• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 7
  • 6
  • 5
  • 1
  • 1
  • Tagged with
  • 20
  • 14
  • 13
  • 13
  • 12
  • 9
  • 7
  • 6
  • 6
  • 6
  • 6
  • 6
  • 5
  • 5
  • 5
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
11

Studies on mechanisms of busulphan cytotoxicity and pharmacokinetics : with special reference to liposomal busulphan /

Hassan, Zuzana, January 1900 (has links)
Diss. (sammanfattning) Stockholm : Karol. inst., 2001. / Härtill 6 uppsatser.
12

Modelo de personalização de dose de bussulfano intravenoso baseado no genótipo de GSTA1 durante regime de condicionamento do transplante de células-tronco hematopoiéticas em crianças

Nava, Tiago Rodrigues January 2017 (has links)
O bussulfano (Bu) é um agente alquilante usado no condicionamento que precede o transplante de células-tronco hematopoiéticas (TCTH) em crianças. Sua farmacocinética (FC) apresenta uma grande variabilidade interindivíduo, que pode ser parcialmente explicada pelas variantes genéticas de GSTA1, gene da enzima glutationa S-transferase α1, crucial para o metabolismo do Bu. Vários métodos de predição da FC do Bu são usados para calcular sua dose, essencialmente com base na idade e peso do paciente. Até o momento, apenas um modelo adulto incorporou as variantes de GSTA1 no cálculo da sua dose do Bu. No presente trabalho, avaliou-se, inicialmente, o desempenho de métodos atualmente disponíveis em pediatria, em função das variantes genéticas de GSTA1. Foram avaliados os parâmetros de FC da primeira dose de 101 crianças e adolescentes submetidos a TCTH alogênico no CHU Sainte-Justine, Montreal, Canadá, após regime de condicionamento que incluía Bu intravenoso (BuCR, do inglês busulfan-containing regimen). Os haplótipos GSTA1 foram interpretados em pares (diplótipos) e depois classificados em três grupos com base nos seus diferentes potenciais de expressão enzimática. As AUCs (area under the curve) medidas e as AUCs calculadas a partir de doses de Bu preditas por 11 modelos diferentes foram classificadas de acordo com a sua capacidade para atingir a AUC-alvo (900 a 1.500 μM.min). Também foram calculados os erros de previsão do clearance do Bu. Após a primeira dose, as AUCs medidas atingiram a AUC-alvo em 38,7%. Os diplótipos de GSTA1 relacionados ao metabolismo lento (G3) e regimes contendo fludarabina (FluCR, do inglês fludarabine-containing regimen) foram os únicos fatores associados à AUC no alvo (OR 4,7, IC 95%, 1,1 - 19,8, p = 0,04 e OR 9,9, IC 95%, 1,6 - 61,7, p = 0,01, respectivamente). Utilizando os outros métodos para o cálculo da dose, a percentagem de AUC no alvo variou de 16% a 74%. G3 e FluCR foram, em alguns modelos, associados à AUC no alvo ou na faixa tóxica, enquanto que os metabolizadores rápidos (G1) foram por vezes associados a AUCs subterapêuticas. Essas associações foram confirmadas na análise de predição do clearance, em que os diplótipos da GSTA1 e o regime de condicionamento influenciaram significativamente a maioria dos erros de previsão dos métodos testados. Uma vez que GSTA1 mostrou influenciar significativamente os algoritmos disponíveis, pretendeu-se desenvolver um modelo de FC de população que incluísse variantes genéticas de GSTA1 como um fator no cálculo de dose do Bu. Para tanto, foram analisados os dados de concentração-tempo de 112 crianças e adolescentes que receberam um BuCR mieloablativo antes de 115 TCTH (autólogos e alogênicos), realizados também no CHU Sainte-Justine. Para a construção do modelo de FC de população, utilizou-se uma análise mista não linear. Sexo, doença de base (maligna vs. não maligna), idade pós-menstrual (PMA) ou idade cronológica, regime de condicionamento e diplótipos de GSTA1 foram avaliados como fatores potenciais. Um modelo de um compartimento com eliminação de primeira ordem foi o que melhor descreveu os dados disponíveis. Um fator de maturação do metabolismo de Bu (Fmat) e o peso elevado a exponencial alométrico teórico foram incluídos no modelo de base. A análise dos fatores revelou PMA (ΔOFV = -26,7, p = 2,3x10-7) e grupos de diplótipos de GSTA1 (ΔOFV = -11,7, p = 0,003) como fatores significativamente associados, respectivamente, ao volume e ao CL do Bu. Os CL dos metabolizadores rápidos (G1) foram preditos como sendo 7% mais elevados que os definidos como metabolizadores normais (G2), enquanto que os metabolizadores lentos (G3) foram descritos com CL 12% menor que os G2. Em conclusão, após se evidenciar que os métodos disponíveis para o cálculo de dose do Bu não são adequados para todos os grupos de diplótipos de GSTA1, propôs-se o primeiro algoritmo de cálculo de dose de Bu em pediatria baseado em farmacogenética. Seu uso pode contribuir para uma melhor previsibilidade da FC do Bu e, desta forma, melhor predizer a exposição de crianças e adolescentes à droga, de acordo com a capacidade metabólica de cada indivíduo. / Busulfan (Bu) is an alkylating agent used in the conditioning before hematopoietic stem cells transplantation (HSCT) in children. Its pharmacokinetics (PK) presents a great inter-individual variability, which can be partially explained by GSTA1 genetic variants, gene coding for the enzyme glutathione s-tranferase α1, crucial for Bu metabolism. Several methods of predicting PK are available and are used to calculate the Bu dose, based essentially on patients’ age and anthropometric characteristics. So far, a single adult model successfully incorporated this factor into the Bu dose calculation. In the present work, we initially evaluate the performance of the currently available guidelines across the different GSTA1 genetic variants. The PK parameters from the Bu first doses from 101 children and adolescents who have undergone allogenic SCT at the CHU Sainte-Justine, Montreal, Canada following a IV Bu-containing conditioning regimen (BuCR). GSTA1 haplotypes were interpreted in pairs (diplotypes) and then classified in 3 groups based on different potentials of enzyme expression. Measured AUCs and AUCs calculated from Bu doses predicted by 11 different models were classified according to their ability to achieve the AUC target (900 and 1500μM.min). Clearance prediction errors were also calculated. After the first dose, measured AUCs achieved the target in 38.7%. GSTA1 diplotypes groups related to poor Bu metabolism (G3) and fludarabine-containing regimens (FluCR) were the only factors associated with AUC within target (OR 4.7, 95% CI, 1.1 - 19.8, p=0.04 and OR 9.9, 95% CI, 1.6 - 61.7, p=0.01, respectively). Using other methods for dose calculation, percentage of AUCs within target varied from 16% to 74%. G3 and FluCR were, in some models, associated to AUC within the target and in the toxic range, whereas rapid-metabolizers (G1) were correlated with sub therapeutic AUCs. These associations were confirmed in clearance-prediction analysis, where GSTA1 diplotypes groups and conditioning regimen consistently influenced methods’ most prediction errors. Once GSTA1 status was demonstrated to influence significantly the available Bu dosing algorithms, we aimed to develop a population PK (PPK) model which included GSTA1 genetic variants as a covariate. For that, concentration-time data from 112 children and adolescents receiving IV Bu as a component of the conditioning regimen for 115 stem cell transplantations (autologous and allogenic) performed at CHU Sainte-Justine were analyzed. Non-linear mixed effects analysis was used to build a PPK model. Sex, baseline disease (malignant vs. non-malignant), post-menstrual age (PMA) or chronological age, conditioning regimen and GSTA1 diplotypes groups were evaluated as potential covariates. A one-compartment model with first-order elimination best described the data. A factor of Bu metabolism maturation (Fmat) and theoretical allometric scaling of weight were included in the base model. Covariate analysis revealed PMA (ΔOFV=-26.7, p=2.3x10-7) and GSTA1 diplotypes groups (ΔOFV=-11.7, p=0.003), as significant factors on volume and clearance (CL), respectively. CL of rapid metabolizers (G1) were predicted as being 7% higher and that of poor ones (G3) 12% lower than CL of those defined as normal metabolizers (G2). In conclusion, after evidencing that available Bu dosing methods are not suitable for all GSTA1 diplotypes groups, we have proposed the first pharmacogenomics-based dosing algorithm for Bu to be used in a pediatrics. Its use may contribute considerably to better predict Bu exposure in children and adolescents tailoring the dose according to individual metabolic capacity.
13

Modelo de personalização de dose de bussulfano intravenoso baseado no genótipo de GSTA1 durante regime de condicionamento do transplante de células-tronco hematopoiéticas em crianças

Nava, Tiago Rodrigues January 2017 (has links)
O bussulfano (Bu) é um agente alquilante usado no condicionamento que precede o transplante de células-tronco hematopoiéticas (TCTH) em crianças. Sua farmacocinética (FC) apresenta uma grande variabilidade interindivíduo, que pode ser parcialmente explicada pelas variantes genéticas de GSTA1, gene da enzima glutationa S-transferase α1, crucial para o metabolismo do Bu. Vários métodos de predição da FC do Bu são usados para calcular sua dose, essencialmente com base na idade e peso do paciente. Até o momento, apenas um modelo adulto incorporou as variantes de GSTA1 no cálculo da sua dose do Bu. No presente trabalho, avaliou-se, inicialmente, o desempenho de métodos atualmente disponíveis em pediatria, em função das variantes genéticas de GSTA1. Foram avaliados os parâmetros de FC da primeira dose de 101 crianças e adolescentes submetidos a TCTH alogênico no CHU Sainte-Justine, Montreal, Canadá, após regime de condicionamento que incluía Bu intravenoso (BuCR, do inglês busulfan-containing regimen). Os haplótipos GSTA1 foram interpretados em pares (diplótipos) e depois classificados em três grupos com base nos seus diferentes potenciais de expressão enzimática. As AUCs (area under the curve) medidas e as AUCs calculadas a partir de doses de Bu preditas por 11 modelos diferentes foram classificadas de acordo com a sua capacidade para atingir a AUC-alvo (900 a 1.500 μM.min). Também foram calculados os erros de previsão do clearance do Bu. Após a primeira dose, as AUCs medidas atingiram a AUC-alvo em 38,7%. Os diplótipos de GSTA1 relacionados ao metabolismo lento (G3) e regimes contendo fludarabina (FluCR, do inglês fludarabine-containing regimen) foram os únicos fatores associados à AUC no alvo (OR 4,7, IC 95%, 1,1 - 19,8, p = 0,04 e OR 9,9, IC 95%, 1,6 - 61,7, p = 0,01, respectivamente). Utilizando os outros métodos para o cálculo da dose, a percentagem de AUC no alvo variou de 16% a 74%. G3 e FluCR foram, em alguns modelos, associados à AUC no alvo ou na faixa tóxica, enquanto que os metabolizadores rápidos (G1) foram por vezes associados a AUCs subterapêuticas. Essas associações foram confirmadas na análise de predição do clearance, em que os diplótipos da GSTA1 e o regime de condicionamento influenciaram significativamente a maioria dos erros de previsão dos métodos testados. Uma vez que GSTA1 mostrou influenciar significativamente os algoritmos disponíveis, pretendeu-se desenvolver um modelo de FC de população que incluísse variantes genéticas de GSTA1 como um fator no cálculo de dose do Bu. Para tanto, foram analisados os dados de concentração-tempo de 112 crianças e adolescentes que receberam um BuCR mieloablativo antes de 115 TCTH (autólogos e alogênicos), realizados também no CHU Sainte-Justine. Para a construção do modelo de FC de população, utilizou-se uma análise mista não linear. Sexo, doença de base (maligna vs. não maligna), idade pós-menstrual (PMA) ou idade cronológica, regime de condicionamento e diplótipos de GSTA1 foram avaliados como fatores potenciais. Um modelo de um compartimento com eliminação de primeira ordem foi o que melhor descreveu os dados disponíveis. Um fator de maturação do metabolismo de Bu (Fmat) e o peso elevado a exponencial alométrico teórico foram incluídos no modelo de base. A análise dos fatores revelou PMA (ΔOFV = -26,7, p = 2,3x10-7) e grupos de diplótipos de GSTA1 (ΔOFV = -11,7, p = 0,003) como fatores significativamente associados, respectivamente, ao volume e ao CL do Bu. Os CL dos metabolizadores rápidos (G1) foram preditos como sendo 7% mais elevados que os definidos como metabolizadores normais (G2), enquanto que os metabolizadores lentos (G3) foram descritos com CL 12% menor que os G2. Em conclusão, após se evidenciar que os métodos disponíveis para o cálculo de dose do Bu não são adequados para todos os grupos de diplótipos de GSTA1, propôs-se o primeiro algoritmo de cálculo de dose de Bu em pediatria baseado em farmacogenética. Seu uso pode contribuir para uma melhor previsibilidade da FC do Bu e, desta forma, melhor predizer a exposição de crianças e adolescentes à droga, de acordo com a capacidade metabólica de cada indivíduo. / Busulfan (Bu) is an alkylating agent used in the conditioning before hematopoietic stem cells transplantation (HSCT) in children. Its pharmacokinetics (PK) presents a great inter-individual variability, which can be partially explained by GSTA1 genetic variants, gene coding for the enzyme glutathione s-tranferase α1, crucial for Bu metabolism. Several methods of predicting PK are available and are used to calculate the Bu dose, based essentially on patients’ age and anthropometric characteristics. So far, a single adult model successfully incorporated this factor into the Bu dose calculation. In the present work, we initially evaluate the performance of the currently available guidelines across the different GSTA1 genetic variants. The PK parameters from the Bu first doses from 101 children and adolescents who have undergone allogenic SCT at the CHU Sainte-Justine, Montreal, Canada following a IV Bu-containing conditioning regimen (BuCR). GSTA1 haplotypes were interpreted in pairs (diplotypes) and then classified in 3 groups based on different potentials of enzyme expression. Measured AUCs and AUCs calculated from Bu doses predicted by 11 different models were classified according to their ability to achieve the AUC target (900 and 1500μM.min). Clearance prediction errors were also calculated. After the first dose, measured AUCs achieved the target in 38.7%. GSTA1 diplotypes groups related to poor Bu metabolism (G3) and fludarabine-containing regimens (FluCR) were the only factors associated with AUC within target (OR 4.7, 95% CI, 1.1 - 19.8, p=0.04 and OR 9.9, 95% CI, 1.6 - 61.7, p=0.01, respectively). Using other methods for dose calculation, percentage of AUCs within target varied from 16% to 74%. G3 and FluCR were, in some models, associated to AUC within the target and in the toxic range, whereas rapid-metabolizers (G1) were correlated with sub therapeutic AUCs. These associations were confirmed in clearance-prediction analysis, where GSTA1 diplotypes groups and conditioning regimen consistently influenced methods’ most prediction errors. Once GSTA1 status was demonstrated to influence significantly the available Bu dosing algorithms, we aimed to develop a population PK (PPK) model which included GSTA1 genetic variants as a covariate. For that, concentration-time data from 112 children and adolescents receiving IV Bu as a component of the conditioning regimen for 115 stem cell transplantations (autologous and allogenic) performed at CHU Sainte-Justine were analyzed. Non-linear mixed effects analysis was used to build a PPK model. Sex, baseline disease (malignant vs. non-malignant), post-menstrual age (PMA) or chronological age, conditioning regimen and GSTA1 diplotypes groups were evaluated as potential covariates. A one-compartment model with first-order elimination best described the data. A factor of Bu metabolism maturation (Fmat) and theoretical allometric scaling of weight were included in the base model. Covariate analysis revealed PMA (ΔOFV=-26.7, p=2.3x10-7) and GSTA1 diplotypes groups (ΔOFV=-11.7, p=0.003), as significant factors on volume and clearance (CL), respectively. CL of rapid metabolizers (G1) were predicted as being 7% higher and that of poor ones (G3) 12% lower than CL of those defined as normal metabolizers (G2). In conclusion, after evidencing that available Bu dosing methods are not suitable for all GSTA1 diplotypes groups, we have proposed the first pharmacogenomics-based dosing algorithm for Bu to be used in a pediatrics. Its use may contribute considerably to better predict Bu exposure in children and adolescents tailoring the dose according to individual metabolic capacity.
14

Les Déterminants Génétiques de la Pharmacocinétique du Busulfan et les Résultats de la Transplantation

Rezgui, Mohamed Aziz 12 1900 (has links)
Le busulfan (Bu) est un composé clé de la phase de conditionnement chez les enfants subissant une transplantation des cellules souches hématopoïétiques (TCSH). Les différences inter-individuelles de la pharmacocinétique (PK) du Bu pourraient affecter son efficacité et sa toxicité. Le Bu est principalement métabolisé par la glutathion-S-transférase (GST). Nous avons étudié la relation des génotypes GSTA1, GSTM1 et GSTP1 avec la PK de la première dose de Bu et la relation avec les résultats de la TCSH chez 69 enfants recevant un régime de conditionnement myéloablatif. Le génotype GSTM1 nul a corrélé avec une exposition élevée du Bu et une faible clairance (CL) chez les patients âgés de 4 ans (p ≤ 0,04). Dans le respect du rôle fonctionnel suggéré d’haplotype GSTA1 *A2, il a été associé à des niveaux plus faibles de médicaments et des niveaux élevés de CL (p ≤ 0,03). L’effet Gène-dose a également été observé (p = ≤ 0,007). L’haplotype de GSTA1 était associé avec les résultats de la TCSH. Les porteurs de deux copies d’haplotype *A2 avaient une meilleure survie sans événement (p = 0,03). En revanche, les individus homozygotes pour haplotypes * B et *B1 ont un risque plus élevé d’atteindre la maladie veino-occlusive (MVO) (p = 0,009). Les individus porteurs de GSTM1 nul âgés de 4 ans possèdent un risque plus fréquent d’avoir la maladie du greffon contre l'hôte (GvHD) (p = 0,03). En conclusion, nous avons montré que les variantes génétiques de GST influencent la PK du BU et les résultats de la TCSH chez les enfants. Pour l'ajustement de la posologie, un modèle avec l'inclusion des facteurs génétiques et non génétiques devrait être évalué et validé dans une étude prospective. / Busulfan (Bu) is a key compound of conditioning regimen in children undergoing hematopoietic stem cell transplantation (HSCT). Inter-individual differences in Bu pharmacokinetics might affect Bu efficacy and toxicity. Since Bu is mainly metabolized by glutathione S-transferase (GST), we investigated the relationship between GSTA1, GSTM1 and GSTP1 genotypes with first-dose Bu pharmacokinetics (PK), and relationship with HSCT outcomes in 69 children receiving myeloablative conditioning regimen. GSTM1 null genotype correlated with higher Bu exposure and lower clearance in patients older than 4 years (p≤0.04). In accordance with the suggested functional role GSTA1*A2 haplotype was associated with lower drug levels and higher drug clearance (p≤0.03). Gene-dosage effect was also observed (p=≤0.007). GSTA1 haplotypes were associated with HSCT outcomes Patients with two copies of haplotype *A2 had better event free survival (p=0.03). In contrast, homozygous individuals for haplotypes *B and *B1 had higher occurrence of veno-occlusive disease (p=0.009). GSTM1 null individuals older than 4 years had more frequently graft versus host disease (p=0.03). In conclusion, we showed that GST gene variants influence Bu PK and outcomes of HSCT in children. A model for the dosage adjustment with the inclusion of genetic and non-genetic factors should be evaluated in a future prospective validation cohort.
15

Optimisation de l’administration des médicaments chez les enfants transplantés grâce à la pharmacocinétique de population

Kassir, Nastya 03 1900 (has links)
Ce travail de thèse porte sur l’application de la pharmacocinétique de population dans le but d’optimiser l’utilisation de certains médicaments chez les enfants immunosupprimés et subissant une greffe. Parmi les différents médicaments utilisés chez les enfants immunosupprimés, l’utilisation du busulfan, du tacrolimus et du voriconazole reste problématique, notamment à cause d’une très grande variabilité interindividuelle de leur pharmacocinétique rendant nécessaire l’individualisation des doses par le suivi thérapeutique pharmacologique. De plus, ces médicaments n’ont pas fait l’objet d’études chez les enfants et les doses sont adaptées à partir des adultes. Cette dernière pratique ne prend pas en compte les particularités pharmacologiques qui caractérisent l’enfant tout au long de son développement et rend illusoire l’extrapolation aux enfants des données acquises chez les adultes. Les travaux effectués dans le cadre de cette thèse ont étudié successivement la pharmacocinétique du busulfan, du voriconazole et du tacrolimus par une approche de population en une étape (modèles non-linéaires à effets mixtes). Ces modèles ont permis d’identifier les principales sources de variabilités interindividuelles sur les paramètres pharmacocinétiques. Les covariables identifiées sont la surface corporelle et le poids. Ces résultats confirment l’importance de tenir en compte l’effet de la croissance en pédiatrie. Ces paramètres ont été inclus de façon allométrique dans les modèles. Cette approche permet de séparer l’effet de la mesure anthropométrique d’autres covariables et permet la comparaison des paramètres pharmacocinétiques en pédiatrie avec ceux des adultes. La prise en compte de ces covariables explicatives devrait permettre d’améliorer la prise en charge a priori des patients. Ces modèles développés ont été évalués pour confirmer leur stabilité, leur performance de simulation et leur capacité à répondre aux objectifs initiaux de la modélisation. Dans le cas du busulfan, le modèle validé a été utilisé pour proposer par simulation une posologie qui améliorerait l’atteinte de l’exposition cible, diminuerait l’échec thérapeutique et les risques de toxicité. Le modèle développé pour le voriconazole, a permis de confirmer la grande variabilité interindividuelle dans sa pharmacocinétique chez les enfants immunosupprimés. Le nombre limité de patients n’a pas permis d’identifier des covariables expliquant cette variabilité. Sur la base du modèle de pharmacocinétique de population du tacrolimus, un estimateur Bayesien a été mis au point, qui est le premier dans cette population de transplantés hépatiques pédiatriques. Cet estimateur permet de prédire les paramètres pharmacocinétiques et l’exposition individuelle au tacrolimus sur la base d’un nombre limité de prélèvements. En conclusion, les travaux de cette thèse ont permis d’appliquer la pharmacocinétique de population en pédiatrie pour explorer les caractéristiques propres à cette population, de décrire la variabilité pharmacocinétique des médicaments utilisés chez les enfants immunosupprimés, en vue de l’individualisation du traitement. Les outils pharmacocinétiques développés s’inscrivent dans une démarche visant à diminuer le taux d'échec thérapeutique et l’incidence des effets indésirables ou toxiques chez les enfants immunosupprimés suite à une transplantation. / This thesis deals with the application of population pharmacokinetics in order to optimize the use of certain medications in immunocompromised children undergoing transplantation. Among the various drugs used in immunocompromised children, the use of busulfan, tacrolimus and voriconazole remains problematic, particularly because of high interindividual variability in their pharmacokinetics necessitating individualized doses based on therapeutic drug monitoring. In addition, these drugs have not been studied in children and the doses are adapted from adults. This practice does not take into account the pharmacological characteristics of pediatrics throughout their development and makes illusory the extrapolation of data acquired in adults to children. The work done in this thesis studied sequentially the pharmacokinetics of busulfan, voriconazole and tacrolimus by a population approach (non-linear mixed effects models). The developed models have identified the main sources of interindividual variability in the pharmacokinetic parameters of these drugs. The identified covariates are body surface area and weight. These results confirm the importance of taking into account the effect of growth in children. These parameters were allometrically included in the models. This approach allows separating the effect of size from other covariates and enables the comparison of pediatric pharmacokinetic parameters with those of adults. The inclusion of these explanatory covariates should improve the management a priori of patients. The developed models were evaluated to confirm their stability, performance, and their ability to answer the original objectives of modeling. In the case of busulfan, the validated model was used to simulate dosing regimens that improve reaching the target exposure, reduce treatment failure and toxicity episodes. The developed population pharmacokinetic model for voriconazole confirmed the large variability in its pharmacokinetics in immunocompromised children. The limited data did not allow identification of covariates explaining this variability. Based on the population pharmacokinetic model of tacrolimus, a Bayesian estimator was developed, which is the first in this population of pediatric liver transplant recipients. This estimator can predict pharmacokinetic parameters and individual exposure to tacrolimus based on a limited number of samples. In conclusion, this thesis allowed applying the population pharmacokinetics approach in pediatrics to explore the characteristics of this population and describe the pharmacokinetic variability of drugs used in immunocompromised children, for the individualization of treatment. Pharmacokinetic tools developed are part of efforts to decrease the rate of treatment failure and the incidence of adverse and toxic events in immunocompromised and transplanted pediatrics.
16

Efeito da fotobiomodulação sobre o estresse oxidativo e o dano no DNA induzido pelo bussulfano em doses encontradas na saliva: estudo in vitro / Effects of photobiomodulation on oxidative stress and DNA damage induced by busulfan at doses found in saliva: an in vitro study

Carvalho, Danielle Lima Correa de 21 August 2018 (has links)
O bussulfano (BU) é um quimioterápico amplamente utilizado para o tratamento de doenças hematológicas e para o condicionamento de transplante de células hematopoiéticas (TCH). As doses plasmáticas desse medicamento são altamente citotóxicas, causando mucosite oral (MO) com alta frequência e gravidade. O BU exibe concentrações na saliva que são similares às encontradas no plasma; porém, ainda não foi confirmado se essa concentração salivar também é citotóxica para queratinócitos, fato que poderia constituir um risco a mais para ocorrência de MO. A prevenção e o tratamento da MO têm sido realizados com laserterapia de baixa intensidade (LTBI), em que a fotobiomodução (FBM) favorece a redução da frequência e da duração das lesões. O objetivo deste trabalho foi verificar se o BU encontrado na saliva é citotóxico e genotóxico para queratinócitos em cultura, bem como se a FBM minimiza esses efeitos. Foram estabelecidos os seguintes grupos experimentais: Grupo Controle (C) - queratinócitos sem nenhum tratamento; Grupo Controle Saliva (CS)- queratinócitos expostos a saliva artificial; Grupo Controle Irradiado (CI) - queratinócitos expostos a LTBI; Grupo Controle Saliva Irradiado (CSI) - queratinócitos expostos a saliva e a LTBI; Grupo BU (BU) - queratinócitos expostos a BU veiculado em saliva; Grupo BU Irradiado (BUI) - queratinócitos expostos a BU salivar e concomitantemente a LTBI. Inicialmente testou-se se a saliva artificial formulada não causava modificações na viabilidade dos queratinócitos cultivados. Em seguida, essas células foram expostas ao BU veiculado em saliva ou inserido diretamente no meio, em concentrações que variaram de 4,0?g/mL a 5,5?g/mL. Nesse ensaio, avaliou-se a viabilidade celular tanto no BU veiculado em saliva quanto do BU inserido no meio de cultura. Foi padronizado também um protocolo de LTBI para ser realizado nas células cultivadas, que incluiu três sessões de laserterapia (660nm, 100mW, 0,028 cm² área do spot, 20 segundos de tempo de irradiação, 8J/cm², 3571mW/cm2) com intervalo de 4h entre elas. Após essas padronizações, a concentração salivar de 5,5?g/mL de BU, que provocou menor viabilidade celular, foi utilizada em outros ensaios, que incluíram avaliação da taxa de morte celular pelo teste de TUNEL, do estresse oxidativo (quantificação da porcentagem de sequestro do radical DPPH, quantificação da superóxido dismutase e da catalase e nível de peroxidação lipídica) e da presença de danos no DNA por intermédio do ensaio cometa. A saliva artificial formulada não alterou a viabilidade celular quando comparada ao grupo controle, porém induziu menor taxa de sequestro do DPPH e níveis discretos de dano no DNA. O BU veiculado na saliva provocou maior redução da viabilidade celular quando comparado ao BU inserido diretamente no meio de cultura (p<0.001), sugerindo que a saliva potencializou o efeito tóxico do BU. Comparando com o grupo controle saliva, o Grupo bussulfano exibiu maior frequência de células TUNEL positivas (p<0.05), menor taxa de sequestro do radical DPPH (p<0.05), maior atividade da catalase (p<0.01) e maior taxa de peroxidação lipídica (p<0.01); não houve, contudo, diferenças em relação à presença da superóxido dismutase. Em comparação ao Grupo BU, o Grupo BU Irradiado exibiu menor quantidade de células TUNEL positivas (p<0.05), maior potencial de sequestro do radical DPPH (p<0.05), menor atividade da catalase (p<0.01) e menor taxa de peroxidação lipídica (p<0.01). Na análise de cometa, o Grupo BU exibiu maior porcentagem de DNA fragmentado em relação ao Grupo BUI (p<0.05). Concluiu-se que a concentração salivar do BU é tóxica para queratinócitos, induzindo estresse oxidativo e danos no DNA, porém a LTBI foi eficaz para a redução da citotoxicidade e da genotoxicidade do BU. / Busulfan (BU) is a chemotherapeutic drug largely used into hematological diseases treatment and also on the conditioning hematopoietic cell transplantation (TCH). BU plasma concentrations are highly cytotoxic, causing oral mucositis (OM) with high frequency and severity. BU salivary concentrations are similar to those found in plasma; however, it has not been confirmed whether the salivary concentration is also cytotoxic for keratinocytes, increasing risk for the occurrence of OM. Low intensity laser therapy (LILT) has been used for prevention and treatment of OM. Photobiomoduction (FBM) contributes to the reduction of frequency and lesions duration. This study aimed to verify if the BU found in saliva is cytotoxic and genotoxic for cultured keratinocytes, as well as whether FBM minimizes these effects. The following experimental groups were established: Control Group (C) - keratinocytes without any treatment; Control Group Saliva (CS) - keratinocytes exposed to artificial saliva; Irradiated Control Group (CI)- keratinocytes exposed to LILT; Control Group Irradiated saliva (CSI) - keratinocytes exposed to saliva and LILT; BU Group (BU) - keratinocytes exposed to salivary BU; BU Group Irradiated (BUI) - keratinocytes exposed to salivary BU and concomitantly to LILT. Initially it was tested the artificial formulated saliva effects over the viability of the cultured keratinocytes. Thereafter, in order to verify the cells viability, these cells were exposed to BU diluted into artificial saliva or medium culture, the concentrations ranged from 4.0 ?g / mL to 5.5 ?g / mL. A LILT protocol was also standardized, which consist three laser therapy sessions (660nm, 100mW, 0.028cm² spot areas, 20 seconds irradiation time, 8J / cm², 3571mW / cm ²) with 4 hours interval between them. After these preliminary tests, the BU salivary concentration of 5.5 ?g / mL, which provoked less cell viability, was submitted in other tests, including the evaluation of cell death rate by the TUNEL test, oxidative stress (quantification of the DPPH radical, quantification of superoxide dismutase and catalase and level of lipid peroxidation) and the presence of DNA damage through the comet assay. The formulated artificial saliva did not modify the cell viability when compared to the Control group, but induced a lower DPPH sequestration rate and a discrete level of DNA damage. The salivary BU promoted a greater reduction in cell viability when compared to BU inserted directly into the culture medium (p <0.001), suggesting that saliva increased the toxic effect of BU. Comparing with the Saliva Control group, the BU group showed a higher frequency of TUNEL positive cells (p <0.05), a lower sequestration rate of the DPPH radical (p <0.05), higher catalase activity (p <0.01) and a higher lipid peroxidation rate (p <0.01); however, there were no differences related to the presence of superoxide dismutase. Compared to the BU group, the irradiated BU group had a lower TUNEL positive cells rate (p <0.05), a higher DPPH radical sequestration potential (p <0.05), lower catalase activity (p <0.01) and lower peroxidation rate lipid profile (p <0.01). In the comet analysis, the BU group exhibited a higher percentage of fragmented DNA in relation to the irradiated BU Group (p <0.05). It was concluded that the salivary concentration of BU is toxic to keratinocytes, inducing oxidative stress and DNA damage, but LTBI has been effective in reducing the cytotoxicity and genotoxicity of BU.
17

Monitoração terapêutica do bussulfano oral, após uso de dose teste e durante condicionamento, em pacientes submetidos a transplante alogênico de células-tronco hematopoiéticas / Therapeutic monitoring of oral busulfan, after the use of test dose and during conditioning regimen, in patients undergoing allogeneic hematopoietic stem cell transplantation

EFFTING, Cristiane 13 April 2012 (has links)
Made available in DSpace on 2014-07-29T15:25:20Z (GMT). No. of bitstreams: 1 Tese Cristiane Effting - Ciencias Saude.pdf: 411669 bytes, checksum: 16d94233a45ca7b200c1360cdad9a9d4 (MD5) Previous issue date: 2012-04-13 / Busulfan is an alkylating agent, used for conditioning patients undergoing hematopoietic stem cell transplantation (HSCT). It presents narrow therapeutic range and high variability in pharmacokinetics among patients and doses in the same patient. High plasma concentrations (> 1000 ng mL-1) have been related to toxicity, such as sinusoidal obstruction syndrome, whereas low levels (< 600 ng mL-1) have been associated with primary disease relapse or graft rejection. To avoid problems related to this treatment, therapeutic drug monitoring with dose adjustment has been proposed. Among the methods described, highperformance liquid chromatography (HPLC) is often used. This study aimed at optimizing and validating a technique to dose busulfan by HPLC coupled with photodiode array detector (PDA) and applying it to patientes undergoing HSCT in Goiás. We included eight patients in the group for therapeutic monitoring (MG) and eight in the control group (CG), i.e., with no intervention. Patients in the MG received the test dose (TD) 14 days before the treatment; after determining busulfan pharmacokinetic profile for each patient, the dose was adjusted to the therapeutic objective of 900 ng mL-1. The conditions for chromatography run were: HPLC/PDA, column ACE® C18 (150 mm x 4 mm); mobile phase methanol/water/acetonitrile (65:20:15, v/v/v); eluent flow rate of 1 mL min-1; internal standard 1,6-bis-(methanesulfonyloxy)hexane; UV detection &#955; = 276 nm; derivatization with sodium diethylcarbamate; liquid-liquid extraction with ethyl acetate after precipitation with acetonitrile. We included eight patients in the group for therapeutic monitoring (MG) and eight in the control group (CG). Results obtained: linearity, analyzed through the calibration curve, of 200 5000 ng mL-1; precision, in terms of repeatability (intra-run), of 1.25%-11.25%, and intermediary (inter-run), of 2.17%-10.71%; accuracy of 89.61%-102.18%; recovery of 89%. Half of the patients required dose increase and the mean dose administered was 1.02±0.19 mg kg-1. High variability was observed in assessed pharmacokinetic parameters: 38% variation in Css ____ between TD and conditioning regimen; half-life increased by 11%; ClT/F decreased by 30%, suggesting accumulation of busulfan when the drug is administered in a multiple dose regimen. Although lower than reported in the literature, this variation may be associated with toxicity or failure in treatment, justifying patient monitoring and enhancing validity of previous pharmacokinetic evaluation using TD regimen. Compared to the CG, this variation did not present impact on toxicity, mortality, and survival rates. Other studies with intervention during monitoring and a higher number of patients may present positive impact on the results of HSCT. / O bussulfano é um agente alquilante utilizado em regimes de condicionamento para ablação medular em pacientes submetidos a transplante de células-tronco hematopoiéticas (TCTH). Apresenta estreita faixa terapêutica e grande variabilidade farmacocinética entre pacientes e entre doses no mesmo paciente. Em altas concentrações plasmáticas (> 1000 ng mL-1), associa-se a toxicidade, como síndrome de obstrução sinusoidal, e em baixas (< 600 ng mL-1), a recaída da doença de base ou rejeição do enxerto. Para evitar problemas relacionados ao tratamento, tem sido proposta sua monitoração terapêutica com ajuste da dose. Entre as metodologias descritas, a cromatografia líquida de alta eficiência (CLAE) é frequentemente utilizada. Este estudo objetivou otimizar e validar técnica de dosagem de bussulfano em CLAE acoplada a detector de arranjo de diodos (DAD) e aplicá-la a pacientes submetidos a TCTH em Goiás. Foram incluídos oito pacientes no grupo para monitoração terapêutica (GM) e oito no grupo controle (GC), ou seja, sem intervenção. Os pacientes do GM receberam a dose teste (DT) 14 dias antes do tratamento; após determinação do perfil farmacocinético do fármaco para cada paciente, a dose foi ajustada para o objetivo terapêutico de 900 ng mL-1. As condições cromatográficas foram: CLAE/DAD, coluna ACE® C18 (150 mm x 4 mm); fase móvel metanol/água/acetonitrila (65:20:15, v/v/v); fluxo de 1 mL min-1; padrão interno 1,6-bis-(metanosulfoniloxi)hexano; detecção UV &#955; = 276 nm; derivatização com dietilditiocarbamato de sódio; extração líquida-líquida com acetato de etila após precipitação com acetonitrila. Os resultados incluíram: linearidade, analisada pela curva de calibração, de 200 5000 ng mL-1; precisão, em termos de repetibilidade (intracorrida), de 1,25%-11,25% e intermediária (intercorrida), de 2,17%-10,71%; exatidão de 89,61%-102,18%; recuperação de 89%. Metade dos pacientes necessitou de aumento da dose e a média da dose administrada foi de 1,02±0,19 mg kg-1. Observou-se alta variabilidade nos parâmetros farmacocinéticos avaliados: variação de 38% da Css ____ entre DT e condicionamento; meia-vida aumentada em 11%; ClT/F reduzido em 30%, sugerindo acúmulo do fármaco quando administrado em esquema de dose múltipla. Embora menor do que a relatada na literatura, essa variação pode estar associada a toxicidade ou falha do tratamento, justificando a monitoração e acentuando a validade da avaliação farmacocinética prévia usando o esquema de DT. Comparada com o GC, essa variação não apresentou impacto sobre toxicidade, mortalidade e sobrevida. Outros trabalhos com intervenções durante a monitoração e maior número de pacientes podem apresentar impacto positivo nos desfechos do TCTH.
18

Les Déterminants Génétiques de la Pharmacocinétique du Busulfan et les Résultats de la Transplantation

Rezgui, Mohamed Aziz 12 1900 (has links)
Le busulfan (Bu) est un composé clé de la phase de conditionnement chez les enfants subissant une transplantation des cellules souches hématopoïétiques (TCSH). Les différences inter-individuelles de la pharmacocinétique (PK) du Bu pourraient affecter son efficacité et sa toxicité. Le Bu est principalement métabolisé par la glutathion-S-transférase (GST). Nous avons étudié la relation des génotypes GSTA1, GSTM1 et GSTP1 avec la PK de la première dose de Bu et la relation avec les résultats de la TCSH chez 69 enfants recevant un régime de conditionnement myéloablatif. Le génotype GSTM1 nul a corrélé avec une exposition élevée du Bu et une faible clairance (CL) chez les patients âgés de 4 ans (p ≤ 0,04). Dans le respect du rôle fonctionnel suggéré d’haplotype GSTA1 *A2, il a été associé à des niveaux plus faibles de médicaments et des niveaux élevés de CL (p ≤ 0,03). L’effet Gène-dose a également été observé (p = ≤ 0,007). L’haplotype de GSTA1 était associé avec les résultats de la TCSH. Les porteurs de deux copies d’haplotype *A2 avaient une meilleure survie sans événement (p = 0,03). En revanche, les individus homozygotes pour haplotypes * B et *B1 ont un risque plus élevé d’atteindre la maladie veino-occlusive (MVO) (p = 0,009). Les individus porteurs de GSTM1 nul âgés de 4 ans possèdent un risque plus fréquent d’avoir la maladie du greffon contre l'hôte (GvHD) (p = 0,03). En conclusion, nous avons montré que les variantes génétiques de GST influencent la PK du BU et les résultats de la TCSH chez les enfants. Pour l'ajustement de la posologie, un modèle avec l'inclusion des facteurs génétiques et non génétiques devrait être évalué et validé dans une étude prospective. / Busulfan (Bu) is a key compound of conditioning regimen in children undergoing hematopoietic stem cell transplantation (HSCT). Inter-individual differences in Bu pharmacokinetics might affect Bu efficacy and toxicity. Since Bu is mainly metabolized by glutathione S-transferase (GST), we investigated the relationship between GSTA1, GSTM1 and GSTP1 genotypes with first-dose Bu pharmacokinetics (PK), and relationship with HSCT outcomes in 69 children receiving myeloablative conditioning regimen. GSTM1 null genotype correlated with higher Bu exposure and lower clearance in patients older than 4 years (p≤0.04). In accordance with the suggested functional role GSTA1*A2 haplotype was associated with lower drug levels and higher drug clearance (p≤0.03). Gene-dosage effect was also observed (p=≤0.007). GSTA1 haplotypes were associated with HSCT outcomes Patients with two copies of haplotype *A2 had better event free survival (p=0.03). In contrast, homozygous individuals for haplotypes *B and *B1 had higher occurrence of veno-occlusive disease (p=0.009). GSTM1 null individuals older than 4 years had more frequently graft versus host disease (p=0.03). In conclusion, we showed that GST gene variants influence Bu PK and outcomes of HSCT in children. A model for the dosage adjustment with the inclusion of genetic and non-genetic factors should be evaluated in a future prospective validation cohort.
19

Optimisation de l’administration des médicaments chez les enfants transplantés grâce à la pharmacocinétique de population

Kassir, Nastya 03 1900 (has links)
Ce travail de thèse porte sur l’application de la pharmacocinétique de population dans le but d’optimiser l’utilisation de certains médicaments chez les enfants immunosupprimés et subissant une greffe. Parmi les différents médicaments utilisés chez les enfants immunosupprimés, l’utilisation du busulfan, du tacrolimus et du voriconazole reste problématique, notamment à cause d’une très grande variabilité interindividuelle de leur pharmacocinétique rendant nécessaire l’individualisation des doses par le suivi thérapeutique pharmacologique. De plus, ces médicaments n’ont pas fait l’objet d’études chez les enfants et les doses sont adaptées à partir des adultes. Cette dernière pratique ne prend pas en compte les particularités pharmacologiques qui caractérisent l’enfant tout au long de son développement et rend illusoire l’extrapolation aux enfants des données acquises chez les adultes. Les travaux effectués dans le cadre de cette thèse ont étudié successivement la pharmacocinétique du busulfan, du voriconazole et du tacrolimus par une approche de population en une étape (modèles non-linéaires à effets mixtes). Ces modèles ont permis d’identifier les principales sources de variabilités interindividuelles sur les paramètres pharmacocinétiques. Les covariables identifiées sont la surface corporelle et le poids. Ces résultats confirment l’importance de tenir en compte l’effet de la croissance en pédiatrie. Ces paramètres ont été inclus de façon allométrique dans les modèles. Cette approche permet de séparer l’effet de la mesure anthropométrique d’autres covariables et permet la comparaison des paramètres pharmacocinétiques en pédiatrie avec ceux des adultes. La prise en compte de ces covariables explicatives devrait permettre d’améliorer la prise en charge a priori des patients. Ces modèles développés ont été évalués pour confirmer leur stabilité, leur performance de simulation et leur capacité à répondre aux objectifs initiaux de la modélisation. Dans le cas du busulfan, le modèle validé a été utilisé pour proposer par simulation une posologie qui améliorerait l’atteinte de l’exposition cible, diminuerait l’échec thérapeutique et les risques de toxicité. Le modèle développé pour le voriconazole, a permis de confirmer la grande variabilité interindividuelle dans sa pharmacocinétique chez les enfants immunosupprimés. Le nombre limité de patients n’a pas permis d’identifier des covariables expliquant cette variabilité. Sur la base du modèle de pharmacocinétique de population du tacrolimus, un estimateur Bayesien a été mis au point, qui est le premier dans cette population de transplantés hépatiques pédiatriques. Cet estimateur permet de prédire les paramètres pharmacocinétiques et l’exposition individuelle au tacrolimus sur la base d’un nombre limité de prélèvements. En conclusion, les travaux de cette thèse ont permis d’appliquer la pharmacocinétique de population en pédiatrie pour explorer les caractéristiques propres à cette population, de décrire la variabilité pharmacocinétique des médicaments utilisés chez les enfants immunosupprimés, en vue de l’individualisation du traitement. Les outils pharmacocinétiques développés s’inscrivent dans une démarche visant à diminuer le taux d'échec thérapeutique et l’incidence des effets indésirables ou toxiques chez les enfants immunosupprimés suite à une transplantation. / This thesis deals with the application of population pharmacokinetics in order to optimize the use of certain medications in immunocompromised children undergoing transplantation. Among the various drugs used in immunocompromised children, the use of busulfan, tacrolimus and voriconazole remains problematic, particularly because of high interindividual variability in their pharmacokinetics necessitating individualized doses based on therapeutic drug monitoring. In addition, these drugs have not been studied in children and the doses are adapted from adults. This practice does not take into account the pharmacological characteristics of pediatrics throughout their development and makes illusory the extrapolation of data acquired in adults to children. The work done in this thesis studied sequentially the pharmacokinetics of busulfan, voriconazole and tacrolimus by a population approach (non-linear mixed effects models). The developed models have identified the main sources of interindividual variability in the pharmacokinetic parameters of these drugs. The identified covariates are body surface area and weight. These results confirm the importance of taking into account the effect of growth in children. These parameters were allometrically included in the models. This approach allows separating the effect of size from other covariates and enables the comparison of pediatric pharmacokinetic parameters with those of adults. The inclusion of these explanatory covariates should improve the management a priori of patients. The developed models were evaluated to confirm their stability, performance, and their ability to answer the original objectives of modeling. In the case of busulfan, the validated model was used to simulate dosing regimens that improve reaching the target exposure, reduce treatment failure and toxicity episodes. The developed population pharmacokinetic model for voriconazole confirmed the large variability in its pharmacokinetics in immunocompromised children. The limited data did not allow identification of covariates explaining this variability. Based on the population pharmacokinetic model of tacrolimus, a Bayesian estimator was developed, which is the first in this population of pediatric liver transplant recipients. This estimator can predict pharmacokinetic parameters and individual exposure to tacrolimus based on a limited number of samples. In conclusion, this thesis allowed applying the population pharmacokinetics approach in pediatrics to explore the characteristics of this population and describe the pharmacokinetic variability of drugs used in immunocompromised children, for the individualization of treatment. Pharmacokinetic tools developed are part of efforts to decrease the rate of treatment failure and the incidence of adverse and toxic events in immunocompromised and transplanted pediatrics.
20

Study of genetic factors in treatment-related complications in patients with childhood acute lymphoblastic leukemia and post transplantation of hematopoietic stem cells

Petrykey, Kateryna 12 1900 (has links)
La leucémie lymphoblastique aiguë (LLA) est le cancer le plus fréquent chez les enfants. Malgré le fait que plus de 80% des enfants atteints de LLA sont aujourd'hui guéris de leur maladie, ce succès a toutefois un prix élevé, car l’exposition aux médicaments cytotoxique et/ou à l’irradiation pendant une période vulnérable du développement de l’enfant peut entraîner des conséquences à long terme. En effet, environ 60% des enfants ayant survécu à une LLA devront vivre avec des problèmes de santé liés au traitement, également appelés effets indésirables tardifs (late-adverse effects, LAEs). Parmi ces derniers, on notera des problèmes métaboliques, l’ostéoporose, une altération des fonctions cognitives ou cardiaques, ainsi que la dépression et l’anxiété. Si certains survivants ne présentent aucune de ces complications, d'autres peuvent en avoir plusieurs. Différents facteurs peuvent contribuer à cette variabilité, notamment le traitement reçu, les caractéristiques de la maladie, les habitudes de vie et, surtout, la constitution génétique du patient. Ce projet s'est concentré sur les biomarqueurs génétiques permettant d'identifier les individus les plus susceptibles de souffrir de LAEs. Récemment, une étude exhaustive (évaluations cliniques, psychosociales et biochimiques) s’est déroulée au CHU Sainte-Justine pour caractériser chacune de ces morbidités chez 250 survivants de la LLA de l'enfant (cohorte PETALE). De plus, on a obtenu le profil génétique de chaque participant. Nous avons utilisé cet ensemble de données et des outils statistiques et bio-informatiques pour réaliser des études d'association comparant la fréquence des variants génétiques chez les survivants ayant développé ou non des LAEs; en particulier, les complications cardiovasculaires et neurocognitives, ainsi que les troubles de l'humeur tels que l'anxiété et la dépression. D'autres facteurs de risque tels que les caractéristiques de traitement et/ou de la leucémie ont été pris en compte lors de l'analyse pour dériver les meilleurs prédicteurs génétiques. Ainsi, en utilisant l'approche des gènes candidats, nous avons identifié les variants communs des gènes MTR, PPARA, ABCC3, CALML5, CACNB2 et PCDHB10 qui étaient associés à des déficits de performance des tests neurocognitifs, tandis que les variants des gènes SLCO1B1 et EPHA5 étaient associés à l'anxiété et à la dépression. Deux variants, rs1805087 dans le gène MTR et rs58225473 dans le gène CACNB2 sont particulièrement intéressants, car ces associations ont été validées dans la cohorte de réplication SJLIFE (St. Jude Children's Research Hospital, Memphis, USA). Les analyses d'association ont été complémentées par une étude d'association à l'échelle de l'exome, qui a identifié plusieurs gènes supplémentaires comme des modulateurs potentiels du risque de développer des complications neurocognitives liées au traitement (gènes AK8 et ZNF382), ainsi que l'anxiété et la dépression (gènes PTPRZ1, MUC16, TNRC6C-AS1, APOL2, C6orf165, EXO5, CYP2W1 et PCMTD1). Le variant rs61732180 du gène ZNF382 a ensuite été validé dans la cohorte de réplication SJLIFE. Également, nous avons effectué des analyses d’association concernant les complications cardiaques liées au traitement qui ont identifié plusieurs nouveaux marqueurs associés à ces complications dans les gènes TTN, NOS1, ABCG2, CBR1, ABCC5, AKR1C3, NOD2 et ZNF267. De plus, nous avons résumé les connaissances actuelles sur les marqueurs pharmacogénomiques qui ont été associés aux effets de cardiotoxicités, induites par les anthracyclines, qui affectent les patients atteints de cancer pédiatrique. Nous avons également inclus un aperçu de l'applicabilité des résultats rapportés, notamment ceux qui ont été validés dans la cohorte PETALE. Par ailleurs, nous nous sommes intéressés aux complications qui surviennent après une greffe de cellules souches hématopoïétiques. Nous avons appliqué des approches bio-informatiques et statistiques similaires pour obtenir un profil plus complet de la composante génétique derrière ces complications potentiellement mortelles. Ainsi, une étude d'association à l'échelle de l'exome a été réalisée dans une cohorte de patients pédiatriques subissant une greffe de cellules souches hématopoïétiques après un régime de conditionnement contenant du busulfan. Nous avons identifié de nouvelles variations génétiques conférant un risque plus élevé de syndrome d'obstruction sinusoïdale (notamment dans les gènes UGT2B10, BHLHE22, et KIAA1715) et de maladie aiguë du greffon contre l'hôte (dans les gènes ERC1, PLEK, NOP9 et SPRED1), qui pourraient être utiles pour des stratégies personnalisées de prévention et de traitement. Ces travaux contribuent à la compréhension de l'influence des facteurs génétiques sur le risque de développer des complications liées au traitement, tant au cours du traitement qu'à long terme. De plus, les marqueurs génétiques signalés ainsi que d'autres facteurs de risque connus peuvent conduire à des modèles de prédiction identifiant les patients à risque accru de ces complications. / Acute lymphoblastic leukemia (ALL) is the most common cancer in children. Even though more than 80% of children with ALL are now cured of their disease, this success comes at a high price as exposure to cytotoxic drugs and/or radiation during a vulnerable period of child development may have long-term consequences. In fact, approximately 60% of children who survive ALL will have to live with treatment-related health problems, also called late-adverse effects (LAEs). These include metabolic problems, osteoporosis, impaired cardiac or cognitive functions, as well as depression and anxiety. While some survivors do not have any of these complications, others may have more than one. Different factors can contribute to this variability, in particular, the treatment received, the characteristics of the disease, the lifestyle, and, above all, the genetic makeup of the patient. This project focused on genetic biomarkers capable of identifying the individuals most likely to suffer from LAEs. Recently, an exhaustive study (clinical, psychosocial, and biochemical evaluations) took place at Sainte-Justine University Health Center (Montreal, Canada), with the goal to characterize each of these morbidities in 250 survivors of childhood ALL (PETALE cohort). In addition, the genetic profile of each participant was obtained, and we used statistical and bioinformatics tools to perform association studies on this dataset in order to compare the frequency of genetic variants in survivors with or without LAEs. We evaluated cardiovascular and neurocognitive complications, as well as mood disorders such as anxiety and depression. Other risk factors, such as treatment and/or leukemia characteristics were also considered during the analysis to derive the best genetic predictors. Thus, using the candidate gene approach, we identified common variants in the MTR, PPARA, ABCC3, CALML5, CACNB2, and PCDHB10 genes that were associated with deficits in neurocognitive tests performance, whereas variants in the SLCO1B1 and EPHA5 genes were associated with anxiety and depression. Two variants, rs1805087 in the MTR gene and rs58225473 in the CACNB2 gene, are of particular interest since these associations were validated in an independent SJLIFE replication cohort (St. Jude Children's Research Hospital, Memphis, USA). The association analyses were complemented by an exome-wide association study, which identified several additional genes as potential modulators of the risk of developing treatment-related neurocognitive complications (genes AK8 and ZNF382), as well as anxiety and depression (genes PTPRZ1, MUC16, TNRC6C-AS1, APOL2, C6orf165, EXO5, CYP2W1, and PCMTD1). Variant rs61732180 in the ZNF382 gene was further validated in the replication SJLIFE cohort. To a great extent, we performed association analyses regarding treatment-related cardiac complications which identified several novel markers associated with these toxicities in the TTN, NOS1, ABCG2, CBR1, ABCC5, AKR1C3, NOD2, and ZNF267 genes in survivors of childhood ALL. In addition, we summarized the current knowledge on pharmacogenomic markers related to anthracycline-induced cardiotoxicity affecting pediatric cancer patients. We also included a brief overview of the applicability of reported findings to the PETALE cohort, validating several of them. Besides, we were interested in the complications that arise after a hematopoietic stem cell transplantation. We applied similar bioinformatics and statistical approaches to gain a more complete insight into the genetic component behind these life-threatening complications. Thus, an exome-wide association study was performed in a cohort of pediatric patients undergoing hematopoietic stem cell transplantation following a conditioning regimen containing busulfan. Our results identified new genetic variations conferring a higher risk of sinusoidal obstruction syndrome (notably in the UGT2B10, BHLHE22, and KIAA1715 genes) and acute graft-versus-host disease (ERC1, PLEK, NOP9, and SPRED1 genes), which could be useful for personalized prevention and treatment strategies. This work contributes to the understanding of the influence of genetic factors on the risk of developing treatment-related complications, both during treatment and in the long term. Furthermore, the reported genetic markers along with other known risk factors can lead to prediction models identifying patients at increased risk for these complications.

Page generated in 0.2565 seconds