• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 22
  • 5
  • 3
  • 2
  • 1
  • Tagged with
  • 37
  • 37
  • 10
  • 5
  • 5
  • 5
  • 4
  • 4
  • 4
  • 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.
11

Developmental toxicity of alkylating agents in differentiating cell cultures /

Seeley, Marguerite R. January 1996 (has links)
Thesis (Ph. D.)--University of Washington, 1996. / Vita. Includes bibliographical references (leaves [98]-111).
12

Adalimumab-Induced Acute Myelogenic Leukemia

Saba, Nakhle, Kosseifi, Semaan G., Charaf, Edris A., Hammad, Ahmad N. 01 December 2008 (has links)
Newer biological treatment strategies have been developed in the last decade with some promising outcomes. Their safety, however, has been questioned lately with multiple reports of increased risk for malignancies and infectious complications. These reports render their use suboptimal. We report a 44-year-old woman receiving adalimumab (Humira®) for advanced juvenile rheumatoid arthritis who then developed acute myelogenic leukemia.
13

Conformational studies on sperm whale metmyoglobin with alkylated histidyl residues

Clark, Julia Freeman January 1966 (has links)
This document only includes an excerpt of the corresponding thesis or dissertation. To request a digital scan of the full text, please contact the Ruth Lilly Medical Library's Interlibrary Loan Department (rlmlill@iu.edu).
14

Mécanisme d'action de nouveaux agents alkylants ciblant l'ADN ou les protéines / Mecanism of action of new alkylating agents which target dna or proteins

Lenglet, Gaëlle 15 December 2010 (has links)
Le S23906-1 est un dérivé diacétate de la benzo-[b]-acronycine. Sélectionné pour son potentiel cytotoxique in vitro et son activité anti-tumorale in vivo, ce composé très prometteur est entré en essai clinique en 2006-2007. Cet agent alkylant présente la particularité d’interagir avec l’azote en position 2 des guanines localisé dans le petit sillon de l’ADN et d’induire ensuite une ouverture locale de la double hélice. La conformation de ce composé est importante pour son activité puisque la présence du groupement réactif acétate en orientation S induit une plus forte déstabilisation de l’ADN par rapport à l’orientation R, ceci en corrélation avec une plus forte cytotoxicité et une meilleure activité anti-tumorale. Cependant le mécanisme moléculaire mis en place, pour conduire à la mort de la cellule tumorale reste peu connu. Afin de mieux comprendre le lien entre la déstabilisation de l’ADN et l’activité cytotoxique du S23906-1, nous avons cherché à déterminer la nature des protéines nucléaires qui reconnaissent spécifiquement cette lésion. Une approche protéomique par chromatographie d’affinité suivie d’une électrophorèse séparative et d’analyse en MALDI-TOF nous a permis d’identifier la GAPDH. Des expériences de retard en gel ont validé l’interaction de la GAPDH à l’adduit S23906-1/ADN simple ou double brin. La recherche d’une éventuelle séquence consensus de fixation à l’ADN de la GAPDH a été entreprise par méthode de CASTing. Si aucun consensus proprement-dit n’est clairement apparu, une prévalence de fixation pour les séquences riches en guanines a été observée. Des études de relation structure/fonction ont montré les adduits formés par les dérivés diacétylés ou benzo-[a]-diacétylés de l’acronycine déstabilisent également l’ADN et que la GAPDH se fixe également de manière efficace à ces adduits. Par comparaison à d’autres composés interagissant au niveau de l’azote 2 des guanines, nous n’avons observé aucune interaction de la GAPDH avec l’adduit ET-743/ADN (l’ET-743 stabilise l’ADN), alors que d’autres auteurs ont mis en évidence une interaction avec l’adduit QAD/ADN, où QAD est un dérivé de la Saframycine A, molécule structurellement proche de l’ET-743. Au niveau cellulaire, un traitement au composé QAD engendre une translocation de la GAPDH du cytoplasme vers le noyau. Nous n'avons pas retrouvé une telle translocation en utilisant le S23906-1. De plus, et contrairement à ce dérivé de la Saframycine A, nos cellules traitées avec un siRNA dirigé contre la GAPDH sont plus résistantes au S23906-1, suggérant un rôle anti-apoptotique de la GAPDH suite au traitement au S23906-1, contrairement à l’effet pro-apoptotique observé avec le dérivé QAD de la Saframycine A. En parallèle, nous nous sommes intéressés à de nouveaux agents cytotoxiques, dont des composés de série benzylamine bis-8-hydroxyquinoline. En utilisant des approches spectrométriques et biochimiques, nous avons montré que le composé principal JLK1486 ne cible pas l'ADN, mais réagit de manière covalente avec des groupements thiol comme celui du glutathion. Cependant, des études cellulaires de déplétion du glutathion endogène ont montré que le glutathion n’était pas la cible du composé JLK1486 mais participait à sa détoxification. Afin d’identifier la ou les cible(s) protéique(s) responsable(s) de l’effet cytotoxique du JLK1486, nous avons mis au point une approche dérivée de l’électrophorèse 2D-DIGE. / S23906-1 is a diacetate benzo-[b] acronycine derivative. Selected for its cytotoxic potential in vitro and its antitumoral activity in vivo, this very promising compound entered clinical trial in 2006-2007. This alkylating agent presents the peculiarity to interact with the nitrogen in position 2 (N2) of guanines, localized in the minor groove of the DNA, and to subsequently lead local opening of the double helix. The conformation of this compound is important for its activity. Indeed, the presence of the active acetate group in the S-orientation leads to a stronger destabilization of the DNA with regard to the R-orientation, in correlation with a stronger cytotoxicity and a better antitumoral activity of the S- rather than R-stereoisomer. However, the molecular mechanism leading to the death of treated tumoral cell lines remains poorly understood. To address the link between the destabilization of the DNA and the cytotoxic activity of the S23906-1, we tried to determine the nature of nuclear proteins that specifically recognize this lesion. A proteomic approach, using chromatographic affinity followed by SDS-PAGE electrophoresis and MALDI-TOF analysis, identified the GAPDH protein. EMSA experiments validated the interaction of GAPDH to the S23906-1/DNA adduct (as a single- or double-stranded DNA template). The search for a possible consensus DNA-binding sequence of GAPDH was performed using a CASTing method. Even though no consensus site was clearly identified, prevalence for G-rich and GT-rich sequences was evidenced. From structure/activity relationships studies, we showed that DNA alkylation using the diacetylated or benzo-[a]-diacetylated forms of acronycine also destabilize the DNA helix and that GAPDH also binds in an efficient manner to these adducts. By comparison to other compounds also interacting with the N2 group of guanines, no interaction of the GAPDH with the ET-743/ADN adduct was evidenced (ET-743 stabilizes the DNA), while other authors evidenced an interaction with DNA of QAD, a Saframycine A derivative and a molecule structurally related to ET-743. At the cellular level, cellular treatment with the QAD resulted in GAPDH translocation from the cytoplasm to the nucleus. We did not observe such a translocation using S23906-1. Furthermore cells treated with a siRNA directed to the GAPDH are more resistant to S23906-1, suggesting an anti-apoptotic role of GAPDH after S23906-1 treatment, by contrast with the pro-apoptotic effect observed in the literature with QAD . In parallel, we were interested in new cytotoxic agents, among which abis-8-hydroxyquinoline benzylamine series. Using spectrometric and biochemical approaches, we showed that the lead compound JLK1486 does not target DNA, but covalently reacts with thiol groups as that of the glutathione. However, cellular studies using depletion of the endogenous glutathione showed that the glutathione was not the target of JLK1486 but that it participated in its detoxification as it does for S23906-1. To identify the protein(s) target(s) responsible for the cytotoxic effect of JLK1486, we developed an approach derived from the 2D-DIGE electrophoresis which clearly evidenced proteins that are the cellular targets for JLK1486.
15

Screening of natural products and alkylating agents for antineoplastic activity

Kanyanda Stonard Sofiel Elisa January 2007 (has links)
<p><b><font face="TimesNewRomanPS-BoldMT"> <p align="left">Apoptosis is a process in which a cell programmes its own death. It is a highly organized physiological mechanism in which injured or damaged cells are destroyed. Apart from physiological stimuli however, exogenous factors can induce apoptosis. Many anti-cancer drugs work by activating apoptosis in cancer cells. Natural substances have been found to have the ability to induce apoptosis in various tumour cells and these substances have been used as templates for the construction of novel lead compounds in anticancer treatment. On the other hand, alkylating agents such as cisplatin, cis- [PtCl2 (NH3) 2]have been widely used as antineoplastic agents for a wide variety of cancers including testicular, ovarian, neck and head cancers, amongst others. However, the use of cisplatin as an anticancer agent is limited due to toxicity and resistance problems. <font face="TimesNewRomanPSMT">The aim of this present study was to screen the leaves of </font><i><font face="TimesNewRomanPS-ItalicMT">Rhus laevigata</font><font face="TimesNewRomanPSMT">, a South African indigenous plant, for the presence of pro-apoptotic and anti-proliferative natural compounds and also to screen newly synthesised palladium based complexes (15 and 57) and a platinum based complex (58) for their antineoplastic activities tested against a panel of cell lines.</font></i></p> </font><font face="TimesNewRomanPS-BoldMT"> <p align="left">&nbsp / </p> </font></b></p>
16

Screening of natural products and alkylating agents for antineoplastic activity

Kanyanda Stonard Sofiel Elisa January 2007 (has links)
<p><b><font face="TimesNewRomanPS-BoldMT"> <p align="left">Apoptosis is a process in which a cell programmes its own death. It is a highly organized physiological mechanism in which injured or damaged cells are destroyed. Apart from physiological stimuli however, exogenous factors can induce apoptosis. Many anti-cancer drugs work by activating apoptosis in cancer cells. Natural substances have been found to have the ability to induce apoptosis in various tumour cells and these substances have been used as templates for the construction of novel lead compounds in anticancer treatment. On the other hand, alkylating agents such as cisplatin, cis- [PtCl2 (NH3) 2]have been widely used as antineoplastic agents for a wide variety of cancers including testicular, ovarian, neck and head cancers, amongst others. However, the use of cisplatin as an anticancer agent is limited due to toxicity and resistance problems. <font face="TimesNewRomanPSMT">The aim of this present study was to screen the leaves of </font><i><font face="TimesNewRomanPS-ItalicMT">Rhus laevigata</font><font face="TimesNewRomanPSMT">, a South African indigenous plant, for the presence of pro-apoptotic and anti-proliferative natural compounds and also to screen newly synthesised palladium based complexes (15 and 57) and a platinum based complex (58) for their antineoplastic activities tested against a panel of cell lines.</font></i></p> </font><font face="TimesNewRomanPS-BoldMT"> <p align="left">&nbsp / </p> </font></b></p>
17

Kinetic studies of DNA interstrand crosslinking by nitrogen mustard and phenylalanine mustard. /

Kaminsky, Margaret I. January 1990 (has links)
Thesis (M.S.)--Rochester Institute of Technology, 1990. / Includes bibliographical references, (leaves 108-110).
18

Συγκριτική μελέτη της μεταλλαξιγόνου δράσης νέων αντικαρκινικών αλκυλιωτικών ενώσεων στο βακτήριο Salmonella typhimurium και στο μύκητα Aspergillus nidulans / Comparative study on the mutagenic action of new anticancer alkylating agents in Salmonella typhimurium and Aspergillus nidulans

Βουτσινάς, Γεράσιμος 09 June 2010 (has links)
- / -
19

Synthesis and Characterization of Oxazaphospholidinone Phosphorus Mustard Derivatives

Harper, Marc Alan 06 August 2020 (has links)
No description available.
20

Contribution to the study of the efficacy and the mechanism of action of the alkylating peptide prolyl-m-sarcolysyl-p-fluorophenylalanine (PSF)

Dierickx, Karen 05 November 2008 (has links)
The search for more effective treatment strategies in melanoma led to many new innovative approaches aiming at different molecular targets. Chemotherapy still remains the most effective treatment and many efforts are put in order to improve targeting and delivery of the chemotherapeutic agents. Among these, peptide conjugates of anticancer drugs were designed to increase stability, cell penetration, specificity and accumulation in cancer cells. We as well as others evaluated such a conjugate, termed PSF (L-prolyl-m-L-sarcolysyl-L-p-fluorophenylalanine-ethylester) in terms of its cytotoxicity in vitro and in vivo using a human melanoma tumor as a model, its stability, transport, and metabolisation. <p>By comparing the cytotoxicity of PSF and melphalan towards different cancer primary melanoma cell cultures, we noticed some interesting observations: PSF displayed the same toxicity pattern both in short (2h) and long term (24h) cell exposures whereas melphalan and m-sarcolysin needed long term exposure to reach the same toxicity. This could indicate that PSF very quickly penetrates the cells in accordance with what has been shown with red blood cells (RBCs). PSF has shown a much better and quicker penetration into the cells in vitro as compared to melphalan. <p>In this present work, the cytotoxic effect of PSF was further evaluated in vivo using a standardized nude mice tumor model bearing a human melanoma. First, the acute toxicity in rats and mice and the maximum tolerated dose were determined. After a dose-escalation study one dose was singled out and tested as a single dose and as a fractionated dose. PSF was able to reach the tumor site and a dose-response relationship was observed. The IP administration of fractionated doses of PSF had significantly better effect on tumor growth inhibition, regression and regrowth than single dose administration and this without any evidence for general toxicity monitored by animal weight loss. We also compared the efficacy of PSF to its parent drug m-sarcolysin, melphalan and cyclophosphamide and observed that PSF was much more active than both melphalan and m-sarcolysin at the same molar doses.<p>Body distribution of the 14C-labelled PSF revealed ratios of 2.4 and 1.5 compared to muscle tissue for the two melanoma tumors evaluated with no significant and stable accumulation in any vital organ. The amount of tracer was still high in the blood after 24 hours explaining the high radioactivity in the kidney and partly in the liver. Interestingly, the spleen had an unusual high radioactivity uptake reflecting the exceptional binding of the tracer to blood cells (BC), while the pancreas very high load was an indicator of protease-mediated specific delivery and strongly support our hypothesis elaborated on the basis of in vitro results. <p><p>Our in vitro data point to a particular mechanism of action of PSF based on the transport of PSF through the body by the rapid binding to blood cells and the delivery at the tumor site by the subsequent release of its active metabolites due to cleavage by tumor-associated proteases.<p>Concerning the binding of PSF to membranes and its transport the following observations were made: while PSF was stable in human plasma, it disappeared very quickly in whole blood along with the generation of a main metabolite: m-sarcolysin. The presence of BC membranes was required for both binding and generating the metabolites. Binding to natural or artificial membranes was achieved and only competition with melanoma cells or proteolytic enzymes such as dispase, led to the generation of active metabolites. The different metabolites were isolated using preparative LC and were then identified using Electrospray Ionisation Mass Spectrometry (ESI). Three metabolites, of which m-sarcolysin was the main one, were identified all bearing the chloroethyl alkylating group. <p>Enzymatic catalysis was further supported by a set of experiments where the enzymatic activity was non-specifically and specifically inhibited. In order to look at the effect of extracellular matrix proteases on PSF, three representatives of ECM proteases were incubated with PSF: collagenase A had no effect, but both dispase and trypsine were able to process PSF. <p>The following data indicate the higher processing of PSF in the presence of cells with a higher proteolytic activity and thus the delivery of the blood cell-bound PSF. When comparing BC with melanoma cells (MC), the latter showed a higher ability to bind and process PSF both by membrane-associated and most interestingly soluble proteases. A lot of families of enzymes are reported to be overexpressed by melanoma cells including: metalloproteases, cysteine cathepsins, serine proteases and aminopeptidases. All the melanoma cells and cell lines evaluated were able to generate PSF active metabolites. <p>To identify the families of enzymes expressed on the membrane of melanoma cells that might be involved in the mechanism of action of PSF, we performed 2D-gel electrophoresis on their membrane extracts. The 2D-gels experiments revealed the presence of proteins compatible with enzymes known to be important in melanoma and further work is needed to identify the individual enzymes involved by using mass spectrometry and Western blotting. <p><p>Both our in vitro and in vivo findings strongly suggest that not only melanoma tumor cells and tumor sites but other types of tumors as well may be targets for the toxic activity of PSF owing to their much higher load in proteolytic enzymes that are closely related to their invasive potential. The transport of PSF by the blood cells and the release of its metabolites at the tumor site result in a low amount of drug in its free soluble form within the blood and this may explain the relatively lower side-effects observed. PSF is thus expected to have a much better therapeutic index than conventional alkylating agents. This original mechanism of drug delivery may well be extended to other cancer and non-cancer drugs than alkylating agents.<p> / Doctorat en Sciences biomédicales et pharmaceutiques / info:eu-repo/semantics/nonPublished

Page generated in 0.31 seconds