• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 10
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 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.
1

Therapeutic drugs in cancer and resistance

Martin, Aditi Pandya. January 1900 (has links)
Thesis (Ph.D)--Virginia Commonwealth University, 2009. / Prepared for: Dept. of Pharmacology and Toxicology. Title from title-page of electronic thesis. Bibliography: leaves 134-160.
2

Therapeutic Drugs in Cancer

Walker, Teneille 16 April 2009 (has links)
The first study examined the interaction between low doses of the multi-kinase inhibitor sorafenib and the histone deacetylase inhibitor vorinostat in colon cancer cells. Sorafenib and vorinostat synergized to kill HCT116 and SW480 cells. In SW480 cells, sorafenib+vorinostat toxicity correlated with CD95 activation and CD95-stimulated autophagy. Drug lethality in SW480 cells was blocked by knock down of CD95. In SW620 cells that are patient matched to SW480 cells, sorafenib+vorinostat toxicity was significantly reduced that correlated with a lack of CD95 activation and lower expression of ceramide synthase 6 (LASS6). Overexpression of LASS6 in SW620 cells enhanced drug-induced CD95 activation and tumor cell killing, whereas knock down of LASS6 in SW480 cells suppressed CD95 activation. In HCT116 cells, sorafenib+vorinostat did not increase CD95 plasma membrane levels, weakly induced caspase 8 association with CD95, and knock down of CD95 enhanced drug lethality. In HCT116 cells sorafenib+vorinostat treatment caused CD95-dependent autophagy that was a protective signal. Thus, treatment of tumor cells with sorafenib+vorinostat activates CD95 that promotes viability via autophagy or degrades survival via extrinsic or intrinsic pathways. Drug-induced activation of the de novo ceramide synthesis pathway plays a key role in CD95 activation. The second project explores the mechanism by which the combination of 17AAG, an hsp90 inhibitor, and PD184352, a MEK1/2 inhibitor alters survival in colon cancer cells. 17AAG and PD184352 synergized to kill HCT116 and SW480 cells. In HCT116 cells drug-exposure increased CD95 plasma membrane levels In SW620 cells, 17AAG and PD184352 toxicity was significantly reduced that correlated with a lack of CD95 activation and lower expression LASS6. Overexpression of LASS6 in SW620 cells enhanced drug-induced CD95 activation and tumor cell killing. In Mia Paca2 cells, a pancreatic cell line, inhibition of caspase 8 or overexpression of c-FLIP-s suppressed cell killing by PD184352 and 17AAG exposure. Drug lethality in Mia Paca2 cells was blocked by knock down of CD95. Additionally, overexpression of Bcl-xL or knockdown of caspase 9 decreased cell killing in 17AAG and PD184352 combination treatment. Thus, 17AAG+PD184352 exposure activates the extrinsic and intrinsic apoptotic pathways to kill Mia Paca2 cells. This document was created in Microsoft Word 2000.
3

Etude de la balance réactivation/apoptose des cellules B infectées par l' EBV suite au traitement par un HDACi, le vorinostat / Study of the balance reactivation / apoptosis of B cells infected with EBV following treatment with a HDACi, vorinostat

Al Mohamad, Hazar 10 May 2016 (has links)
Les inhibiteurs des histones désacétylase (HDACi) constituent une classe prometteuse de médicaments anticancéreux. Ils peuvent déclencher la voie apoptotique et sont proposés pour le traitement des désordres hématologiques. Cependant, les HDACi qui ciblent les HDAC de classe II, tel que le vorinostat, sont également des agents réactivateurs potentiels de l’EBV, un virus qui infecte de manière latente plus de 90% de la population adulte dans le monde et est associée à de nombreux lymphomes de type B. L’étude de la commutation entre le cycle latent et le cycle lytique de l’EBV est essentielle pour appréhender l’impact des HDACi lors du!traitement des lymphomes B associés à l’EBV (risque du relargage de virions en grande quantité lors des traitements chimio thérapeutiques).Notre étude a porté sur l'effet de vorinostat (25 μM pendant 48h) sur des cellules tumorales B infectées par l’EBV : trois lignées de lymphomes de burkitt (BL2B95.8, BL41B95.8 et P3HR1), trois lignées lymphoblastoides (1602, PRI et RUD) et la lignée B95.8 de marmouset en cycle lytique de l’EBV (contrôle positif). Nous avons mis en évidence que le vorinostat peut induire la réactivation de l’EBV (P3HR1 et B95.8) ou à l’apoptose (BL41B95.8, BL2B95.8, 1602, PRI et RUD) avec une inhibition mutuelle de ces deux processus. Au niveau moléculaire, nous avons pu montrer que le vorinostat active constitutivement et simultanément le facteur de transcription initiateur de la réactivation MEF2D (par déphosphorylation) et la MAP kinase pro-apototique p38 (par phosphorylation) suite à la diminution de l’expression de la MAP kinase phosphatase (MPK1) dont p38 est un substrat. Le pré-traitement avec un inhibiteur de p38 (SB203580) a mis en évidence que cette MAP kinase est à la fois impliquée dans les processus de réactivation de l’EBV et d’’apoptose. Cependant, les lignées cellulaires pour lesquelles l'activation de p38 augmente fortement lors du traitement par le vorinostat, entrent directement en apoptose, sans qu’il puisse y avoir réactivation de l’EBV.Nos résultats suggèrent que le niveau d’activation de la MAP kinase p38 permet de réguler la balance réactivation/apoptose des cellules B infectées par l’EBV lorsqu’elles sont soumises à un agent inducteur de la réactivation, en particulier dans le cas de cellules de lymphomes B traitées par le vorinostat. Ils posent la question de l’utilisation des HDACi lors du traitement des lymphomes associés à l'EBV, avec le risque d’une réactivation virale selon le niveau d’activation intracellulaire de p38 et la nécessité d’utiliser simultanément un anti-viral tel que le ganciclovir. / Histone deacetylase inhibitors (HDAC) are a promising class of anticancer drugs. They can trigger the apoptotic pathway and are available for treatment of blood disorders. However, the HDACi that target HDAC class II, as vorinostat, also are potential reactivators agents of EBV, a virus that infects latently over 90% of the adult population worldwide and is associated with many type B lymphomas the study of switching between the latent cycle and the lytic cycle of EBV is essential to understand the impact of HDACi when treating B-cell lymphoma associated with EBV (salting risk virions in large quantities during the chemotherapeutic treatment).Our study focused on the effect of vorinostat (25 .mu.M for 48) on tumor B cells infected with EBV: three lines of Burkitt lymphoma (BL2B95.8, BL41B95.8 and P3HR1), three lymphoblastoid cell lines (1602 PRI and RUD) and B95.8 marmoset line in the lytic cycle of the EBV (positive control). We have demonstrated that vorinostat can induce EBV reactivation (P3HR1 and B95.8) or apoptosis (BL41B95.8, BL2B95.8, 1602, PRI and RUD) with a mutual inhibition of these two process. At the molecular level, we have shown that the active vorinostat constitutively and simultaneously initiating transcription factor reactivation MEF2D (by dephosphorylation) and MAP kinase p38 pro-apototique (phosphorylation) following the reduction in the expression of the MAP kinase phosphatase (MPK1) p38 which is a substrate. The pre-treatment with a p38 inhibitor (SB203580) showed that MAP kinase is both involved in the process of EBV reactivation and apoptosis. However, cell lines where p38 activation greatly increases during treatment with vorinostat, come directly into apoptosis, without there may EBV reactivation.Our results suggest that the level of activation of p38 MAP kinase helps regulate the balance reactivation / apoptosis of B cell EBV infected when exposed to an inducing agent for the reactivation, in particular in the case of cells B lymphoma treated with vorinostat. They raise the question of the use of HDACi in the treatment of lymphomas associated with EBV, with the risk of viral reactivation by level of intracellular activation of p38 and the need to simultaneously use an antiviral such as ganciclovir.
4

Therapeutic drugs in cancer and resistance.

Martin, Aditi Pandya 02 April 2009 (has links)
We investigated the mechanism of toxicity and resistance development of small molecule tyrosine kinase inhibitor lapatinib in HCT 116 colon cancer cells. Lapatinib mediated cell death in HCT 116 cells was caspase independent and involved cytosolic release of apoptosis inducing factor. Treatment of HCT 116 cells with 10µM Lapatinib lead to the outgrowth of lapatinib resistant HCT 116 cells. Our studies show that alterations in the expression and activation of Bcl-2 family proteins allow lapatinib resistant HCT 116 cells to resist cytotoxic effects of lapatinib as well as of other commonly used chemotherapeutic agents. In hepatoma and pancreatic cancer cells, the effects of combining multi-kinase inhibitor sorafenib with histone deacetylase inhibitors (HDACIs) namely, vorinostat and sodium valproate were investigated. It was found that sorafenib synergizes with HDACIs resulting in enhanced cell death compared to death induced by the drugs individually. The mechanism of action of sorafenib and vorinostat combination treatment as well as sorafenib and sodium valproate combined treatment was shown to involve activation of the CD95 death receptor pathway. Alterations in the CD95 pathway can render cancer cells resistant to chemotherapeutic agents. Hence, we combined sorafenib+sodium valproate with a BH-3 domain mimetic named obatoclax (GX-15-070) which resulted in enhanced toxicity to cancer cells. More importantly, knock-down of CD95 (to mimic non-functional CD95 pathway) reduced cell death induced by sorafenib+sodium valproate combined but failed to protect cells from cell death induced by sorafenib+sodium valproate+obatoclax combined. This suggests that combining sorafenib+HDACI with obatoclax may not only enhance toxicity to cancer cells but may also reduce chances of resistance development via alterations in the CD95 pathway. These studies enhance our knowledge of existing treatment strategies for cancer as well as throw light on how current approaches can be improved in order to better diagnose and treat cancer. Understanding mechanisms of drug action as well as resistance development will allow us to combine existing therapies effectively in order best target cancer cells as well as provide us with information that can help us design new cancer treatment strategies.
5

The influence of p21WAF1 on cell death pathways in acute lymphoblastic leukaemia

Davies, Carwyn, Children's Cancer Institute Australia for Medical Research, UNSW January 2009 (has links)
The p53 protein is a primary mediator of apoptosis and growth arrest after exposure to DNA-damaging agents. Previous work has categorised a wild type p53 gene in the majority of childhood acute lymphoblastic leukaemia (ALL) cases, in which instance the p53 protein functions as a modulator of chemotherapy-induced cell death. In contrast, certain p53-induced proteins, such as p21WAF1, can act in an anti-apoptotic manner, and bestow resistance to chemotherapy. Previous studies of the p53 pathway in ALL have utilised cell lines and primary material. In this study a model of ALL was utilised that had previously been developed from a heterogeneous panel of patient biopsies established as xenografts in immune-deficient mice, and are adaptable for short term in vitro culture. A wild-type p53 protein response to etoposide and nutlin-3 exposure was a feature of the whole ALL xenograft panel, irrespective of clinical characteristics and disease biology. While a range of p53 target genes were induced in B-cell precursor (BCP)-ALL and T-ALL xenografts after etoposide exposure, there was negligible induction of p21WAF1 in T- ALL samples. Further work with the histone deacetylase inhibitor vorinostat facilitated p53-independent induction of p21WAF1 in BCP-ALL samples, yet failed to induce p21WAF1 in T- ALL. An association was observed between reduced p21WAF1 expression in the T-ALL samples and decreased histone H3 acetylation in the p21WAF1 promoter together with increased cytosine methylation in the first exon/intron of the p21WAF1 gene. These results suggest that p21WAF1 in T-ALL cells is subject to epigenetic modifications that cause transcriptional silencing. Defective induction of p21WAF1 in T-ALL xenografts was associated with increased sensitivity to the death-inducing effects of drugs, phosphatidylserine (PS) externalisation and caspase-3/-7 activity after drug exposure, indicating that p21WAF1 may exert an anti-apoptotic activity. As proof of principle, p21WAF1 was silenced in Nalm-6 cells by micro-RNA transduction and these cells exhibited increased sensitivity and rapid PS externalisation after drug exposure. A combination of a p21WAF1 inhibitory agent and vorinostat gave some pharmacological evidence to suggest that p21WAF1 inhibition could enhance drug efficacy. Overall, these investigations provide insight into the epigenetic regulation of p21WAF1 and demonstrate an anti-apoptotic role for p21WAF1 in childhood ALL cells.
6

The influence of p21WAF1 on cell death pathways in acute lymphoblastic leukaemia

Davies, Carwyn, Children's Cancer Institute Australia for Medical Research, UNSW January 2009 (has links)
The p53 protein is a primary mediator of apoptosis and growth arrest after exposure to DNA-damaging agents. Previous work has categorised a wild type p53 gene in the majority of childhood acute lymphoblastic leukaemia (ALL) cases, in which instance the p53 protein functions as a modulator of chemotherapy-induced cell death. In contrast, certain p53-induced proteins, such as p21WAF1, can act in an anti-apoptotic manner, and bestow resistance to chemotherapy. Previous studies of the p53 pathway in ALL have utilised cell lines and primary material. In this study a model of ALL was utilised that had previously been developed from a heterogeneous panel of patient biopsies established as xenografts in immune-deficient mice, and are adaptable for short term in vitro culture. A wild-type p53 protein response to etoposide and nutlin-3 exposure was a feature of the whole ALL xenograft panel, irrespective of clinical characteristics and disease biology. While a range of p53 target genes were induced in B-cell precursor (BCP)-ALL and T-ALL xenografts after etoposide exposure, there was negligible induction of p21WAF1 in T- ALL samples. Further work with the histone deacetylase inhibitor vorinostat facilitated p53-independent induction of p21WAF1 in BCP-ALL samples, yet failed to induce p21WAF1 in T- ALL. An association was observed between reduced p21WAF1 expression in the T-ALL samples and decreased histone H3 acetylation in the p21WAF1 promoter together with increased cytosine methylation in the first exon/intron of the p21WAF1 gene. These results suggest that p21WAF1 in T-ALL cells is subject to epigenetic modifications that cause transcriptional silencing. Defective induction of p21WAF1 in T-ALL xenografts was associated with increased sensitivity to the death-inducing effects of drugs, phosphatidylserine (PS) externalisation and caspase-3/-7 activity after drug exposure, indicating that p21WAF1 may exert an anti-apoptotic activity. As proof of principle, p21WAF1 was silenced in Nalm-6 cells by micro-RNA transduction and these cells exhibited increased sensitivity and rapid PS externalisation after drug exposure. A combination of a p21WAF1 inhibitory agent and vorinostat gave some pharmacological evidence to suggest that p21WAF1 inhibition could enhance drug efficacy. Overall, these investigations provide insight into the epigenetic regulation of p21WAF1 and demonstrate an anti-apoptotic role for p21WAF1 in childhood ALL cells.
7

NF-kappaB-dependent regulation of the diagnostic marker CD10 and role of BCL-2 activity in histone deacetylase inhibitor-induced apoptosis in human B-lymphoma cell lines

Thompson, Ryan C. 22 January 2016 (has links)
Diffuse large B-cell lymphoma (DLBCL) is a genetically heterogeneous disease with multiple distinct molecular subtypes. Increased NF-κB activity and expression of the microRNA miR-155 (product of the BIC gene) are associated with one subtype, called the activated B-cell (ABC) subtype. It is shown here that induction of NF-κB activity leads to increased miR-155 expression, the levels of miR-155 in a panel of B-lymphoma cell lines correlate with increased NF-κB activity, and the NF-κB p50/p65 heterodimer binds to a specific DNA site in the BIC promoter. Also described is a regulatory network wherein NF-κB-dependent up-regulation of miR-155 leads to reduced PU.1 transcription factor expression and consequently reduced PU.1-driven expression of B-lymphoma marker CD10 in the human B-lymphoma cell line BJAB. Genetic variation in DLBCL can be used to explain the response of individual patients to chemotherapy. One cancer therapeutic approach currently in clinical trials uses histone deacetylase inhibitors (HDACi's) as a monotherapy or in combination with other vi agents. It is shown here that two pan-HDACi's, trichostatin A and vorinostat, induce apoptosis in seven of eight human DLBCL cell lines. Ectopic over-expression of antiapoptotic proteins BCL-2 and BCL-XL or the pro-apoptotic protein BIM in select DLBCL cell lines can confer further resistance or sensitivity, respectively, to HDACi treatment. Additionally, the BCL-2 family antagonist ABT-737 can increase the sensitivity of several DLBCL cell lines to vorinostat-induced apoptosis, including the HDACi-resistant SUDHL6 cell line. Moreover, one vorinostat-resistant variant of the HDACi-sensitive cell line SUDHL4 has increased expression of anti-apoptotic proteins BCL-XL and MCL-1 and decreased sensitivity to ABT-737, and a second such variant cell line has increased expression of anti-apoptotic protein MCL-1. These results suggest that the balance of anti- to pro-apoptotic BCL-2 family protein expression is important in determining the sensitivity of DLBCL cell lines to HDACi-induced apoptosis. Thus, the sensitivity of DLBCL cell lines to treatment with HDACi's appears to depend on the complex regulation of BCL-2 family members, suggesting that the response of a subset of DLBCL patients to HDACi treatment may benefit from co-treatment with BCL-2 antagonists.
8

Epigenetic Biomarker and Therapeutic Intervention for Dementia

Islam, Md Rezaul 30 October 2019 (has links)
No description available.
9

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.
10

Synthèse, caractérisation et évaluation des effets biologiques des hybrides "FcTAM-SAHA" et composés dérivés

Cázares Marinero, José De Jesús 02 September 2013 (has links) (PDF)
Dans le cadre de la poursuite de l'exploration des effets anticancéreux des complexes ferrocéniques du tamoxifène, nous avons préparé une série de complexes hybrides résultant de la substitution de la chaîne latérale du ferrocifène (FcTAM) par celle du SAHA, un inhibiteur des histones-déacétylases (HDACi) utilisé dans le traitement de certains lymphomes. A partir de la structure hybride " FcTAM-SAHA ", cinq types de modifications ont été réalisées. Il s'agit [1] de la modification de la fonction acide hydroxamique (CONHOH) en fonction amide primaire (CONH2) ou acide carboxylique (COOH), [2] de la variation de la longueur de la chaîne latérale, [3] de la présence ou non d'un substituant sur le cycle aromatique, [4] de la substitution du métallocène et [5] du remplacement du groupement ferrocényle par un phényle. Les effets antiprolifératifs des nouveaux complexes ont été étudiés sur deux lignées de cancer du sein, MDA-MB-231 (lignée hormono-indépendante et triple-négative) et MCF-7 (lignée hormono-dépendante). Un effet synergique de la toxicité est observé en série FcTAM-SAHA sur les deux lignées cellulaires. De façon un peu inattendue, nous avons trouvé que l'effet antiprolifératif des hybrides phénoliques (type FcOHTAM-SAHA) et des hybrides ferrocénophaniques (type FnTAM-SAHA) était comparable voire légèrement inférieur à celui des complexes non hydroxylés (type FcTAM-SAHA). Cette observation semble indiquer que les propriétés redox des molécules ne s'expriment pas de façon identique pour les précurseurs et les hybrides. Dans tous les cas, les complexes porteurs d'une fonctionnalité chimique acide hydroxamique ou amide primaire sont plus cytotoxiques que les acides carboxyliques et ces effets sont peu influencés par la longueur de la chaîne latérale. Enfin, dans tous les cas, les complexes ferrocéniques ont des effets antiprolifératifs supérieurs à leurs analogues organiques. Cette différence souligne l'importance de la présence du motif ferrocénique et de ses propriétés redox uniques.

Page generated in 0.2916 seconds