Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Oncol Rep ; 33(1): 471-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25371289

ABSTRACT

The present in vitro study aimed to assess the effects of combining the mTOR inhibitor RAD001 and temozolomide (TMZ) together with irradiation by either low-linear energy transfer (LET) radiation (γ-rays) or high-LET radiation (fast neutrons) on the growth and cell survival of the human glioblastoma cell line U-87. We observed a strong decrease in cell proliferation along with a concomitant increase in cell death as a function of the radiation dose. As expected, high-LET radiation was more effective and induced more sustained damage to DNA than low-LET radiation. While RAD001 in association with TMZ induced autophagic cell death, additional combination with either type of radiation did not further increase its occurrence. On the contrary, apoptosis remained at a low level in all experimental groups.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Apoptosis/drug effects , Apoptosis/radiation effects , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dose-Response Relationship, Radiation , Everolimus , Gamma Rays , Glioblastoma/pathology , Histones/metabolism , Humans , Linear Energy Transfer , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Temozolomide
2.
Anticancer Res ; 33(5): 1845-51, 2013 May.
Article in English | MEDLINE | ID: mdl-23645729

ABSTRACT

We have studied the consequences of the combination of the mammalian target of rapamycin (mTOR) inhibitor RAD001 and temozolomide on the growth and cell death of the glioblastoma cell line U-87 in vitro. A progressive decrease of cell proliferation was recorded with increasing concentrations of temozolomide, which was markedly reinforced and prolonged by the addition of RAD001. While this combination treatment resulted in only a low level of apoptosis, it led to a pronounced enhancement of autophagic cell death. When combined with γ-ray irradiation, a significant reinforcement of the overall cytotoxicity was obtained, suggesting the efficacy of such a multipronged approach for the treatment of glioblastoma. RAD001 strongly contributes to the reinforcement of temozolomide-induced autophagy, which appears to represent a major form of cell death in glioblastoma. The association of such combined chemotherapies with radiotherapy could be useful for the management of these hard-to-treat malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Autophagy , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Glioblastoma/pathology , Apoptosis/radiation effects , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Cell Proliferation/radiation effects , Cesium Radioisotopes , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Synergism , Everolimus , Flow Cytometry , Gamma Rays , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Temozolomide , Tumor Cells, Cultured
3.
Expert Opin Ther Pat ; 22(9): 1053-79, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22860892

ABSTRACT

INTRODUCTION: Autophagy is an intracellular process of self-digestion involving the lysosomal degradation of cytoplasmic organelles and macromolecules. It occurs at low basal levels to perform housekeeping functions and is dramatically augmented upon nutrient depletion or exposure to other stresses, thus maintaining cellular homeostasis and energy balance and providing cytoprotective responses to adverse conditions. Mounting evidence that autophagy malfunction contributes to the pathogenesis of diverse human diseases has stimulated efforts to identify pharmacological agents that modulate autophagy in potentially beneficial ways. Here, we review the progresses accomplished toward this goal in recent years, as reflected by the patent literature. AREAS COVERED: Patent applications published from 2008 to mid-2012 that pertain to the pharmacological modulation of autophagy are reviewed and their potential therapeutic utilities are discussed. EXPERT OPINION: Of 40 patents related to autophagy, 21 claim novel enhancers or inhibitors of autophagy. One of the most promising applications of these compounds concerns cancer therapy, a few of them being already considered for clinical evaluation. Further work is, however, needed to identify compounds that target unique molecular effectors/regulators of autophagy to selectively modulate its various stages in different tissues and to design therapeutic interventions applicable to a broad variety of dysfunctional autophagy-associated disorders.


Subject(s)
Autophagy/drug effects , Drug Delivery Systems , Drug Design , Animals , Homeostasis , Humans , Lysosomes/metabolism , Organelles/metabolism , Patents as Topic
4.
Int J Oncol ; 41(4): 1381-6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22895785

ABSTRACT

Treatment of hepatocellular carcinoma (HCC) is a major concern for physicians as its response to chemotherapy and radiotherapy remains generally poor, due, in part, to intrinsic resistance to either form of treatment. We previously reported that an irradiation with fast neutrons, which are high-linear energy transfer (LET) particles, massively induced autophagic cell death in the human HCC SK-Hep1 cell line. In the present study, we tested the capacity of the mammalian target of rapamycin (mTOR) inhibitor RAD001 to augment the cytotoxicity of low and high-LET radiation in these cells. As mTOR is a key component in a series of pathways involved in tumor growth and development, it represents a potential molecular target for cancer treatment. Results indicate that RAD001, at clinically relevant nanomolar concentrations, enhances the efficacy of both high- and low-LET radiation in SK-Hep1 cells, and that the induction of autophagy may account for this effect. However, fast neutrons were found to be more efficient at reducing tumor cell growth than low-LET radiation.


Subject(s)
Carcinoma, Hepatocellular/radiotherapy , Cell Proliferation/radiation effects , Liver Neoplasms/radiotherapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Autophagy/drug effects , Autophagy/radiation effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Cell Proliferation/drug effects , Everolimus , Growth Inhibitors/pharmacology , Humans , Linear Energy Transfer , Liver Neoplasms/pathology , Neutrons , Radiation-Sensitizing Agents/pharmacology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
5.
Curr Cancer Drug Targets ; 12(8): 899-924, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22831276

ABSTRACT

Radiotherapy (RT) allows for tumor control through the cytotoxic action of ionizing radiation (IR). Although modern technologies permit precise IR delivery to the tumor mass while minimizing exposure of surrounding healthy tissues, the efficacy of RT remains limited by the intrinsic or acquired radioresistance of many tumors. There is thus an ongoing search for agents that augment the sensitivity of tumor cells to IR cytotoxicity, with recent interest in targeting components of signaling pathways involved in tumor growth and radioresistance. Here, we review the evidence suggesting that disabling one of these components, the mechanistic target of rapamycin (mTOR) kinase, may enhance RT efficacy. This molecule constitutes the catalytic subunit of the mTORC1 and mTORC2 protein complexes, which regulate cell growth and other processes implicated in tumorigenesis. Much work has focused on mTORC1 because it is selectively blocked by the microbial product rapamycin and its analogs (collectively designated rapamycins) that are approved for cancer treatment, and is frequently hyperactivated in malignant cells. In several, but not all human cancer cell lines, rapamycins increased IR cytotoxicity in vitro, apparently through multiple mechanisms, including the promotion of autophagic cell death. Rapamycins also potentiated fractionated RT in tumor xenograft models, in part by suppressing tumor angiogenesis. Synthetic kinase inhibitors that simultaneously target PI3K and both mTOR complexes also enhanced RT in vitro and in vivo, but with greater efficiency than rapamycins. These encouraging data have led to early clinical trials of rapamycins and catalytic mTOR inhibitors combined with RT in various cancers.


Subject(s)
Neoplasms/metabolism , Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy/drug effects , Cell Line, Tumor , DNA Repair/drug effects , Humans , Mice , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , Xenograft Model Antitumor Assays
6.
J Chem Inf Model ; 51(12): 3050-9, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22077795

ABSTRACT

Antioxidative activity expressed as protection of thymidine has been investigated for a set of 30 pulvinic acid derivatives. A combination of in vitro testing and in silico modeling was used for synthesis of new potential antioxidants. Experimental data obtained from a primary screening test based on oxidation under Fenton conditions and by an UV exposure followed by back-titration of the amount of thymidine remaining intact have been used to develop a computer model for prediction of antioxidant activity. Structural descriptors of 30 compounds tested for their thymidine protection activity were calculated in order to define the structure-property relationship and to construct predictive models. Due to the potential nonlinearity, the counter-propagation artificial neural networks were assessed for modeling of the antioxidant activity of these compounds. The optimized model was challenged with 80 new molecules not present in the initial training set. The compounds with the highest predicted antioxidant activity were considered for synthesis. Among the predicted structures, some coumarine derivatives appeared to be especially interesting. One of them was synthesized and tested on in vitro assays and showed some antioxidant and radioprotective activities, which turned out as a promising lead toward more potent antioxidants.


Subject(s)
Antioxidants/chemistry , Antioxidants/pharmacology , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Drug Design , Lactones/chemistry , Lactones/pharmacology , Thymidine/metabolism , Cell Line , Humans , Hydrogen Peroxide/metabolism , Iron/metabolism , Neural Networks, Computer , Oxidation-Reduction/drug effects , Quantitative Structure-Activity Relationship , Ultraviolet Rays
7.
In Vivo ; 25(1): 1-9, 2011.
Article in English | MEDLINE | ID: mdl-21282728

ABSTRACT

Hepatocellular carcinoma (HCC) represents the sixth most common cancer worldwide and a major health problem since the choice of treatment is limited due to chemo- and radio-resistance. It was previously reported that high linear energy transfer (LET) radiation induced massive autophagic cell death in the human HCC SK-Hep1 cell line in vitro. This study analyzed the effects of high-LET radiation on the same HCC tumor model, orthotopically transplanted into nude mice. For this purpose, after surgical xenograft in the liver, animals were irradiated with fast neutrons and cell death occurring in the tumors was assessed with various techniques, including electron microscopy and probe-based confocal laser endomicroscopy. Results indicate that considerable autophagy and only limited apoptosis took place in the tumor xenografts after high-LET irradiation. These data confirm the previous in vitro results, suggesting that autophagy may act as a predominant mode of cell death in the efficacy of high-LET radiation.


Subject(s)
Apoptosis/radiation effects , Autophagy/radiation effects , Carcinoma, Hepatocellular/radiotherapy , Linear Energy Transfer , Liver Neoplasms/radiotherapy , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Fast Neutrons , Humans , Liver Neoplasms/pathology , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Electron , Xenograft Model Antitumor Assays
8.
ChemMedChem ; 6(3): 561-9, 2011 Mar 07.
Article in English | MEDLINE | ID: mdl-21246736

ABSTRACT

A high-throughput screening method has highlighted the marked antioxidant activity of some pulvinic acid derivatives (PADs) towards oxidation of thymidine, under γ and UV irradiation, and Fenton-like conditions. Here, we report the synthesis of a series of new hydrophilic PADs and the evaluation of their radioprotective efficacy in cell culture. Using a cell-based fluorescent assay, we show that some of these compounds have a pronounced ability to prevent cell death caused by radiation and to allow the subsequent resumption of proliferation. Thus, PADs may be considered as a novel class of radioprotective agents.


Subject(s)
Carboxylic Acids/chemistry , Lactones/chemistry , Radiation-Protective Agents/chemical synthesis , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/chemical synthesis , 4-Butyrolactone/chemistry , 4-Butyrolactone/pharmacology , Carboxylic Acids/chemical synthesis , Carboxylic Acids/pharmacology , Cell Line , High-Throughput Screening Assays , Humans , Lactones/chemical synthesis , Lactones/pharmacology , Phenylacetates/chemical synthesis , Phenylacetates/chemistry , Phenylacetates/pharmacology , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/pharmacology , Ultraviolet Rays
9.
Expert Opin Ther Pat ; 20(5): 715-23, 2010 May.
Article in English | MEDLINE | ID: mdl-20192905

ABSTRACT

Erythropoietin (EPO) is a cytokine with erythropoietic and tissue protective activities. Its action as a tissue protective agent requires, however, high dosage that results in limiting side effects associated with abnormally augmented erythropoiesis. Elimination of the erythropoietic activity of EPO while preserving its tissue protective properties was nevertheless achieved in carbamoylated EPO (CEPO), whose therapeutic activity and apparent safety were documented in experimental models of nervous, heart, kidney and other tissue damage, justifying ongoing clinical trials. Here, we review patent application WO2009094172 by Araim Pharmaceuticals, which describes novel EPO-derived peptides having tissue protective but no erythropoietic activity. The preferred peptide, UEQLERALNSS, which mimics the external 3D structure of the helix B of EPO, was shown to exhibit the same spectrum of tissue protective activity as CEPO in several in vivo models. In addition, it could reduce radiation-induced mortality when administered 24 h after irradiation in mice, suggesting its possible utility in emergency situations after mass irradiation casualties. Owing to their low manufacturing cost, high stability and low immunogenicity, such peptides might well offer a superior alternative to CEPO for therapeutic tissue protection in human pathologies and are likely to provide valuable probes to study the molecular mechanisms of EPO-mediated cytoprotection.


Subject(s)
Erythropoietin/analogs & derivatives , Peptides/pharmacology , Protective Agents/pharmacology , Animals , Cytoprotection/drug effects , Disease Models, Animal , Erythropoietin/adverse effects , Erythropoietin/pharmacology , Humans , Mice , Patents as Topic , Peptides/adverse effects , Peptides/chemistry , Protective Agents/adverse effects , Protective Agents/chemistry , Protein Stability , Radiation Injuries/drug therapy , Wounds and Injuries/drug therapy , Wounds and Injuries/etiology , Wounds and Injuries/physiopathology
10.
Anticancer Res ; 30(2): 303-10, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20332433

ABSTRACT

The aim of the present study was to determine the cytotoxic consequences of high-linear energy transfer (LET) irradiation in the presence of oxaliplatin on hepatocellular carcinoma (HCC) cells in vitro. We attempted to correlate the induction of apoptosis and autophagy with the formation of DNA double-strand breaks (DSBs). SK-Hep1 cells were irradiated by 65 MeV neutrons in the presence of oxaliplatin and/or the poly(ADP-ribose) polymerase (PARP) inhibitor PJ34. DSBs were measured by the formation of gammaH2AX foci. Results show that in SK-Hep1 cells exposed to fast neutrons in the presence of oxaliplatin, DSBs occurred and persisted with time after irradiation. While apoptosis remained low in co-treated cells, autophagy was considerably increased after irradiation and augmented by the addition of oxaliplatin. Thus, autophagic cell death appears to play a prominent role in the cytotoxicity of the combined treatment and may be linked to the generation of heavy damage to DNA.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/radiation effects , Carcinoma, Hepatocellular/pathology , Linear Energy Transfer , Liver Neoplasms/pathology , Organoplatinum Compounds/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/radiotherapy , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Histones/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/radiotherapy , Neutrons , Oxaliplatin , Poly(ADP-ribose) Polymerases/metabolism
11.
Expert Opin Ther Pat ; 20(1): 73-101, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20021286

ABSTRACT

IMPORTANCE OF THE FIELD: Ionizing radiation (IR) can produce deleterious effects in living tissues, leading to significant morbidity and a potentially fatal illness affecting various organs dose-dependently. As people may be exposed to IR during cancer radiotherapy or as a result of a radiological/nuclear incident or act of terrorism, the danger of irradiation represents a serious public health problem. At present, however, this problem remains largely impervious to medical management. There is, therefore, a pressing need to develop safe and effective radiation countermeasure (RC) agents to prevent, mitigate or treat the harmful consequences of IR exposure. AREAS COVERED IN THIS REVIEW: Recent advances in the search for RC agents as reflected by the relevant patent literature of the past five years along with peer-reviewed publications are surveyed. WHAT THE READER WILL GAIN: A total of 43 patents, describing approximately 38 chemically diverse compounds with RC potential are analyzed. These include antioxidants capable of scavenging IR-induced free radicals, modulators of cell death signaling or cell cycle progression, cytokines or growth factors promoting tissue repair and inhibitors of inflammatory cytokines. TAKE HOME MESSAGE: Several of these RC candidates appear promising, including at least two that are undergoing evaluation for fast-track clinical development.


Subject(s)
Radiation Injuries/prevention & control , Radiation Protection/methods , Radiation-Protective Agents/pharmacology , Animals , Dose-Response Relationship, Radiation , Drug Design , Humans , Patents as Topic , Radiation, Ionizing , Radiation-Protective Agents/adverse effects
12.
Cancer Res ; 69(13): 5458-66, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19549908

ABSTRACT

Cisplatin-derived anticancer therapy has been used for three decades despite its side effects. Other types of organometallic complexes, namely, some ruthenium-derived compounds (RDC), which would display cytotoxicity through different modes of action, might represent alternative therapeutic agents. We have studied both in vitro and in vivo the biological properties of RDC11, one of the most active compounds of a new class of RDCs that contain a covalent bond between the ruthenium atom and a carbon. We showed that RDC11 inhibited the growth of various tumors implanted in mice more efficiently than cisplatin. Importantly, in striking contrast with cisplatin, RDC11 did not cause severe side effects on the liver, kidneys, or the neuronal sensory system. We analyzed the mode of action of RDC11 and showed that RDC11 interacted poorly with DNA and induced only limited DNA damages compared with cisplatin, suggesting alternative transduction pathways. Indeed, we found that target genes of the endoplasmic reticulum stress pathway, such as Bip, XBP1, PDI, and CHOP, were activated in RDC11-treated cells. Induction of the transcription factor CHOP, a crucial mediator of endoplasmic reticulum stress apoptosis, was also confirmed in tumors treated with RDC11. Activation of CHOP led to the expression of several of its target genes, including proapoptotic genes. In addition, the silencing of CHOP by RNA interference significantly reduced the cytotoxicity of RDC11. Altogether, our results led us to conclude that RDC11 acts by an atypical pathway involving CHOP and endoplasmic reticulum stress, and thus might provide an interesting alternative for anticancer therapy.


Subject(s)
Cell Division/drug effects , Endoplasmic Reticulum/genetics , Melanoma, Experimental/pathology , Organometallic Compounds/therapeutic use , Ruthenium/therapeutic use , Transcription Factor CHOP/genetics , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cisplatin/therapeutic use , Cisplatin/toxicity , Endoplasmic Reticulum/drug effects , Flow Cytometry , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Luciferases/genetics , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
13.
Expert Opin Ther Pat ; 19(6): 775-99, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19456277

ABSTRACT

BACKGROUND: The efficacy of radiotherapy (RT) for cancer treatment is limited by normal tissue toxicity and by the intrinsic or acquired radioresistance of many tumours. Therefore, continuing efforts are conducted to identify radiosensitising agents that preferentially sensitise tumour cells to the cytotoxic action of RT. Recent progresses in molecular oncology have uncovered an array of novel targets, which may be exploited for RT enhancement. OBJECTIVE: To survey the patent literature of the past 4 years pertaining to the development of molecularly targeted agents as potential tumour radiosensitisers. METHODS: Patents were searched with a set of relevant keywords using several search engines. A Medline search on the same topics was performed in parallel. RESULTS/CONCLUSION: A total of 48 patents/applications were selected. These concerned agents target molecular components of pathways involved in DNA damage repair, cell growth and survival signalling, apoptosis modulation and tumour angiogenesis. Current trials of some of these agents may reveal their value as clinical radiosensitisers.


Subject(s)
Drug Delivery Systems , Neoplasms/therapy , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Clinical Trials as Topic , DNA Repair/drug effects , Humans , Neoplasms/physiopathology , Neovascularization, Pathologic/drug therapy , Patents as Topic
14.
Expert Opin Ther Pat ; 19(5): 643-62, 2009 May.
Article in English | MEDLINE | ID: mdl-19441939

ABSTRACT

BACKGROUND: Radiotherapy is utilised for the treatment of approximately 50% of patients with solid tumours, but its efficacy is limited by normal tissue toxicity and by the intrinsic or acquired radioresistance of many tumours. The combination of radiotherapy with chemotherapeutic agents that preferentially sensitise tumour cells to its cytotoxic effects has thus long been considered as a strategy to enhance cancer therapy. However, current chemoradiotherapy protocols remain highly unsatisfactory. Therefore, continuing efforts are being conducted to identify improved radiosensitising agents. OBJECTIVE: To survey the patent literature and associated peer-reviewed publications of the past 4 years pertaining to the development of novel radiosensitising agents, with a focus on anticancer drugs traditionally used as radiosensitisers and on agents targeting radioresistant hypoxic tumour cells. METHODS: Patents were searched with a set of relevant keywords using several search engines ( ep.espacenet.com/ , www.freepatentsonline.com/ , patft.uspto.gov/ ). A Medline search on the same topics was performed in parallel. RESULTS/CONCLUSION: A total of 37 patents/applications were retrieved. Of these, 14 concern the use of conventional anticancer cytotoxic drugs for tumour radiosensitisation. The other patents mostly disclose novel hypoxic radiosensitisers, bioreductive drugs and inhibitors of hypoxia-inducible factor-1. Whether these advances will translate into clinically valuable radiosensitisers is, however, unclear.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Combined Modality Therapy , Drug Delivery Systems , Humans , Neoplasms/radiotherapy , Patents as Topic
15.
Cancer Lett ; 264(1): 63-70, 2008 Jun 08.
Article in English | MEDLINE | ID: mdl-18329790

ABSTRACT

Modern protocols of concomitant chemo/radiotherapy provide a very effective strategy to treat certain types of tumors. High-linear energy transfer (LET) radiations, on the other hand, have an increased efficacy against cancer with low radiosensibility and critical localization. We previously reported that oxaliplatin, a third generation platinum drug, was able to reinforce the cytotoxicity of an irradiation by fast neutrons towards human glioblastoma U-87 cells in culture. We show here that such a combination has the capacity to enhance the number of double strand breaks in DNA and to induce autophagy in these cells. Xenografts experiments were further performed in nude mice subcutaneously transplanted with U-87 cells. When injected shortly before a single irradiation by fast neutrons, oxaliplatin causes a marked reduction of tumor growth compared with the irradiation alone. Overall, our data indicate the unique cytotoxic mechanism of a combined high-LET irradiation and oxaliplatin treatment modality and suggest its potential application in anticancer therapy.


Subject(s)
Autophagy/radiation effects , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Linear Energy Transfer , Organoplatinum Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cells, Cultured , Combined Modality Therapy , Fast Neutrons , Humans , Male , Mice , Mice, Nude , Oxaliplatin , Xenograft Model Antitumor Assays
16.
Cancer Lett ; 254(1): 54-62, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17383816

ABSTRACT

The combination of high-linear energy transfer (LET) radiation with chemotherapeutic agents may offer new perspectives in cancer treatment. We therefore assessed the consequences of a treatment in which U-87 human glioblastoma cells were irradiated with p(65)+Be neutrons in the presence of oxaliplatin, a third generation platinum anticancer drug having higher apoptosis-inducing activity than cisplatin. Cell survival, apoptosis, cell cycle progression as well as p21 and p53 protein expressions were analyzed. Results show that an enhanced cytotoxic effect was obtained when the two treatments were combined and that, unlike what we previously observed with cisplatin, this was not due to a reinforcement of apoptosis. Altogether, our results also indicate the potential of oxaliplatin for use in association with high-LET radiation against tumors refractory to conventional photon radiotherapy.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Flow Cytometry , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Linear Energy Transfer , Neutrons , Oxaliplatin , Time Factors , Tumor Suppressor Protein p53/metabolism
17.
Life Sci ; 79(6): 513-8, 2006 Jul 04.
Article in English | MEDLINE | ID: mdl-16516239

ABSTRACT

High linear energy transfer (LET) radiation have the ability to kill cancer cells resistant to conventional radiotherapy. On the other hand, protocols combining radiotherapy and chemotherapy are effective in eradicating certain inoperable cancers. In this study, we investigated the cytotoxicity of a co-treatment with fast neutrons and cisplatin in a human glioblastoma cell line, U-87. Cells cultured in vitro were irradiated with p(65)+Be neutrons in the presence of cisplatin. Cell survival and the induction of apoptosis and premature senescence were assessed at different time intervals thereafter, using a variety of methods. A marked reinforcement of the cytotoxicity was obtained when irradiation and cisplatin were associated. This reflected both an amplification of the apoptotic process and the induction of premature cell senescence. The efficiency of a combination between fast neutrons and cisplatin in inducing cell death in U-87 is more than additive. The present data concur with those we previously reported in a mouse lymphoma and suggest the potential utility of platinum compounds as adjuncts to future cancer therapy protocols using high-LET radiation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Cisplatin/pharmacology , Fast Neutrons , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 3 , Caspases/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Culture Media , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Glioblastoma/enzymology , Glioblastoma/pathology , Humans
18.
Biochem Biophys Res Commun ; 334(2): 533-42, 2005 Aug 26.
Article in English | MEDLINE | ID: mdl-16018969

ABSTRACT

We have previously shown that ionizing radiation-induced apoptosis in human lymphoblastoid cells differs according to their p53 status, and that caspase 8-mediated cleavage of BID is involved in the p53-dependent pathway. In the present study, we investigated the role of Fas signaling in caspase 8 activation induced by fast neutrons irradiation in these cells. Fas and FasL expression was assessed by flow cytometry and by immunoblot. We also measured Fas aggregation after irradiation by fluorescence microscopy. We found a decrease of Fas expression after irradiation, but no change in Fas ligand expression. We also showed that, in contrast to the stimulation of Fas by an agonistic antibody, Fas aggregation did not occur after irradiation. Altogether, our data strongly suggest that fast neutrons induced-apoptosis is Fas-independent, even in p53-dependent apoptosis.


Subject(s)
Apoptosis/radiation effects , Lymphocytes/metabolism , Lymphocytes/radiation effects , Neutrons , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/metabolism , fas Receptor/metabolism , Humans
19.
Cancer Lett ; 211(2): 155-61, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15219939

ABSTRACT

The pro-apoptotic ability of (Z)-3,5,4'-Tri-O-methyl-resveratrol (R3) was investigated in vitro on the human lymphoblastoid cell line TK6 and its p53-knockout counterpart (NH32). In both cell lines, R3 induced the stimulation of caspase-3. Although R3 induced growth inhibition and apoptosis of both cell lines, two distinct mechanisms were observed. The p53-knockout NH32 cells were shown to override the G2/M phase checkpoint with development of hyperdiploid cells, whereas TK6 cells accumulated at G2/M. As p53 function is often altered in human cancer cells, these results show that the pro-apototic effects of R3 against tumor cells are independent of their p53 status.


Subject(s)
Anisoles/pharmacology , Apoptosis/drug effects , Genes, p53/genetics , Stilbenes/pharmacology , Caspase 3 , Caspases/pharmacology , Cell Culture Techniques , Cell Cycle/drug effects , Genetic Engineering , Humans , Leukemia, Lymphoid/pathology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/pharmacology
20.
Bioorg Med Chem ; 12(13): 3619-25, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15186846

ABSTRACT

The synthesis of a new alpha-methylene-gamma-butyrolactone-psoralen heterodimer 2 is reported. Its photoantiproliferative activity and skin phototoxicity were compared with that of 5-methoxypsoralen (5-MOP) and another heterodimer 1. Both derivatives show a significant phototoxicity toward malignant cell lines including melanoma cells A375 compared to their intrinsic cytotoxicity in the dark. Both compounds were found to be nonphototoxic on mice skin and therefore could be active potential drugs in photochemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Ficusin/chemical synthesis , Ficusin/pharmacology , Furocoumarins , Lactones/chemical synthesis , Lactones/pharmacology , Photochemotherapy , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Dimerization , Female , Ficusin/adverse effects , Ficusin/chemistry , Humans , Inhibitory Concentration 50 , Lactones/adverse effects , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Inbred BALB C , Molecular Structure , Skin/drug effects , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...