• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 6
  • 4
  • 4
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 29
  • 7
  • 6
  • 6
  • 5
  • 5
  • 5
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 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.
21

Sorafenib-Induced Apoptosis in Hepatocellular Carcinoma Is Reversed by SIRT1

Garten, Antje, Grohmann, Theresa, Klockova, Katarina, Lavery, Gareth G., Kiess, Wieland, Penke, Melanie 06 February 2024 (has links)
Sorafenib is a multi-kinase inhibitor and one of the few systemic treatment options for patients with advanced hepatocellular carcinomas (HCCs). Resistance to sorafenib develops frequently and could be mediated by the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin (SIRT)1. We aimed to test whether sorafenib efficacy is influenced by cellular NAD levels and NAD-dependent SIRT1 function. We analyzed sorafenib effects on apoptosis induction, NAD salvage, mitochondrial function, and related signaling pathways in HCC cell lines (HepG2, Hep3B, und HUH7) overexpressing SIRT1 or supplemented with the NAD metabolite nicotinamide mononucleotide (NMN) compared to controls. Treatment of HCC cell lines with sorafenib dose-dependently induced apoptosis and a significant decrease in cellular NAD concentrations. The SIRT1 protein was downregulated in HUH7 cells but not in Hep3B cells. After sorafenib treatment, mitochondrial respiration in permeabilized cells was lower, citrate synthase activity was attenuated, and cellular adenosine triphosphate (ATP) levels were decreased. Concomitant to increased phosphorylation of adenosine monophosphate (AMP)-activated protein kinase (AMPK), sorafenib treatment led to decreased activity of the mechanistic target of rapamycin (mTOR), indicative of energy deprivation. Transient overexpression of SIRT1, as well as NAD repletion by NMN, decreased sorafenib-induced apoptosis. We can, therefore, conclude that sorafenib influences the NAD/SIRT1/AMPK axis. Overexpression of SIRT1 could be an underlying mechanism of resistance to sorafenib treatment in HCC
22

Efeito do sorafenibe na carcinogênese hepática experimental secundária à doença hepática gordurosa não alcoólica / Sorafenib effect\'s on liver experimental carcinogenesis secondary to non-alcoholic fatty liver disease

Costa, Fernando Gomes de Barros 16 December 2016 (has links)
INTRODUÇÃO E OBJETIVOS: A doença hepática gordurosa não alcoólica (DHGNA) tem sido associada ao carcinoma hepatocelular (CHC), muitas vezes em paciente com hepatopatia avançada. Este estudo objetivou avaliar o efeito do sorafenibe no modelo experimental de CHC avançado secundário à DHGNA, padronizar o PET com 18F-FDG para avaliar o CHC neste modelo e avaliar se há relação entre o grau de avidez pelo 18F-FDG e o grau de diferenciação tumoral do CHC. METODOLOGIA: Estudo foi aprovado pela Comissão de Ética no Uso de Animais. Foram utilizados trinta ratos Sprague-Dawley, machos, com 3 meses de vida, pesando entre 300-400g. O CHC secundário à DHGNA foi induzido pela combinação de dieta hiperlipídica deficiente em colina e dietilnitrosamina na dose de 100 mg/ L na água de beber ad libitum por 16 semanas. Após este período foram suspensos os estímulos carcinogênicos, realizou-se ultrassonografia abdominal para caracterização dos nódulos hepáticos maiores que 2 mm, e foi feita a divisão dos dois grupos segundo randomização e iniciada a administração diária do fármaco por gavagem durante 3 semanas: Controle (n=10) - 1 mL salina, Sorafenibe (n=20): 5mg/ kg/ dia. Ao término do tratamento, os animais realizaram PET (Gamma Medica-Ideas, USA) com 18F-FDG (média de 18F-FDG injetada de 1,02 ± 0,17 mCi ou 37,7 ± 6,29 MBq). Três dias após o PET, os animais foram anestesiados e foi feita a eutanásia, quando foi coletado material hepático. As lâminas foram avaliadas, por patologista veterinário experiente, na coloração de hematoxilina-eosina e imunohistoquímica para glutamina sintetase, antígeno específico de hepatócitos 1 e citoqueratina-19. RESULTADOS: A mortalidade nos dois grupos foi de 60% (p=0,07). Os achados ultrassonográficos mostraram grupos homogêneos com média de nódulos por animal: 4,88 ± 2,75 no controle e 4,95 ± 3,11 no sorafenibe (p=0,48). Na 19ª semana, viu-se que a média de lesões hipercaptantes por animal no PET foi de 4,37 ± 1,59 no grupo sorafenibe e 8,5 ± 3,7 no controle (p=0,006). A avidez máxima do 18F-FDG (SUVmáx) foi diferente entre os grupos estudados: 2,4 ± 1,98 no sorafenibe e 3,8 ± 1,74 no controle (p=0,01). Houve correlação direta entre o CHC pouco diferenciado/indiferenciado e os maiores valores de SUVmed (R2 = 0,34, p=0,04), SUVmax (R2 = 0,44, p=0,01), relação Tumor SUVmax/Fígado SUVmax (R2 = 0,42, p=0,02) e relação Tumor SUVmax/ Músculo SUVmax (R2 = 0,54, p=0,006). A média por animal de CHC confirmado pela histologia foi menor no grupo sorafenibe que no controle (5,5 ± 1,5 vs 3,3 ± 0,48, p=0,01). E o grupo tratado com sorafenibe apresentou mais CHC bem diferenciado que o controle (39% vs 5%, respectivamente, p=0,01), bem como, menor presença de CHC pouco diferenciado que o grupo controle (52% vs 81%, p-0,003). CONCLUSÃO: O sorafenibe reduziu o número médio de CHC, a agressividade dos CHC e menor SUVmax dos tumores. A metodologia do PET foi padronizada para este modelo animal específico. O PET 18F-FDG pode ser utilizado para avaliar não invasivamente o grau de diferenciação histológica do CHC, pois valores maiores de SUVmed, SUVmax, Tumor SUVmax/Fígado SUVmax e Tumor SUVmax/ Músculo SUVmax foram correlacionados com CHC pouco diferenciado / BACKGROUND AND AIMS: Non-alcoholic fatty liver disease (NAFLD) has been linked to hepatocellular carcinoma (HCC), often in patients with advanced liver disease. This study aimed to: assess the effect of sorafenib in the experimental model of NAFLD related HCC, standardize PET 18F-FDG to be an assessment tool of HCC in this model and to assess if there is a correlation between the degree of avidity for 18F-FDG and the degree of tumor differentiation. METHODS: The ethics committee on animal use approved this study. Thirty male sprague-dawley rats were used, weighing between 300-400g. NAFLD related HCC was induced by the combination of fat and choline deficient diet with diethylnitrosamine (100 mg/L) in drinking water for 16 weeks. After this period these carcinogenic stimuli were suspended, and liver nodules were identified by abdominal ultrasound. Two groups were randomized: control (n=10) and treatment (n=20). Rats received daily gavage administration of 1 mL saline in the control group and sorafenib (5mg/kg/day) in the treatment group. After treatment, animals performed PET (Gamma Medica-Ideas, USA) with 18F-FDG (average of 18F-FDG injected 1.02 ± 0.17 mCi or 37.7 ± 6.29 MBq). Three days after the PET, the animals were anesthetized and euthanized. Histological aspect was evaluated by experienced veterinary pathologist. RESULTS: The mortality in both groups was 60% (p = 0.07). The sonographic findings showed homogeneous groups with average nodules per animal of: 4.88 ± 2.75 in control and 4.95 ± 3.11 in sorafenib (p = 0.48). On the 19th week, it was observed that the average hypercaptant lesion per animal in PET was 4.37 ± 1.59 in the sorafenib group and 8.5 ± 3.7 in control group (p = 0.006). Average SUVmax was different between groups: 2.4 ± 1.98 in the sorafenib group and 3.8 ± 1.74 in the control group (p = 0.01). A direct correlation was found between the poorly differentiated HCC and larger values of: SUVmed (R2 = 0.34, p = 0.04), SUVmax (R2 = 0.44, p = 0.01) tumorSUVmax / LiverSUVmax ratio (R2 = 0.42, p = 0.02) and tumorSUVmax / MuscleSUVmax ratio (R2 = 0.54, p = 0.006). HCC average per animal was lower in the sorafenib group than in the control group (5.5 ± 1.5 vs. 3.3 ± 0.48; p = 0.01). And sorafenib group had more well differentiated HCC (39% vs 5%, respectively, p = 0.01) and lower presence of poorly differentiated HCC (52% vs 81%, p -0.003) than the control group. CONCLUSION: Sorafenib reduced the average number of HCC, the aggressiveness of HCC and lowered values of tumors SUVmax. The methodology of PET was standardized for this particular animal model. PET 18F-FDG can be used noninvasively to assess the degree of histological differentiation of HCC, as higher values of SUVmed, SUVmax, tumorSUVmax/ LiverSUVmax ratio and tumorSUVmax / MuscleSUVmax ratio were correlated with poorly differentiated HCC
23

Efeito do sorafenibe na carcinogênese hepática experimental secundária à doença hepática gordurosa não alcoólica / Sorafenib effect\'s on liver experimental carcinogenesis secondary to non-alcoholic fatty liver disease

Fernando Gomes de Barros Costa 16 December 2016 (has links)
INTRODUÇÃO E OBJETIVOS: A doença hepática gordurosa não alcoólica (DHGNA) tem sido associada ao carcinoma hepatocelular (CHC), muitas vezes em paciente com hepatopatia avançada. Este estudo objetivou avaliar o efeito do sorafenibe no modelo experimental de CHC avançado secundário à DHGNA, padronizar o PET com 18F-FDG para avaliar o CHC neste modelo e avaliar se há relação entre o grau de avidez pelo 18F-FDG e o grau de diferenciação tumoral do CHC. METODOLOGIA: Estudo foi aprovado pela Comissão de Ética no Uso de Animais. Foram utilizados trinta ratos Sprague-Dawley, machos, com 3 meses de vida, pesando entre 300-400g. O CHC secundário à DHGNA foi induzido pela combinação de dieta hiperlipídica deficiente em colina e dietilnitrosamina na dose de 100 mg/ L na água de beber ad libitum por 16 semanas. Após este período foram suspensos os estímulos carcinogênicos, realizou-se ultrassonografia abdominal para caracterização dos nódulos hepáticos maiores que 2 mm, e foi feita a divisão dos dois grupos segundo randomização e iniciada a administração diária do fármaco por gavagem durante 3 semanas: Controle (n=10) - 1 mL salina, Sorafenibe (n=20): 5mg/ kg/ dia. Ao término do tratamento, os animais realizaram PET (Gamma Medica-Ideas, USA) com 18F-FDG (média de 18F-FDG injetada de 1,02 ± 0,17 mCi ou 37,7 ± 6,29 MBq). Três dias após o PET, os animais foram anestesiados e foi feita a eutanásia, quando foi coletado material hepático. As lâminas foram avaliadas, por patologista veterinário experiente, na coloração de hematoxilina-eosina e imunohistoquímica para glutamina sintetase, antígeno específico de hepatócitos 1 e citoqueratina-19. RESULTADOS: A mortalidade nos dois grupos foi de 60% (p=0,07). Os achados ultrassonográficos mostraram grupos homogêneos com média de nódulos por animal: 4,88 ± 2,75 no controle e 4,95 ± 3,11 no sorafenibe (p=0,48). Na 19ª semana, viu-se que a média de lesões hipercaptantes por animal no PET foi de 4,37 ± 1,59 no grupo sorafenibe e 8,5 ± 3,7 no controle (p=0,006). A avidez máxima do 18F-FDG (SUVmáx) foi diferente entre os grupos estudados: 2,4 ± 1,98 no sorafenibe e 3,8 ± 1,74 no controle (p=0,01). Houve correlação direta entre o CHC pouco diferenciado/indiferenciado e os maiores valores de SUVmed (R2 = 0,34, p=0,04), SUVmax (R2 = 0,44, p=0,01), relação Tumor SUVmax/Fígado SUVmax (R2 = 0,42, p=0,02) e relação Tumor SUVmax/ Músculo SUVmax (R2 = 0,54, p=0,006). A média por animal de CHC confirmado pela histologia foi menor no grupo sorafenibe que no controle (5,5 ± 1,5 vs 3,3 ± 0,48, p=0,01). E o grupo tratado com sorafenibe apresentou mais CHC bem diferenciado que o controle (39% vs 5%, respectivamente, p=0,01), bem como, menor presença de CHC pouco diferenciado que o grupo controle (52% vs 81%, p-0,003). CONCLUSÃO: O sorafenibe reduziu o número médio de CHC, a agressividade dos CHC e menor SUVmax dos tumores. A metodologia do PET foi padronizada para este modelo animal específico. O PET 18F-FDG pode ser utilizado para avaliar não invasivamente o grau de diferenciação histológica do CHC, pois valores maiores de SUVmed, SUVmax, Tumor SUVmax/Fígado SUVmax e Tumor SUVmax/ Músculo SUVmax foram correlacionados com CHC pouco diferenciado / BACKGROUND AND AIMS: Non-alcoholic fatty liver disease (NAFLD) has been linked to hepatocellular carcinoma (HCC), often in patients with advanced liver disease. This study aimed to: assess the effect of sorafenib in the experimental model of NAFLD related HCC, standardize PET 18F-FDG to be an assessment tool of HCC in this model and to assess if there is a correlation between the degree of avidity for 18F-FDG and the degree of tumor differentiation. METHODS: The ethics committee on animal use approved this study. Thirty male sprague-dawley rats were used, weighing between 300-400g. NAFLD related HCC was induced by the combination of fat and choline deficient diet with diethylnitrosamine (100 mg/L) in drinking water for 16 weeks. After this period these carcinogenic stimuli were suspended, and liver nodules were identified by abdominal ultrasound. Two groups were randomized: control (n=10) and treatment (n=20). Rats received daily gavage administration of 1 mL saline in the control group and sorafenib (5mg/kg/day) in the treatment group. After treatment, animals performed PET (Gamma Medica-Ideas, USA) with 18F-FDG (average of 18F-FDG injected 1.02 ± 0.17 mCi or 37.7 ± 6.29 MBq). Three days after the PET, the animals were anesthetized and euthanized. Histological aspect was evaluated by experienced veterinary pathologist. RESULTS: The mortality in both groups was 60% (p = 0.07). The sonographic findings showed homogeneous groups with average nodules per animal of: 4.88 ± 2.75 in control and 4.95 ± 3.11 in sorafenib (p = 0.48). On the 19th week, it was observed that the average hypercaptant lesion per animal in PET was 4.37 ± 1.59 in the sorafenib group and 8.5 ± 3.7 in control group (p = 0.006). Average SUVmax was different between groups: 2.4 ± 1.98 in the sorafenib group and 3.8 ± 1.74 in the control group (p = 0.01). A direct correlation was found between the poorly differentiated HCC and larger values of: SUVmed (R2 = 0.34, p = 0.04), SUVmax (R2 = 0.44, p = 0.01) tumorSUVmax / LiverSUVmax ratio (R2 = 0.42, p = 0.02) and tumorSUVmax / MuscleSUVmax ratio (R2 = 0.54, p = 0.006). HCC average per animal was lower in the sorafenib group than in the control group (5.5 ± 1.5 vs. 3.3 ± 0.48; p = 0.01). And sorafenib group had more well differentiated HCC (39% vs 5%, respectively, p = 0.01) and lower presence of poorly differentiated HCC (52% vs 81%, p -0.003) than the control group. CONCLUSION: Sorafenib reduced the average number of HCC, the aggressiveness of HCC and lowered values of tumors SUVmax. The methodology of PET was standardized for this particular animal model. PET 18F-FDG can be used noninvasively to assess the degree of histological differentiation of HCC, as higher values of SUVmed, SUVmax, tumorSUVmax/ LiverSUVmax ratio and tumorSUVmax / MuscleSUVmax ratio were correlated with poorly differentiated HCC
24

CRISPR-barcoding pour l'étude fonctionnelle de mutations oncogéniques dans un contexte d'hétérogénéité intra-tumorale. / Functional analysis of oncogenic driver mutations through CRISPR-barcoding in a context of intratumoral heterogeneity

Guernet, Alexis 27 September 2017 (has links)
Les tumeurs sont généralement constituées de différentes sous-populations de cellules cancéreuses génétiquement hétérogènes, responsables en grande partie de la capacité de la tumeur à évoluer rapidement et à s’adapter aux conditions environnementales. Cette diversité génétique a des conséquences majeures pour le patient, notamment au cours de la progression tumorale et pour l’acquisition d’une résistance aux traitements.Nous avons développé une nouvelle stratégie basée sur la technologie CRISPR/Cas9 qui consiste à introduire, en plus d’une altération de séquence voulue d’un gène d’intérêt, une série de mutations silencieuses, constituant une sorte d’étiquette génétique qui peut être détectée par PCR quantitative ou séquençage de nouvelle génération. En parallèle, un code-barres constitué exclusivement de mutations silencieuses est utilisé comme contrôle interne pour les effets non spécifiques potentiels qui peuvent être engendrés suite au clivage hors-cible par le système CRISPR/Cas9. Cette approche, que nous avons appelée CRISPR-barcoding, permet de générer et de suivre l’émergence d’un petit groupe de cellules cancéreuses contenant une mutation voulue au sein d’une population de cellules non modifiées, représentant ainsi un nouveau modèle expérimental d’hétérogénéité génétique intratumorale. Grâce à une série de preuves de concept, nous avons montré que CRISPR-barcoding est une nouvelle approche qui permet d’étudier de façon simple et rapide les conséquences fonctionnelles de différents types de modifications génétiques apportées directement au niveau de la séquence génomique.Dans la deuxième partie de ma thèse, nous avons utilisé cette nouvelle approche pour l'étude de la résistance du cancer bronchique non à petites cellules (CBNPC) à la thérapie ciblée. Les patients de CBNPC dont la tumeur présente une mutation activatrice de l'epidermal growth factor receptor (EGFR) sont généralement traités avec des inhibiteurs de ce récepteur. Malheureusement, malgré une réponse initiale, presque invariablement ces patients rechutent, suite au développement d’une résistance causée, dans la majorité des cas, par l’apparition de mutations secondaires ou tertiaire de l'EGFR. Grâce à un criblage réalisé en utilisant un modèle de CBNPC basé sur la stratégie CRISPR-barcoding, nous avons pu identifier l’inhibiteur multikinase sorafenib pour sa capacité à prévenir la résistance de ces cellules aux inhibiteurs d’EGFR. Ce composé présente un mécanisme d’action original, impliquant une diminution précoce du niveau de phosphorylation de STAT3, suivie par une baisse considérable de l’expression de l’EGFR, aboutissant à une inhibition des voies intracellulaires en aval de ce récepteur, telles que RAS-MAPK et PI3K-AKT-mTOR. Ces données ont été confirmées in vivo en utilisant un modèle de xénogreffe de cellules de CBNPC modifiées par CRISPR-barcoding.En conclusion, l’ensemble de nos résultats montre que le sorafenib peut prévenir l’émergence de cellules de CBNPC résistantes aux inhibiteurs d’EGFR, indiquant que ce composé pourrait représenter une nouvelle stratégie thérapeutique pour le traitement de ce type de tumeur. / Individual tumors are composed of multiple and genetically distinct subpopulations of transformed cells that can adapt and evolve in a different way based on environmental conditions. This genetic diversity has major consequences for the patient, particularly during tumor progression and for cancer treatment.We devised a new strategy based on CRISPR/Cas9 technology in which a potentially functional modification in the sequence of a gene of interest is coupled with a series of silent point mutations, functioning as a genetic label for cell tracing. In parallel, a second barcode consisting of distinct silent mutations is inserted in the same cell population and used as a control for CRISPR/Cas9 off-target cleavage. This approach, that we named CRISPR barcoding, enables detection of cells containing the mutation of interest within a mass population of unmodified cells using real-time quantitative PCR or deep sequencing. Through a series of proof-of-concept studies, we demonstrated that CRISPR-barcoding is a fast and highly flexible strategy to investigate the functional consequences of a specific genetic modification in a broad range of assays.In the second part of my thesis, we used CRISPR-barcoding to investigate non-small cell lung cancer (NSCLC) resistance to targeted therapy. Some NSCLCs harbor activating mutations of the epidermal growth factor receptor (EGFR) and are addicted to this signaling pathway. These tumors initially show a good response to EGFR inhibitors (EGFRi), but they almost invariably relapse, due to the acquisition of a resistance, as a result of additional genetic alterations, including secondary and tertiary EGFR mutations. Using a CRISPR-barcoding model, we identified the multikinase inhibitor sorafenib for its ability to prevent EGFRi resistance in NSCLC cells. This compound acts through an original mechanism that involves early reduction of STAT3 phosphorylation and late down-regulation of EGFR, resulting in the inhibition of different downstream pathways activated by this receptor, including, RAS-MAPK and PI3K-AKT-mTOR. These results were confirmed in vivo, using a CRISPR-barcoding xenograft model for NSCLC.Altogether, our data indicate that sorafenib can prevent NSCLC resistance to EGFRi through a novel mechanism, thus providing a new potential therapeutic strategy for the treatment of this type of cancer.
25

Identificació de noves dianes terapèutiques en neoplàsies limfoides

Xargay i Torrent, Sílvia 29 October 2012 (has links)
El limfoma de cèl•lules de mantell (MCL) i la leucèmia limfàtica crònica (CLL) són dues neoplàsies limfoides fins al moment incurables, per la qual cosa requereixen la recerca de nous fàrmacs. La identificació de noves dianes terapèutiques per al MCL i la CLL, i del mecanisme molecular d’actuació d’aquests fàrmacs en aquestes neoplàsies, té una gran rellevància translacional. El concepte de medicina personalitzada està basat en l’ús de teràpies dirigides a través d’un coneixement profund dels mecanismes moleculars d’acció d’aquests agents. Amb l’augment del coneixement sobre l’etiologia del càncer, s’han descobert noves dianes alterades específicament en les cèl•lules tumorals. Entre elles, la sobreexpressió de les HDACs, que provoca un estat epigenètic aberrant a la cèl•lula. Això ha motivat el desenvolupament del vorinostat, un inhibidor de les HDACs. Els resultats obtinguts demostren el vorinostat és efectiu i selectiu per a les cèl•lules tumorals de MCL. A nivell molecular, el vorinostat en el MCL indueix l’acetilació de les histones de la regió promotora dels gens de les proapoptòtiques de la família de Bcl-2 BH3-only BIM, BMF i NOXA, que són activats transcripcionalment. Totes tres proteïnes, Bmf, Bim i Noxa, cooperen en el procés d’apoptosi. A més, el vorinostat és sinèrgic amb l’agent que mimetitza les BH3-only, ABT-263. En els últims anys, s’ha identificat un gran nombre de cinases, que regulen moltes vies de supervivència i proliferació cel•lulars, que estan específicament activades i sobreexpressades en cèl•lules tumorals. En particular, recentment les cinases associades al receptor de cèl•lules B (BCR) han esdevingut un focus d’atenció important, ja que està activament implicat en la patogènesi de les neoplàsies limfoides. En aquesta tesi, hem analitzat l’efecte anti-tumoral de l’inhibidor multi-cinasa sorafenib en la CLL i el MCL. Els nostres resultats confirmen que el sorafenib indueix apoptosi selectiva en cèl•lules de CLL i de MCL, fins i tot en els casos de més mal pronòstic de la CLL. A nivell molecular, els nostres resultats han demostrat que el sorafenib indueix una desfosforilació ràpida, sostinguda i simultània de les cinases associades al BCR, Syk i de la família Src cinases. En la CLL i el MCL a temps d’incubació molt curts, el sorafenib provoca una disminució traduccional de l’anti-apoptòtica Mcl-1 alhora que de ciclina D1, sobreexpressada en el MCL. Tant Mcl-1 com ciclina D1 participen en l’apoptosi induïda per sorafenib, la disminució de Mcl-1 altera directament els senyals anti-apoptòtics, mentre que el sorafenib, a més de disminuir els nivells de ciclina D1, també allibera Bax del segrestament per part de ciclina D1, finalment activant mecanismes d’apoptosi caspasa-dependents i independents com AIF. El sorafenib també modula les interaccions del microambient amb la CLL i el MCL. En aquest sentit, s’ha demostrat que el sorafenib bloqueja la migració induïda per la quimiocina CXCL12 via inhibició de FAK, una cinasa substrat de Src que regula i promou la invasió. El complex Src-FAK afecta múltiples proteïnes, com ara les del citoesquelet d’actina. En consistència, el sorafenib bloqueja la reorganització del citoesquelet en cèl•lules de MCL estimulades amb CXCL12. Les cèl•lules estromals del microambient dels teixits, com ara el moll d’os i els teixits limfoides secundaris, interaccionen amb el tumor i en promouen el seu creixement i resistència a fàrmacs. En aquest context, s’ha demostrat que, en presència de l’estroma del microambient, les cèl•lules de MCL i CLL esdevenen resistents als agents quimioterapèutics habituals i el sorafenib és capaç de resensibilitzar-les. Tant la proliferació com l’activació del BCR en la CLL són majors en el gangli limfàtic que en sang. En aquest sentit, s’ha demostrat que el sorafenib és més efectiu en cèl•lules de CLL derivades de gangli limfàtic que les respectives derivades de moll d’os i de sang perifèrica. L’estimulació del BCR en la CLL també provoca la secreció de les citocines atraients de cèl•lules accessòries CCL3 i CCL4, que el sorafenib bloqueja amb eficiència. Globalment, els treballs que constitueixen aquesta tesi doctoral suposen una contribució important en el coneixement del mecanisme molecular d’acció del vorinostat i el sorafenib, fàrmacs nous que podrien ser efectius per al tractament d’aquestes dues entitats, el MCL i la CLL. / Mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL) are two incurable B-lymphoid malignancies, thus new therapeutic strategies are required. In the recent years, anti-tumor therapy has focused on pathways specifically altered in cancer cells. Among them, overexpression of histone deacetylases (HDACs) is a common feature. This led to the development of the HDAC inhibitor vorinostat. This thesis has shown that vorinostat could become a new therapeutic approach for MCL. The results show that vorinostat is effective and selective for MCL cells. The molecular mechanism of action of vorinostat involves acetylation of the histones in the gene promoter of the pro-apoptotic BH3-only from Bcl-2 family, BMF, BIM and NOXA, which induces its transcriptional activation. The respective proteins cooperate in the apoptosis induction. Moreover, vorinostat synergizes with the BH3-only mimetic ABT-263 in MCL. Plenty of kinases that have been recognized to regulate survival and proliferation pathways are overexpressed in tumor cells. In this work, we analyzed the anti-tumor effect of sorafenib, a multi-kinase inhibitor approved by the FDA, in CLL and MCL. Sorafenib induces selective apoptosis in CLL and MCL cells, being also effective in poor prognosis cases of CLL. At the molecular level, sorafenib dephosphorylates the B-cell receptor (BCR) associated kinases, specifically Syk and the Src family kinases. In CLL and MCL, sorafenib inhibits Mcl-1 and cyclin D1 translation. Mcl-1 downregulation is directly impairing anti-apoptotic signals. As for cyclin D1, sorafenib releases Bax from cyclin D1 sequestering. Both processes are involved in apoptosis induction, which occurs through caspase-dependent and independent mechanisms. In addition, sorafenib blocks CXCL12-induced migration and actin polymerization via FAK inhibition, a Src kinase substrate that regulates migration. Stromal cells from the microenvironment interact with and promote tumor growth and drug resistance. In this context, MCL and CLL cells become resistant to conventional chemotherapeutic drugs in the presence of stromal microenvironment, which is counteracted by sorafenib. Activation of the BCR and proliferation in CLL cells is higher in the lymph node microenvironments, a setting where sorafenib has been demonstrated to be more effective. BCR stimulation also leads to CCL3 and CLL4 chemoattractant cytokines secretion in CLL, that sorafenib blocks efficiently. Overall, this thesis is an important contribution to the understanding of the molecular mechanism of action of vorinostat and sorafenib, new drugs that might be effective for the treatment of these two entities, MCL and CLL.
26

Tyrosinkinaseinhibition bei humanen Non-Hodgkin-Lymphomen: Präklinische Evaluation von Sorafenib / Tyrosine Kinase Inhibition at Human Non-Hodgkin s Lymphomas: Preclinical Evaluation of Sorafenib

Schuelper, Nikolai 30 November 2010 (has links)
No description available.
27

Applications d'une modulation pharmacologique des dérives des formes réactives de l'oxygène pour une optimisation thérapeutique des patients traités par chimiothérapie

Coriat, Romain 16 November 2012 (has links) (PDF)
Les formes réactives de l'oxygène (FRO) ont un rôle bien établi dans l'oncogénèse et le fonctionnement des cellules tumorales en augmentant les capacités de prolifération et d'invasion cellulaire. Les FRO présentent une moindre toxicité dans les cellules normales où le niveau de stress oxydant est bas et les systèmes d'oxydoréduction opérationnels. Une modulation pharmacologique de l'équilibre d'oxydoréduction permet d'améliorer l'efficacité des molécules cytotoxiques qui agissent sur les FRO. L'activité anti tumorale de la chimiothérapie étant médiée en partie par le stress oxydant, nous nous sommes dans un premier temps intéressé aux variations du stress oxydant lors d'un traitement par le sorafenib (Nexavar©), un inhibiteur de tyrosine kinases. Nous avons mis en évidence une activité anti tumorale du sorafenib liée à l'augmentation de la production d'anion superoxyde par les cellules. Ce phénomène nous a permis d'identifier un marqueur prédictif d'efficacité du sorafenib, le dosage des Produits d'Oxydation Avancée des Protéines (AOPP), dans le sérum des patients cirrhotiques développant un carcinome hépatocellulaire. Sachant que l'efficacité anti tumorale des chimiothérapies conventionnelles passe en partie par l'induction de FRO, nous nous sommes intéressés dans un deuxième temps au mangafodipir, un modulateur du stress oxydant, qui est connu pour augmenter l'index thérapeutique des agents cytotoxiques in vivo. Le mangafodipir diminue les effets toxiques du stress oxydatif dans des cellules non tumorales et potentialise l'activité anti tumorale de l'oxaliplatine. Ces données nous ont conduits à explorer in vivo et in vitro le mécanisme de la neurotoxicité induite par l'oxaliplatine et le rôle protecteur du mangafodipir. Nous avons ainsi observé dans un modèle murin que le mangafodipir prévient la neurotoxicité induite par l'oxaliplatine au niveau de la gaine de myéline. Nous avons confirmé ces résultats chez l'homme lors d'une étude de phase II. Compte tenu des taux plus élevés de FRO dans les cellules tumorales par rapport aux cellules normales, plusieurs stratégies ont été proposées pour cibler sélectivement les tumeurs sans endommager les tissus sains avec des modulateurs du stress oxydant. Notre troisième axe de travail a eu pour objectif d'identifier et de valider de nouvelles molécules liées à la modulation des FRO. Cette étude a été réalisée dans le cadre d'un projet du programme Européen Pierre et Marie Curie (International Training Network 7-FP7-2007-1-1-ITN201114), au sein du réseau REDCAT. Cette collaboration a permis de concevoir et de synthétiser de nouveaux agents thérapeutiques, les organochalcogenes. Nous avons mis en évidence in vitro et in vivo le potentiel anti tumoral de cette nouvelle classe thérapeutique et principalement celui du composé LAB027. Le LAB027 présente une activité anti tumorale seul ou associé à l'oxaliplatine. L'ensemble de ces travaux nous a permis d'envisager l'évaluation précoce de l'efficacité du sorafenib par un marqueur du stress oxydant, les AOPP, de mettre en évidence l'effet anti-neurotoxique d'un mimétique de la superoxyde dismutase, le mangafodipir et d'identifier une nouvelle classe médicamenteuse en oncologie, les organochalcogènes. Ces optimisations thérapeutiques permettent d'envisager une meilleure prise en charge des malades traités par chimiothérapie afin d'optimiser l'efficacité des traitements utilisés en oncologie.
28

Sorafenib enhances pemetrexed-induced cytotoxicity through and autophagy-dependent mechanism in cancer cells

Mary, Bareford 03 August 2012 (has links)
Acquired cellular resistance to traditional chemotherapeutics is a common obstacle in the treatment of most cancer cell types. This resistance occurs as a result of changes in the underlying molecular mechanisms of disease progression. The development of novel chemotherapeutic approaches designed to enhance the efficacy of protypical anti-cancer drugs is important in order to overcome this issue. Such approaches will aid in understanding the biomolecular phenomena responsible for drug resistance and disease progression. Combining signaling pathway inhibitors has become an effective strategy for enhancing tumor cell death by targeting multiple pathways known to regulate cell survival. Pemetrexed, an FDA-approved anti-folate drug, targets thymidylate synthase (TS) and a secondary folate-dependent enzyme, 5’ aminoimidazole-carboximide ribonucleotide formyltransferase (AICART); both important for DNA synthesis. Studies performed by our collaborator demonstrated that TS inhibition causes intracellular accumulation of ZMP+ and activation of AMPK which is known to induce autophagy in mammalian cells. Previous studies from our lab and others showed that sorafenib, a multi-kinase inhibitor of Raf-1 and class III receptor tyrosine kinases, was able to induce a cytotoxic form of autophagy in a variety of tumor cell types. Combination treatment using pemetrexed and sorafenib in these cancer cells resulted in an enhancement of autophagy and cell lethality beyond that of individual drugs alone. Inhibition of autophagy suppressed the toxic interactions of these drugs in all cell types examined. Pemetrexed/sorafenib cotherapy also proved to be an effective treatment for triple negative breast cancer cells having advanced to a stage of estrogen independence. Fulvestrant-resistant MCF7 cells were more sensitive to the drug combination than parental, estrogen-dependent MCF7 cells. Breast cancer cells cotreated with pemetrexed and sorafenib exhibited enhanced MEK/ERK signaling, Src activation that was dependent on platelet-derived growth factor β (PDGFRβ) downregulation, elevated protein phosphatase 2A (PP2A) activity, and increased de novo ceramide synthesis. Studies using a mouse model of experimentally-induced breast cancer validated drug combination effectiveness through inhibition of tumor growth, while no deleterious effects on normal tissues were observed. The data presented demonstrates that pemetrexed/sorafenib cotreatment augments chemosensitivity in both in vitro and in vivo systems. Based upon these findings, a Phase I clinical trial involving pemetrexed and sorafenib in breast cancer patients with solid, recurrent tumors was begun in 2011. In conclusion, this work strongly supports a promising therapeutic utility for the pemetrexed/sorafenib combination in treatment of various cancer cell types.
29

IDENTIFICATION OF CLINICAL, LABORATORY AND GENETIC COVARIATES FOR PHARMACOKINETICS, EFFICACY AND TOXICITY OF SORAFENIB IN PATIENTS WITH SOLID TUMORS

JAIN, LOKESH 10 August 2009 (has links)
The goal of this research work was to understand the clinical-pharmacology based treatment approaches for sorafenib. Treatment with sorafenib is associated with high inter-patient variability in pharmacokinetic exposures, efficacy and toxicity. We explored the demographic, laboratory, clinical and pharmacogenetic factors to elucidate the sources of variability. In addition, we examined the impact of pharmacogenetic variation in VEGFR2, an important mediator of the VEGF pathway, on risk of prostate cancer. To support these investigations, (mainly single-dose) pharmacokinetic, pharmacogenetic, efficacy and toxicity information were collected from patients with solid tumors, enrolled in five phase I / II clinical trials at National Cancer Institute. Non-compartmental analysis-general linear modeling (NCA-GLM), population pharmacokinetic analysis and several correlative studies were performed to characterize the sources of variability in pharmacokinetics and response. The role of prostate specific antigen (PSA) and ex-vivo anti-angiogenic activity as efficacy markers was evaluated, respectively, for patients with prostate cancer treated with sorafenib and patients with solid tumors treated with combination of sorafenib and bevacizumab. Sweat concentrations of sorafenib were measured to study its association with development of hand-foot skin reaction (HFSR). Only body weight was a significant covariate for volume of distribution by population pharmacokinetic analysis, while BSA, albumin and UGT1A9*3 appeared to be significant by NCA-GLM. However, the contribution of these covariates in overall exposure variability was very small; hence, these were considered clinically irrelevant. The association of sorafenib exposure with efficacy in patients with prostate cancer, colorectal cancer and combined solid tumors were not significant; exposure-efficacy relationship for lung cancer patients requires further evaluation. Sorafenib exposures appeared to be associated with incidences of rash in single agent trials and with HFSR in trials involving treatment with sorafenib and bevacizumab combination. In-vitro cell-line experiments determined that prostate specific antigen (PSA) is not a suitable marker of efficacy in patients with prostate cancer treated with sorafenib. The ex-vivo anti-angiogenic activity, measured by rat-aortic ring assay using patient serum samples, appeared to be not associated with clinical response. Sorafenib concentration in sweat, upto ≥5 ng/mL, apparently was not associated with HFSR. The VEGFR2 H472Q polymorphism was associated with progression-free survival (PFS) (with an apparent heterozygous advantage for survival) and toxicities in patients treated with drugs against the VEGF pathway. Patients who developed hypertension and HFSR on bevacizumab and sorafenib therapy, respectively, appeared to have longer PFS. Therefore, these side effects should be effectively managed to avoid/delay the treatment discontinuation. The VEGFR2 H472Q and V297I genotype were not predictive of risk of prostate cancer in Caucasian subjects.

Page generated in 0.4388 seconds