Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Allergy Clin Immunol ; 153(1): 287-296, 2024 01.
Article in English | MEDLINE | ID: mdl-37793572

ABSTRACT

BACKGROUND: The Primary Immune Deficiency Treatment Consortium (PIDTC) enrolled children in the United States and Canada onto a retrospective multicenter natural history study of hematopoietic cell transplantation (HCT). OBJECTIVE: We investigated outcomes of HCT for severe combined immunodeficiency (SCID). METHODS: We evaluated the chronic and late effects (CLE) after HCT for SCID in 399 patients transplanted from 1982 to 2012 at 32 PIDTC centers. Eligibility criteria included survival to at least 2 years after HCT without need for subsequent cellular therapy. CLE were defined as either conditions present at any time before 2 years from HCT that remained unresolved (chronic), or new conditions that developed beyond 2 years after HCT (late). RESULTS: The cumulative incidence of CLE was 25% in those alive at 2 years, increasing to 41% at 15 years after HCT. CLE were most prevalent in the neurologic (9%), neurodevelopmental (8%), and dental (8%) categories. Chemotherapy-based conditioning was associated with decreased-height z score at 2 to 5 years after HCT (P < .001), and with endocrine (P < .001) and dental (P = .05) CLE. CD4 count of ≤500 cells/µL and/or continued need for immunoglobulin replacement therapy >2 years after transplantation were associated with lower-height z scores. Continued survival from 2 to 15 years after HCT was 90%. The presence of any CLE was associated with increased risk of late death (hazard ratio, 7.21; 95% confidence interval, 2.71-19.18; P < .001). CONCLUSION: Late morbidity after HCT for SCID was substantial, with an adverse impact on overall survival. This study provides evidence for development of survivorship guidelines based on disease characteristics and treatment exposure for patients after HCT for SCID.


Subject(s)
Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Child , Humans , Severe Combined Immunodeficiency/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Incidence , Canada/epidemiology , Retrospective Studies , Transplantation Conditioning
2.
Eur J Haematol ; 108(4): 278-287, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34897809

ABSTRACT

Classification of inherited bone marrow failure syndromes (IBMFSs) according to clinical and genetic diagnoses enables proper adjustment of treatment. Unfortunately, 30% of patients enrolled in the Canadian Inherited Marrow Failure Registry (CIMFR) with features suggesting hereditability could not be classified with a specific syndromic diagnosis. We analyzed the outcome of hematopoietic stem cell transplantation (HSCT) in unclassified IBMFSs (uIBMFSs) and the factors associated with outcome. Twenty-two patients with uIBMFSs and 70 patients with classified IBMFSs underwent HSCT. Five-year overall survival of uIBMFS patients after HSCT was inferior to that of patients with classified IBMFSs (56% vs 76.5%). The outcome of patients with uIBMFS who received cord blood was significantly lower than that of patients who received other stem cell sources (14.8% vs 90.9%). Engraftment failure was higher among patients with uIBMFS who received cord blood than those who received bone marrow. None of the following factors were significantly associated with poor survival: transfusion load, transplant indication, the intensity of conditioning regimen, human leukocyte antigen-identical sibling/alternative donor. We suggest that identifying the genetic diagnosis is essential to modulate the transplant procedure including conditioning agents and stem cell sources for better outcome and the standard cord blood transplantation (CBT) should be avoided in uIBMFS.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Bone Marrow Transplantation , Canada/epidemiology , Congenital Bone Marrow Failure Syndromes , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , HLA Antigens , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Retrospective Studies , Transplantation Conditioning/methods
3.
Transplant Cell Ther ; 28(3): 174.e1-174.e5, 2022 03.
Article in English | MEDLINE | ID: mdl-34958973

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) used after hematopoietic stem cell transplantation (HSCT) can enhance neutrophil recovery in patients rendered neutropenic by the preparative regimen. G-CSF is contraindicated in patients with sickle cell disease (SCD), because life-threatening complications can ensue in the presence of sickle vasculopathy. The safety profile of G-CSF after HSCT for SCD has not been described, however. We report clinical outcomes in the first 100 days post-HSCT in 62 patients supported with G-CSF until neutrophil recovery on a clinical trial of reduced- intensity conditioning HSCT for SCD. The patients received G-CSF for a median of 9 days (range, 5 to 33 days) post-transplantation from the best available stem cell source. Preparation for transplantation included a target hemoglobin S level of ≤45%. Neutrophil engraftment (absolute neutrophil count >0.5 × 103/mL) was achieved at a median of 13 days (range, 10 to 34 days), and platelet engraftment (>50 × 103/mL) was achieved at a median of 19 days (range, 12 to 71 days). The median duration of inpatient hospitalization following stem cell infusion (day 0) was 21.5 days (range, 11 to 33 days). No patient developed SCD-related complications following G-CSF use. The most common organ toxicities encountered between G-CSF initiation (on day +7) and day +100 were anorexia (n = 14), hypertension (n = 11), and electrolyte imbalance requiring correction (n = 9). Central nervous system-related events were noted in 5 patients, all of whom had preexisting cerebral vasculopathy/moyamoya disease, attributed to reversible posterior leukoencephalopathy syndrome in the presence of calcineurin inhibitor therapy and hypertension. We conclude that G-CSF does not adversely impact SCD HSCT recipients and can be safely used post-transplantation to enhance neutrophil recovery.


Subject(s)
Anemia, Sickle Cell , Granulocyte Colony-Stimulating Factor , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Granulocyte Colony-Stimulating Factor/adverse effects , Hematopoietic Stem Cell Transplantation , Humans , Hypertension/epidemiology , Transplantation, Homologous
5.
Front Immunol ; 11: 571884, 2020.
Article in English | MEDLINE | ID: mdl-33193355

ABSTRACT

It is established that pediatric hematopoietic stem cell transplant (HSCT) recipients have a lower rate of chronic graft-versus-host disease (cGvHD) compared to adults. Our group has previously published immune profiles changes associated with cGvHD of clinically well-defined adult and pediatric HSCT cohorts. Since all analyses were performed by the same research group and analyzed using identical methodology, we first compared our previous immune profile analyses between adults and children. We then performed additional analyses comparing the T cell populations across age groups, and a sub-analysis of the impact of the estimated pubertal status at time of HSCT in our pediatric cohort. In all analyses, we corrected for clinical covariates including total body irradiation and time of onset of cGvHD. Three consistent findings were seen in both children and adults, including elevations of ST2 and naive helper T (Th) cells and depression of NKreg cells. However, significant differences exist between children and adults in certain cytokines, B cell, and Treg populations. In children, we saw a broad suppression of newly formed B (NF-B) cells, whereas adults exhibited an increase in T1-CD21lo B cells and a decrease in T1-CD24hiCD38hi B cells. Prepubertal children had elevations of aminopeptidase N (sCD13) and ICAM-1. Treg abnormalities in children appeared to be primarily in memory Treg cells, whereas in adults the abnormalities were in naïve Treg cells. In adults, the loss of PD1 expression in naïve Treg and naïve Th cells was associated with cGvHD. We discuss the possible mechanisms for these age-related differences, and how they might theoretically impact on different therapeutic approaches to cGvHD between children and adults.


Subject(s)
Age Factors , B-Lymphocytes/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Child , Chronic Disease , Female , Graft vs Host Disease/epidemiology , Humans , Immunophenotyping , Interleukin-1 Receptor-Like 1 Protein/metabolism , Lymphocyte Activation , Male , Middle Aged , Up-Regulation
6.
Pediatr Blood Cancer ; 67(7): e28263, 2020 07.
Article in English | MEDLINE | ID: mdl-32323913

ABSTRACT

INTRODUCTION: Indications for hematopoietic stem cell transplantation (HSCT) in pediatric acute myeloid leukemia (AML) are primarily dependent on risk stratification at diagnosis and relapse status. We sought to determine whether access to HSCT is influenced by regional and socioeconomic factors. METHODS: Children with newly diagnosed AML aged < 15 years between 2001 and 2015 were identified using the Cancer in Young People in Canada national population-based registry. Factors potentially associated with the receipt of HSCT were studied using univariate and multivariable logistic regression models. RESULTS: Overall, 568 children with newly diagnosed AML were included and 262 (46%) received HSCT. A greater proportion of patients, 103/157 (65.6%), underwent HSCT after first or subsequent relapse compared to 159/411 (38.7%) patients who underwent transplant before relapse. Among patients for whom HSCT would be considered before relapse, factors associated with higher odds of HSCT in a multivariable analysis were: poor versus good-risk cytogenetics (Odds ratio [OR]: 30.0, 95% confidence interval [CI]: 7.7-117.0), diagnosis during 2012-2015 versus 2001-2006 (OR: 3.2, 95% CI: 1.6-6.3), diagnosis in eastern Canada versus central Canada (OR: 3.7, 95% CI: 1.9-7.3), and age 10-14 years versus age < 1 year (OR: 5.4, 95% CI: 2.3-12.8). Among patients for whom HSCT would be considered after first relapse, higher odds of HSCT was associated with diagnosis at a HSCT center (OR: 2.1, 95% CI: 1.1-4.1). CONCLUSION: Patients diagnosed at a HSCT performing center and patients from eastern Canada had higher odds of receiving HSCT. This may suggest preferential access to HSCT for certain patients.


Subject(s)
Graft vs Host Disease/epidemiology , Health Services Accessibility/statistics & numerical data , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/therapy , Neoplasm Recurrence, Local/epidemiology , Adolescent , Canada/epidemiology , Child , Child, Preschool , Female , Follow-Up Studies , Graft vs Host Disease/diagnosis , Humans , Incidence , Infant , Leukemia, Myeloid, Acute/pathology , Male , Neoplasm Recurrence, Local/diagnosis , Prognosis , Retrospective Studies , Transplantation Conditioning , Transplantation, Homologous
7.
NPJ Genom Med ; 4: 30, 2019.
Article in English | MEDLINE | ID: mdl-31839986

ABSTRACT

Inherited bone marrow failure syndromes (IBMFSs) are genetically heterogeneous disorders with cytopenia. Many IBMFSs also feature physical malformations and an increased risk of cancer. Point mutations can be identified in about half of patients. Copy number variation (CNVs) have been reported; however, the frequency and spectrum of CNVs are unknown. Unfortunately, current genome-wide methods have major limitations since they may miss small CNVs or may have low sensitivity due to low read depths. Herein, we aimed to determine whether reanalysis of NGS panel data by normalized coverage value could identify CNVs and characterize them. To address this aim, DNA from IBMFS patients was analyzed by a NGS panel assay of known IBMFS genes. After analysis for point mutations, heterozygous and homozygous CNVs were searched by normalized read coverage ratios and specific thresholds. Of the 258 tested patients, 91 were found to have pathogenic point variants. NGS sample data from 165 patients without pathogenic point mutations were re-analyzed for CNVs; 10 patients were found to have deletions. Diamond Blackfan anemia genes most commonly exhibited heterozygous deletions, and included RPS19, RPL11, and RPL5. A diagnosis of GATA2-related disorder was made in a patient with myelodysplastic syndrome who was found to have a heterozygous GATA2 deletion. Importantly, homozygous FANCA deletion were detected in a patient who could not be previously assigned a specific syndromic diagnosis. Lastly, we identified compound heterozygousity for deletions and pathogenic point variants in RBM8A and PARN genes. All deletions were validated by orthogonal methods. We conclude that careful analysis of normalized coverage values can detect CNVs in NGS panels and should be considered as a standard practice prior to do further investigations.

8.
Biol Blood Marrow Transplant ; 25(6): 1172-1178, 2019 06.
Article in English | MEDLINE | ID: mdl-30769192

ABSTRACT

Access to hematopoietic stem cell transplantation (HSCT) in pediatric acute lymphoblastic leukemia (ALL) primarily depends on disease-related factors but may be influenced by social and economic determinants. We included all children aged < 15 years with newly diagnosed ALL in Canada between 2001 and 2018 using the Cancer in Young People in Canada national registry. We examined factors potentially associated with the likelihood of receiving HSCT using univariate and multivariable logistic regression models. A total of 3992 patients with newly diagnosed ALL were included. Three hundred twenty-five (8.1%) received an HSCT and formed the transplant cohort. In multivariable analysis factors independently associated with an increased odds of receiving HSCT were male sex (odds ratio [OR], 1.42; 95% confidence interval [CI], 1.05 to 1.93), initial WBC ≥ 50,000 × 109/L (OR, 1.58; 95% CI, 1.09 to 2.28), mixed phenotype acute leukemia relative to B-precursor ALL (OR, 34.32; 95% CI, 16.64 to 70.79), T cell relative to B-precursor ALL (OR, 1.77; 95% CI, 1.07 to 2.91), unfavorable relative to standard cytogenetics (OR, 3.96; 95% CI, 2.56 to 6.12), and relapse before HSCT (OR, 32.77; 95%, 23.89 to 44.96). No association was found between race, neighborhood income quintile or region at diagnosis, and receipt of HSCT. Diagnosis at an HSCT treating center (OR, 1.51; 95% CI, 1.09 to 2.09) and residential distance from the ALL treating center (OR, 1.84 for ≥300 km compared with <100 km; 95% CI, 1.17 to 2.91) were associated with higher odds of receiving HSCT. In a publically funded healthcare system, children with ALL had equitable access to HSCT, which was largely governed by biologic disease-related factors. Patients diagnosed at an HSCT performing center and patients who live farthest away from their treatment center had higher odds of receiving HSCT, although the effect was small, possibly suggesting preferential referral to HSCT for some patients.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transplantation Conditioning/methods , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male
10.
Respir Care ; 64(2): 209-216, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30584066

ABSTRACT

Pediatric ARDS continues to be a management challenge in the ICU with prolonged hospitalizations and high mortality. Thromboembolic pulmonary embolism and in situ pulmonary artery thrombosis might represent underappreciated thrombotic processes for a subset of these patients. Although well described in the adult literature, descriptions of pulmonary thromboses with pediatric ARDS are limited to case reports. However, many risk factors for pulmonary thromboses are present in children with ARDS (eg, coagulopathy, endothelial injury, central venous catheters, concomitant inflammatory diseases), suggesting a much higher incidence is plausible. Based on an interpretation of animal, pediatric, and adult data, we propose a diagnostic algorithm to facilitate a timely and accurate diagnosis. Observing an alveolar dead space fraction ≥ 0.25, or either a 50% increase in physiologic dead space/tidal volume or a central venous saturation ≤ 60% over 24 h, triggers the algorithm. Together with targeted heparin treatment and right ventricular afterload reduction, clinical outcomes might improve if this particular patient subgroup can be identified early. While anticoagulation is recommended in adults with confirmed pulmonary embolism and low early mortality risk, data for children are limited.


Subject(s)
Algorithms , Pulmonary Artery , Pulmonary Embolism/diagnosis , Respiratory Distress Syndrome, Newborn/complications , Thrombosis/diagnosis , Adult , Animals , Blood Gas Analysis , Child , Female , Humans , Male , Pulmonary Embolism/etiology , Respiratory Dead Space , Risk Assessment/methods , Risk Factors , Thrombosis/etiology , Tidal Volume
11.
Blood ; 130(25): 2718-2727, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29021228

ABSTRACT

The Primary Immune Deficiency Treatment Consortium (PIDTC) is enrolling children with severe combined immunodeficiency (SCID) to a prospective natural history study. We analyzed patients treated with allogeneic hematopoietic cell transplantation (HCT) from 2010 to 2014, including 68 patients with typical SCID and 32 with leaky SCID, Omenn syndrome, or reticular dysgenesis. Most (59%) patients were diagnosed by newborn screening or family history. The 2-year overall survival was 90%, but was 95% for those who were infection-free at HCT vs 81% for those with active infection (P = .009). Other factors, including the diagnosis of typical vs leaky SCID/Omenn syndrome, diagnosis via family history or newborn screening, use of preparative chemotherapy, or the type of donor used, did not impact survival. Although 1-year post-HCT median CD4 counts and freedom from IV immunoglobulin were improved after the use of preparative chemotherapy, other immunologic reconstitution parameters were not affected, and the potential for late sequelae in extremely young infants requires additional evaluation. After a T-cell-replete graft, landmark analysis at day +100 post-HCT revealed that CD3 < 300 cells/µL, CD8 < 50 cells/µL, CD45RA < 10%, or a restricted Vß T-cell receptor repertoire (<13 of 24 families) were associated with the need for a second HCT or death. In the modern era, active infection continues to pose the greatest threat to survival for SCID patients. Although newborn screening has been effective in diagnosing SCID patients early in life, there is an urgent need to identify validated approaches through prospective trials to ensure that patients proceed to HCT infection free. The trial was registered at www.clinicaltrials.gov as #NCT01186913.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immune Reconstitution , Severe Combined Immunodeficiency/therapy , Child, Preschool , Female , Genotype , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immune Reconstitution/genetics , Infant , Infant, Newborn , Infections/etiology , Male , Neonatal Screening , Prospective Studies , Risk Factors , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/mortality , Survival Analysis , Tissue Donors
12.
NPJ Genom Med ; 22017 May 10.
Article in English | MEDLINE | ID: mdl-28690869

ABSTRACT

Inherited bone marrow failure syndromes (IBMFSs) comprise a genetically heterogeneous group of diseases with hematopoietic failure and a wide array of physical malformations. Copy number variants (CNVs) were reported in some IBMFSs. It is unclear what impact CNVs play in patients evaluated for a suspected diagnosis of IBMFS. Clinical and genetic data of 323 patients from the Canadian Inherited Marrow Failure Registry from 2001 to 2014, who had a documented genetic work-up, were analyzed. Cases with pathogenic CNVs (at least 1 kilobasepairs) were compared to cases with other mutations. Genotype-phenotype correlations were performed to assess the impact of CNVs. Pathogenic nucleotide-level mutations were found in 157 of 303 tested patients (51.8%). Genome-wide CNV analysis by single nucleotide polymorphism arrays or comparative genomic hybridization arrays revealed pathogenic CNVs in 11 of 67 patients tested (16.4%). In four of these patients, identification of CNV was crucial for establishing the correct diagnosis as their clinical presentation was ambiguous. Eight additional patients were identified to harbor pathogenic CNVs by other methods. Of the 19 patients with pathogenic CNVs, four had compound-heterozygosity of a CNV with a nucleotide-level mutation. Pathogenic CNVs were associated with more extensive non-hematological organ system involvement (p=0.0006), developmental delay (p=0.006) and short stature (p=0.04) compared to nucleotide-level mutations. In conclusion, a significant proportion of patients with IBMFSs harbor pathogenic CNVs which were associated with a more extensive non-hematological phenotype in this cohort. Patients with a phenotype suggestive of IBMFSs but without identification of pathogenic nucleotide-level mutations should undergo specific testing for CNVs.

13.
Can J Kidney Health Dis ; 4: 2054358117690338, 2017.
Article in English | MEDLINE | ID: mdl-28270931

ABSTRACT

BACKGROUND: Childhood cancer survivors experience adverse drug events leading to lifelong health issues. The Applying Biomarkers to Minimize Long-Term Effects of Childhood/Adolescent Cancer Treatment (ABLE) team was established to validate and apply biomarkers of cancer treatment effects, with a goal of identifying children at high risk of developing cancer treatment complications associated with thrombosis, graft-versus-host disease, hearing loss, and kidney damage. Cisplatin is a chemotherapy well known to cause acute and chronic nephrotoxicity. Data on biomarkers of acute kidney injury (AKI) and late renal outcomes in children treated with cisplatin are limited. OBJECTIVE: To describe the design and methods of the pan-Canadian ABLE Nephrotoxicity study, which aims to evaluate urine biomarkers (neutrophil gelatinase-associated lipocalin [NGAL] and kidney injury molecule-1 [KIM-1]) for AKI diagnosis, and determine whether they predict risk of long-term renal outcomes (chronic kidney disease [CKD], hypertension). DESIGN: This is a 3-year observational prospective cohort study. SETTING: The study includes 12 Canadian pediatric oncology centers. PATIENTS: The target recruitment goal is 150 patients aged less than 18 years receiving cisplatin. Exclusion criteria: Patients with an estimated glomerular filtration rate (eGFR) <30 mL/min/1.73 m2 or a pre-existing renal transplantation at baseline. MEASUREMENTS: Serum creatinine (SCr), urine NGAL, and KIM-1 are measured during cisplatin infusion episodes (pre-infusion, immediate post-infusion, discharge sampling). At follow-up visits, eGFR, microalbuminuria, and blood pressure are measured and outcomes are collected. METHODS: Outcomes: AKI is defined as per SCr criteria of the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines. CKD is defined as eGFR <90 mL/min/1.73m2 or albumin-to-creatinine ratio≥3mg/mmol. Hypertension is defined as per guidelines. Procedure: Patients are recruited before their first or second cisplatin cycle. Participants are evaluated during 2 cisplatin infusion episodes (AKI biomarker validation) and at 3, 12, and 36 months post-cisplatin treatment (late outcomes). LIMITATIONS: The study has a relatively moderate sample size and short follow-up duration. There is potential for variability in data collection since multiple sites are involved. CONCLUSIONS: ABLE will provide a national platform to study biomarkers of late cancer treatment complications. The Nephrotoxicity study is a novel study of AKI biomarkers in children treated with cisplatin that will greatly inform on late cisplatin renal outcomes and follow-up needs.


MISE EN CONTEXTE: Les survivants d'un cancer infantile éprouvent des effets indésirables dus à leurs traitements, ce qui leurs engendrent des problèmes de santé à vie. L'équipe The Applying Biomarkers to Minimize Long-Term Effects of Childhood/Adolescent Cancer Treatment (ABLE) vise à valider des biomarqueurs d'effets indésirables causés par les traitements contre le cancer et identifier les enfants à risque de développer des complications associées aux problèmes de coagulation, à la maladie du greffon contre l'hôte, la perte auditive et l'insuffisance rénale. La chimiothérapie cisplatine cause des dommages aux reins à court et à long terme. Relativement peu de données existent sur les biomarqueurs d'insuffisance rénale aigüe (IRA) et sur les problèmes rénaux à long terme chez les enfants traités avec le cisplatine. OBJECTIFS: Décrire les méthodologies de l'étude pancanadienne néphrotoxique ABLE qui vise à évaluer si les biomarqueurs urinaires (neutrophil gelatinase-associated lipocalin [NGAL] et kidney injury molecule-1[KIM-1]) peuvent diagnostiquer l'IRA, et s'ils peuvent prédire le risque de développer l'insuffisance rénale chronique (IRC) et l'hypertension artérielle à long terme. CADRE ET TYPE D'ÉTUDE: Étude prospective observationnelle de 3 ans dans 12 centres d'oncologie pédiatrique canadiens. PARTICIPANTS: cible de 150 patients âgés <18 ans recevant du cisplatine. Critères d'exclusion: Débit de filtration glomérulaire estimé (DFGe)<30 mL/min/1.73m2 ou avoir reçu une transplantation rénale. MESURES: Créatinine sérique, NGAL/KIM-1 sont mesurés pendant les infusions de cisplatine (échantillonnage avant l'infusion, après, et avant la sortie de l'hôpital). Visites de suivi: DFGe, microalbuminurie et tension artérielle sont mesurés; les résultats sont recueillis. MÉTHODOLOGIE: Critères d'évaluation: L'IRA est définie selon les critères de créatinine sérique de la classification Kidney Disease: Improving Global Outcomes (KDIGO). L'IRC est définie comme ayant un DFGe<90 mL/min/1.73m2 ou un ratio d'albumine/créatinine ≥3mg/mmol. L'hypertension est définie selon les lignes directrices. Procédure: Le recrutement: à lieu au premier ou deuxième cycle de cisplatine. Les patients sont évalués pendant deux infusions de cisplatine (validation des biomarqueurs d'IRA) et 3, 12 et 36 mois après le cisplatine (évaluation des problèmes rénaux à long terme). LIMITES DE L'ÉTUDE: La taille de l'échantillon est relativement modérée et la durée du suivi est moyennement courte. Il pourrait potentiellement avoir de la variabilité dans la collecte de données car plusieurs sites d'études sont impliqués. CONCLUSIONS: ABLE génèrera une plateforme nationale pour étudier les biomarqueurs de complications à long terme des traitements contre le cancer. L'étude néphrotoxique ABLE est une étude novatrice des biomarqueurs de l'IRA chez les enfants traités avec le cisplatine qui contribuera grandement à identifier les problèmes rénaux à long terme causés par le cisplatine et la nécessité de suivis cliniques.

14.
Lancet Haematol ; 3(11): e526-e536, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27746112

ABSTRACT

BACKGROUND: Intravenous busulfan combined with therapeutic drug monitoring to guide dosing improves outcomes after allogeneic haemopoietic cell transplantation (HCT). The best method to estimate busulfan exposure and optimum exposure in children or young adults remains unclear. We therefore assessed three approaches to estimate intravenous busulfan exposure (expressed as cumulative area under the curve [AUC]) and associated busulfan AUC with clinical outcomes in children or young adults undergoing allogeneic HCT. METHODS: In this retrospective analysis, patients from 15 centres in the Netherlands, USA, Canada, Switzerland, UK, Italy, Germany, and Australia who received a busulfan-based conditioning regimen between March 18, 2001, and Feb 12, 2015, were included. Cumulative AUC was calculated by numerical integration using non-linear mixed effect modelling (AUCNONMEM), non-compartmental analysis (AUC from 0 to infinity [AUC0-∞] and to the next dose [AUC0-τ]), and by individual centres using various approaches (AUCcentre). The main outcome of interest was event-free survival. Other outcomes of interest were graft failure or relapse, or both; transplantation-related mortality; acute toxicity (veno-occlusive disease or acute graft versus-host disease [GvHD]); chronic GvHD; overall survival; and chronic-GvHD-free event-free survival. We used propensity-score-adjusted Cox proportional hazard models, Weibull models, and Fine-Gray competing risk regressions for statistical analyses. FINDINGS: 790 patients were enrolled, 674 of whom were included: 274 (41%) with malignant and 400 (59%) with non-malignant disease. Median age was 4·5 years (IQR 1·4-10·7). The median busulfan AUCNONMEM was 74·4 mg × h/L (95% CI 31·1-104·6), which correlated with the standardised method AUC0-∞ (r2=0·74), but the latter correlated poorly with AUCcentre (r2=0·35). Estimated 2-year event-free survival was 69·7% (95% CI 66·2-73·0). Event-free survival at 2 years was 77·0% (95% CI 72·1-82·9) in the 257 patients with an optimum intravenous busulfan AUC of 78-101 mg × h/L compared with 66·1% (60·9-71·4) in the 235 patients at the low historical target of 58-86 mg × h/L and 49·5% (29·2-66·0) in the 44 patients with a high (>101 mg × h/L) busulfan AUC (p=0·011). Compared with the low AUC group, graft failure or relapse occurred less frequently in the optimum AUC group (hazard ratio [HR] 0·57, 95% CI 0·39-0·84; p=0·0041). Acute toxicity (HR 1·69, 1·12-2·57; p=0·013) and transplantation-related mortality (2·99, 1·82-4·92; p<0·0001) were significantly higher in the high AUC group (>101 mg × h/L) than in the low AUC group (<78 mg × h/L), independent of indication; no difference was noted between AUC groups for chronic GvHD (<78 mg × h/L vs ≥78 mg × h/L, HR 1·30, 95% CI 0·73-2·33; p=0·37). INTERPRETATION: Improved clinical outcomes are likely to be achieved by targeting the busulfan AUC to 78-101 mg × h/L using a new validated pharmacokinetic model for all indications. FUNDING: Research Allocation Program and the UCSF Helen Friller Family Comprehensive Cancer Center and the Mt Zion Health Fund of the University of California, San Francisco.


Subject(s)
Area Under Curve , Busulfan/administration & dosage , Busulfan/pharmacokinetics , Dose-Response Relationship, Drug , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Transplantation Conditioning/methods , Transplantation, Homologous/adverse effects , Transplantation, Homologous/mortality , Adolescent , Adult , Busulfan/therapeutic use , Busulfan/toxicity , Child , Child, Preschool , Cohort Studies , Disease-Free Survival , Female , Graft Rejection/epidemiology , Graft Survival/drug effects , Graft vs Host Disease/epidemiology , Humans , Infant , Male , Recurrence , Retrospective Studies , Transplantation Conditioning/adverse effects , Treatment Outcome
15.
Am J Hematol ; 90(12): 1093-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26348869

ABSTRACT

Fifty-two children with symptomatic sickle cell disease sickle cell disease (SCD) (N = 43) or transfusion-dependent thalassemia (N = 9) received matched sibling donor marrow (46), marrow and cord product (5), or cord blood (1) allografts following reduced intensity conditioning (RIC) with alemtuzumab, fludarabine, and melphalan between March 2003 and May 2014*. The Kaplan-Meier probabilities of overall and event-free survival at a median of 3.42 (range, 0.75-11.83) years were 94.2% and 92.3% for the group, 93% and 90.7% for SCD, and 100% and 100% for thalassemia, respectively. Treatment-related mortality (all related to graft versus host disease, GVHD) was noted in three (5.7%) recipients, all 17-18 years of age. Acute and chronic GVHD was noted in 23% and 13%, respectively, with 81% of recipients off immunosuppression by 1 year. Graft rejection was limited to the single umbilical cord blood recipient who had prompt autologous hematopoietic recovery. Fourteen (27%) had mixed chimerism at 1 year and beyond; all had discontinued immunosuppression between 4 and 12 months from transplant with no subsequent consequence on GVHD or rejection. Infectious complications included predominantly bacteremia (48% were staphylococcus) and CMV reactivation (43%) necessitating preemptive therapy. Lymphocyte recovery beyond 6 months was associated with subsidence of infectious complications. All patients who engrafted were transfusion independent; no strokes or pulmonary complications of SCD were noted, and pain symptoms subsided within 6 months posttransplant. These findings support using RIC for patients with hemoglobinopathy undergoing matched sibling marrow transplantation (*www.Clinical Trials.gov: NCT00920972, NCT01050855, NCT02435901).


Subject(s)
Bone Marrow Transplantation/methods , Hemoglobinopathies/surgery , Hemoglobinopathies/therapy , Transplantation Conditioning/methods , Disease-Free Survival , Female , Humans , Male , Siblings , Tissue Donors
16.
Biol Blood Marrow Transplant ; 21(7): 1205-14, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25963921

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are regulatory cell populations that have the ability to suppress effector T cell responses and promote the development of regulatory T cells (Tregs). They are a heterogeneous population of immature myeloid progenitors that include monocytic and granulocytic subsets. We postulated that given the rapid expansion of myeloid cells post-transplant, these members of the innate immune system may be important contributors to the early immune environment post-transplant. To evaluate the kinetics of recovery and function of MDSCs after allogeneic hematopoietic stem cell transplant (HSCT), 26 patients undergoing allogeneic HSCT were studied at 6 time points in the first 3 months after HSCT. Both MDSC subsets recovered between 2 and 4 weeks, well before the recovery of T and B lymphocytes. MDSC subset recovery positively correlated with T, B, and/or double-negative T cell numbers after HSCT. MDSCs isolated from patients post-transplant were functional in that they suppressed third-party CD4(+) T cell proliferation and Th1 differentiation and promoted Treg development. In conclusion, functional MDSC are present early after HSCT and likely contribute to the regulatory cell population post-transplant.


Subject(s)
Cell Lineage/immunology , Granulocytes/immunology , Hematologic Neoplasms/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Monocytes/immunology , Transplantation Conditioning , Adolescent , Adult , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Count , Cell Differentiation , Cell Lineage/drug effects , Cell Proliferation , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Granulocytes/drug effects , Granulocytes/pathology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Lymphocyte Activation , Male , Middle Aged , Monocytes/drug effects , Monocytes/pathology , Myeloablative Agonists/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Time Factors , Transplantation, Homologous
17.
Clin Case Rep ; 3(2): 121-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25767712

ABSTRACT

Diagnostic criteria for hemophagocytic lymphohistiocytosis should be reviewed early in critically ill patients with toxic epidermal necrolysis, multisystem dysfunction, and a deteriorating clinical trajectory.

18.
Pediatr Blood Cancer ; 61(12): 2164-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24616082

ABSTRACT

BACKGROUND: Health disparities between Canadian First Nation (FN) people and the rest of the national population exist. No studies have specifically documented cancer-related health outcomes in Canadian FN children. The purpose of this study was to describe the incidence of pediatric malignancies in Manitoba FN children, and to compare morbidity patterns and survival between FN and non-FN children with cancer in the Canadian province of Manitoba. PROCEDURE: A retrospective, population-based review of all children (0-14.99 years) diagnosed with malignancy (2001-2008) in Manitoba, Canada was undertaken using the Cancer in Young People in Canada registry. FN children were compared to the non-FN population for markers of morbidity and survival. RESULTS: The average annual age-standardized incidence rate for all childhood cancers in FN children was 132 per 1,000,000 per year. 240 children were included in the morbidity and survival analyses (38 FN; 202 non-FN). No differences were found between FN and non-FN children in time from first presentation of symptoms to consultation with an oncology specialist or diagnosis, or number of hospital admissions / total days of admission for treatment complications. Overall survival was inferior for FN children in univariable analysis (P = 0.048) but not when risk group was included in a multivariable analysis (P = 0.15). No difference in event free survival or cumulative incidence of relapse was identified. CONCLUSION: The estimated incidence of childhood cancers in the Manitoba FN population is similar to provincial incidence rates. No differences in morbidity patterns or survival were found between Manitoba FN and non-FN children with cancer.


Subject(s)
Hospitalization/statistics & numerical data , Morbidity , Neoplasms/epidemiology , Neoplasms/mortality , Adolescent , Canada/epidemiology , Child , Child, Preschool , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Incidence , Infant , Infant, Newborn , Male , Neoplasm Staging , Neoplasms/therapy , Prognosis , Referral and Consultation , Registries , Retrospective Studies , Survival Rate
19.
Pediatr Blood Cancer ; 61(4): 672-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24167059

ABSTRACT

BACKGROUND: Megestrol acetate (MA) is an appetite stimulant with efficacy in promoting weight gain in adults with cancer-associated anorexia-cachexia. Studies documenting MA efficacy in children, however, are limited. We present the first randomized, double-blind, placebo-controlled clinical trial of MA versus placebo in children with cancer and weight loss. METHODS: Subjects <18 years of age with weight loss (minimum 5% from highest previous weight; or %ideal body weight <90%) due to cancer and/or cancer therapy were randomized to either MA (7.5 mg/kg/day) or placebo for a planned study duration of 90 days. Primary outcome was the difference between groups in mean percent weight change from beginning to end of the study period. Secondary outcomes included effects on anthropometrics, body composition, need for tube feeding or parenteral nutrition, and toxicities. RESULTS: Twenty-six patients were randomly assigned (13 MA, 13 placebo). The MA group experienced a mean weight gain of +19.7% compared to a mean weight loss of -1.2% in the placebo group, for a difference of +20.9% (95%CI: +11.3% to +30.5%, P = 0.003) in favor of MA over placebo. MA subjects experienced significant increases in weight for age z-scores, body mass index z-scores, and mid upper arm circumference compared to placebo. DXA scanning suggested disproportionate increases in fat accrual. Adrenal suppression was the main toxicity of MA. CONCLUSION: In children with high-risk malignancies, MA resulted in significant increases in mean percent weight change compared to placebo. Further studies of MA should be pursued to better delineate the effect on nutritional status.


Subject(s)
Appetite Stimulants/therapeutic use , Appetite/drug effects , Megestrol Acetate/therapeutic use , Neoplasms/complications , Nutrition Disorders/drug therapy , Weight Loss/drug effects , Adolescent , Adult , Child , Child, Preschool , Double-Blind Method , Female , Follow-Up Studies , Humans , Infant , Male , Neoplasm Staging , Neoplasms/therapy , Nutrition Disorders/diagnosis , Nutrition Disorders/etiology , Prognosis , Quality of Life , Young Adult
20.
Biol Blood Marrow Transplant ; 19(11): 1608-14, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24029650

ABSTRACT

Little information is currently available regarding the pharmacokinetics (PK) of busulfan in infants and small children to help guide decisions for safe and efficacious drug therapy. The objective of this study was to develop an algorithm for individualized dosing of i.v. busulfan in infants and children weighing ≤12 kg, that would achieve targeted exposure with the first dose of busulfan. Population PK modeling was conducted using intensive time-concentration data collected through the routine therapeutic drug monitoring of busulfan in 149 patients from 8 centers. Busulfan PK was well described by a 1-compartment base model with linear elimination. The important clinical covariates affecting busulfan PK were actual body weight and age. Based on our model, the predicted clearance of busulfan increases approximately 1.7-fold between 6 weeks to 2 years of life. For infants age <5 months, the model-predicted doses (mg/kg) required to achieve a therapeutic concentration at steady state of 600-900 ng/mL (area under the curve range, 900-1350 µM·min) were much lower compared with standard busulfan doses of 1.1 mg/kg. These results could help guide clinicians and inform better dosing decisions for busulfan in young infants and small children undergoing hematopoietic cell transplantation.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Busulfan/pharmacokinetics , Hematopoietic Stem Cell Transplantation/methods , Transplantation, Autologous/methods , Antineoplastic Agents, Alkylating/therapeutic use , Busulfan/therapeutic use , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Retrospective Studies , Transplantation Conditioning
SELECTION OF CITATIONS
SEARCH DETAIL
...