• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 2
  • 1
  • Tagged with
  • 8
  • 8
  • 8
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 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

Design of Anticancer Agents Based on the Tetrahydroisoquinoline Alkaloids Containing a Pyrazino[2,1-b]quinazoline-3,6-diones structure

Yang, Ping-Syun 23 August 2010 (has links)
Tetrahydroisoquinoline alkaloids are a class of structurally complex natural products and a huge number of its natural product widely exist in nature which, from the discovery has been more than a century, it compounds with high anti-tumor activity, antibacterial and other physical activity, but also because of its special structure, with low oncentration of biological activity, but these alkaloids are not sold in the market mainly due to a less natural extraction, chemical synthesis method and multi-step, low yield. Therefore, we constructed a combination of tetrahydroisoquinoline alkaloids and the pyrazino [2,1-b] quinazoline-3,6-diones structure of the new compounds, which have the quinazolinone compounds which is the drug synthesis and drug activity on the bond, is also a kind of unique and widely used drug structure, and causes a lot of scientists and drug research interest and discussion, as we develop the motivation.
2

Phase 1 Study Of A Sequence Selective Minor Groove DNA Binding Agent (SJG-136) with Pharmacokinetic and Pharmacodynamic Measurements in Patients with Advanced Solid Tumours.

Hochhauser, Daniel, Meyer, Timothy, Spanswick, Victoria J., Wu, Jenny, Clingen, Peter H., Loadman, Paul M., Cobb, Margaret, Gumbrell, Lindsey, Begent, Richard H., Hartley, J.A., Jodrell, Duncan January 2009 (has links)
PURPOSE: This phase I dose-escalation study was undertaken to establish the maximum tolerated dose of the sequence-selective minor groove DNA binding agent SJG-136 in patients with advanced solid tumors. The study also investigated antitumor activity and provided pharmacokinetic and pharmacodynamic data. EXPERIMENTAL DESIGN: Sixteen patients were assigned sequentially to escalating doses of SJG-136 (15-240 microg/m(2)) given as a 10-minute i.v. infusion every 21 days. The dose was subsequently reduced in incremental steps to 45 microg/m(2) due to unexpected toxicity. RESULTS: The maximum tolerated dose of SJG-136 was 45 microg/m(2). The main drug-related adverse event was vascular leak syndrome (VLS) characterized by hypoalbuminemia, pleural effusions, ascites, and peripheral edema. Other unexpected adverse events included elevated liver function tests and fatigue. The VLS and liver toxicity had delayed onset and increased in severity with subsequent cycles. Disease stabilization was achieved for >6 weeks in 10 patients; in 2 patients this was maintained for >12 weeks. There was no evidence of DNA interstrand cross-linking in human blood lymphocytes with the use of the comet assay. Evidence of DNA interaction in lymphocytes and tumor cells was shown through a sensitive gamma-H2AX assay. SJG-136 had linear pharmacokinetics across the dose range tested. CONCLUSIONS: SJG-136 was associated with dose-limiting VLS and hepatotoxicity when administered by short injection every 21 days. DNA damage was noted, at all dose levels studied, in circulating lymphocytes. The etiology of the observed toxicities is unclear and is the subject of further preclinical research. Alternative clinical dosing strategies are being evaluated.
3

Ciblage de l'activité de l'interféron alpha : de la preuve de concept à l'activité biologique / Cell-specific targeting of interferon alpha activity

Paul, Franciane 24 November 2016 (has links)
Le ciblage de l’activité de l’IFNα est une stratégie développée afin d’augmenter l’index thérapeutique de cette cytokine, dont l’efficacité requiert de fortes doses au site d’action, responsables d’une toxicité systémique. Du fait de l’expression ubiquitaire de son récepteur, le ciblage de l’IFNα par immunocytokine est limité. En se basant sur le concept d'immunocytokine et utilisant un IFNα muté, peu actif, une efficacité de ciblage de 3 log a pu être obtenue, dans le système humain et murin, grâce au rétablissement de l'activité de l'IFNα sur les cellules ciblées. Ces IFNα ciblés sont doués d'une activité biologique, notamment antitumorale, dont la cible cellulaire reste à déterminer. Une stratégie inverse, en cours d'optimisation, permet d'inhiber l’activité des IFN-I spécifiquement sur les cellules ciblées. Cette double stratégie de ciblage de l’activité et de l’inhibition de l’IFN devrait permettre de déterminer les cibles des effets bénéfiques et néfastes des IFN-I, avec des applications thérapeutiques éventuelles. / Targeting IFN activity is a strategy developed to increase the therapeutic index of thiscytokine, whose efficiency requires high doses to site of action, responsible for systemictoxicity. Due to the ubiquitous expression of its receptor, the targeting efficiency of IFNbasedimmunocytokine is limited. Using a mutated IFNα, poorly active, a 3 log targetingefficiency was achieved, in the human and mouse system, by restoring the activity of IFNα ontargeted cells. The biological activity of these targeted IFNα include an antitumoral effect,the cellular target remains to be determined. A reverse strategy being optimized, can inhibitIFN-I activity specifically on targeted cells. This dual targeting strategy of the activity andinhibition of IFN should identify the targets of beneficial and deleterious effects of IFN-I,with possible therapeutic applications.
4

A mathematical model of doxorubicin penetration through multicellular layers,

Evans, C.J., Phillips, Roger M., Jones, P.F., Loadman, Paul, Sleeman, B.D., Twelves, Christopher J., Smye, S.W. January 2009 (has links)
no / Inadequate drug delivery to tumours is now recognised as a key factor that limits the efficacy of anticancer drugs. Extravasation and penetration of therapeutic agents through avascular tissue are critically important processes if sufficient drug is to be delivered to be therapeutic. The purpose of this study is to develop an in silico model that will simulate the transport of the clinically used cytotoxic drug doxorubicin across multicell layers (MCLs) in vitro. Three cell lines were employed: DLD1 (human colon carcinoma), MCF7 (human breast carcinoma) and NCI/ADR-Res (doxorubicin resistant and P-glycoprotein [Pgp] overexpressing ovarian cell line). Cells were cultured on transwell culture inserts to various thicknesses and doxorubicin at various concentrations (100 or 50 microM) was added to the top chamber. The concentration of drug appearing in the bottom chamber was determined as a function of time by HPLC-MS/MS. The rate of drug penetration was inversely proportional to the thickness of the MCL. The rate and extent of doxorubicin penetration was no different in the presence of NCI/ADR-Res cells expressing Pgp compared to MCF7 cells. A mathematical model based upon the premise that the transport of doxorubicin across cell membrane bilayers occurs by a passive "flip-flop" mechanism of the drug between two membrane leaflets was constructed. The mathematical model treats the transwell apparatus as a series of compartments and the MCL is treated as a series of cell layers, separated by small intercellular spaces. This model demonstrates good agreement between predicted and actual drug penetration in vitro and may be applied to the prediction of drug transport in vivo, potentially becoming a useful tool in the study of optimal chemotherapy regimes.
5

Examination of the distribution of the bioreductive drug AQ4N and its active metabolite AQ4 in solid tumours by imaging matrix-assisted laser desorption/ionisation mass spectrometry

Atkinson, S.J., Loadman, Paul, Sutton, Chris W., Patterson, Laurence H., Clench, M.R. January 2007 (has links)
no / AQ4N (banoxatrone) (1,4-bis-{[2-(dimethylamino-N-oxide)ethyl]amino}-5,8-dihydroxyanthracene-9,10-dione) is an example of a bioreductive prodrug in clinical development. In hypoxic cells AQ4N is reduced to the topoisomerase II inhibitor AQ4 (1,4-bis- {[2-(dimethylamino)ethyl]amino}-5,8-dihydroxyanthracene-9,10-dione). By inhibition of topoisomerase II within these hypoxic areas, AQ4N has been shown to sensitise tumours to existing chemo- and radiotherapy treatments. In this study the distribution of AQ4N and AQ4 in treated H460 human tumour xenografts has been examined by imaging matrix-assisted laser desorption/ionisation mass spectrometry. Images of the distribution of AQ4N and AQ4 have been produced that show little overlap. The distribution of ATP in the tumour xenografts was also studied as an endogenous marker of regions of hypoxia since concentrations of ATP are known to be decreased in these regions. The distribution of ATP was similar to that of AQ4N, i.e. in regions of abundant ATP there was no evidence of conversion of AQ4N into AQ4. This indicates that the cytotoxic metabolite AQ4 is confined to hypoxic regions of the tumour as intended.
6

Preclinical pharmacology of the pyrrolobenzodiazepine (PBD) monomer DRH-417 (NSC 709119).

Burger, A.M., Loadman, Paul, Thurston, D.E., Schultz, R., Fiebig, H.H., Bibby, Michael C. January 2007 (has links)
no / The pyrrolobenzodiazepine monomer DRH-417 is a member of the anthramycin group of anti-tumor antibiotics that bind covalently to the N2 of guanine within the minor groove of DNA. DRH-417 emerged from the EORTC-Drug Discovery Committee and NCI 60 cell line in vitro screening programs as a potent antiproliferative agent with differential sensitivity towards certain cancer types such as melanoma, breast and renal cell carcinoma (mean IC(50) = 3 nM). DRH-417 was therefore tested for in vivo activity. The maximum tolerated dose (MTD) was established as 0.5 mg/kg given i.p. Marked anti-tumor activity was seen in two human renal cell cancers, one breast cancer and a murine colon tumor model (p<0.01). A selective HPLC (LC/MS) analytical method was developed and plasma pharmacokinetics determined. At a dose of 0.5 mg kg(-1), the plasma AUC was 540 nM h (197.1 ng h ml(-1)) and the peak plasma concentration (171 nM [62.4 ng ml(-1)]) occurred at 30 min., reaching doses levels well above those needed for in vitro antiproliferative activity. Genomic profiling of in vivo sensitive tumors revealed that the latter have an activated insulin-like growth factor signaling pathway.
7

Expression, purification et cristallisation de l'aminopeptidase-N humaine (APN ou CD13) : évaluation in vitro et in vivo d'inhibiteurs sélectifs / Expression, purification and crystallization of aminopeptidase-N (APN or CD13) : In vitro and in vivo evaluation of selective inhibitors

Schmitt, Céline 18 September 2012 (has links)
L’Aminopeptidase-N (APN ou CD13) [EC.3.4.11.2] est une ectoenzyme homodimérique de nature glycoprotéique appartenant à la famille M1 des zinc-aminopeptidases. Elle est surexprimée à la surface des cellules endothéliales angiogéniques, ainsi que sur un certain nombre de cellules tumorales. Et il existe une corrélation étroite entre l’élévation de l’expression de l’APN, une activité enzymatique accrue et le pouvoir invasif de nombreux types de cellules tumorales. Des inhibiteurs puissants et sélectifs de l’APN, appartenant à la famille des composés de type amino-benzosubérone, ont été synthétisés au laboratoire. Ces composés ont été testés in vitro et in vivo, et il est apparu qu’ils présentaient une affinité variant du nano au picomolaire. En parallèle à ces essais, un nouveau projet a débuté il y a quelques années au laboratoire, visant à déterminer la structure tridimensionnelle de l’APN humaine. La connaissance de cette structure constitue un enjeu majeur car des co-cristallisations avec ces inhibiteurs permettraient de résoudre le mode de liaison de cette nouvelle famille de composés à l’APN. La difficulté de cette étude réside dans le fait que l’APN est une glycoprotéine membranaire particulièrement difficile à purifier à partir de tissus ; de plus, cette protéine étant ancrée dans la membrane de la cellule, sa cristallisation en est d’autant plus complexe. Plusieurs stratégies de clonage et de surexpression de l’APN humaine ont été envisagées, avec pour objectif final, l’obtention d’une protéine cristallisable, glycosylée ou non. / Aminopeptidase-N (APN or CD13) [EC.3.4.11.2] is a highly glycosylated type II membrane-bound ectoenzyme that belongs to the M1 family of zinc-dependent aminopeptidases. The members of this family have a thermolysin-like catalytic domain with the consensus HEXXH-X18-E zinc-binding sequence and an exopeptidase motif, GXMEN, in the active site. APN/CD13 is a widespread enzyme, located in many tissues, organs and cells, whose multiple functions dependent on its location. It is overexpressed on the endothelial cells of angiogenic, but not normal, vasculature, as well as on numerous tumor cells. As it was demonstrated that APN plays a critical role in tumor cell angiogenesis and metastasis, this protein was identified as a potential target for cancer therapy. In this context, highly potent and selective non-peptidic APN inhibitors, with Ki values ranging from micro to nanomolar, were previously designed and synthesized in the laboratory. In vitro and in vivo efficacy of these novel amino-benzosuberone derivatives was tested. In parallel to these works, a new project was started a few years ago which consists to solve the 3D structure of mammalian APN. Co-crystallizations with amino-benzosuberone derivatives would determine the binding mode of these novel inhibitors. Nevertheless solving the structure of a membrane protein like mammalian APN still remains a challenge. Therefore several cloning and expression strategies for human APN production were developed.
8

Chemical synthesis and biological evaluation of a NAD(P)H:quinone oxidoreductase-1-targeted tripartite quinone drug delivery system

Volpato, Milène, Abou-Zeid, N., Tanner, R.W., Glassbrook, L.T., Taylor, James P., Stratford, I.J., Loadman, Paul, Jaffar, M., Phillips, Roger M. January 2007 (has links)
No / NAD(P)H:quinone oxidoreductase-1 (NQO1) is a potential target for therapeutic intervention but attempts to exploit NQO1 using quinone-based bioreductive prodrugs have been largely compromised by toxicity to organs that inherently express high levels of NQO1. In an attempt to circumvent this problem, this study describes the development of a tripartite quinone-based drug delivery system, the ultimate objective of which is to release a targeted therapeutic agent following the reduction of a quinone "trigger" by NQO1. Molecular modeling of drug/NQO1 interactions were conducted prior to the synthesis of N-{4-[bis-(2-chloroethyl)-amino]-phenyl}-beta,beta,2,4,5-pentamethyl-3,6-dioxo-1,4-cyclohexadiene-1-propanamide (prodrug 1). Prodrug 1 is a good substrate for purified NQO1 (V(max) and K(m) values of 11.86 +/- 3.09 micromol/min/mg and 2.70 +/- 1.14 micromol/L, respectively) and liquid chromatography-mass spectrometry analysis of the metabolites generated showed that lactone 3 and aniline mustard 4 were generated in a time- and NQO1-dependent manner. Chemosensitivity studies showed that prodrug 1 is selectively toxic to cells that overexpress NQO1 under aerobic conditions, and comet assay analysis confirmed the presence of elevated interstrand cross-links in NQO1-rich compared with NQO1-deficient cells. Hypoxic sensitization (hypoxic cytotoxicity ratio = 15.8) was observed in T47D cells that overexpress cytochrome P450 reductase. In conclusion, the results of this study provide mechanistic proof of principle that a tripartite benzoquinone drug delivery system is enzymatically reduced to release an active therapeutic agent. Further development of this concept to fine-tune substrate specificity for specific reductases and/or the inclusion of alternative therapeutic agents is warranted.

Page generated in 0.0508 seconds