showed no interaction between the lack of IL-2 effect and any factor, including age, sex, baseline performance status, karyotype, AML subtype, and time from achievement of CR1 to initiation of maintenance therapy. We conclude that IL-2 alone is not an effective remission maintenance therapy for AML patients in CR1.


Subject(s)
Immunotherapy , Interleukin-2/therapeutic use , Leukemia, Myeloid, Acute/prevention & control , Adult , Child , Female , Humans , Leukemia, Myeloid, Acute/therapy , Male , Randomized Controlled Trials as Topic , Recombinant Proteins/therapeutic use , Secondary Prevention , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...