Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Cell Biochem ; 118(7): 1689-1698, 2017 07.
Article in English | MEDLINE | ID: mdl-27922187

ABSTRACT

Tomentosin, a natural sesquiterpene lactone purified from of Inula viscosa L., was investigated for its anti-proliferative, telomere shortening, and apoptotic effects on human cervical cancer HeLa and SiHa cell lines. Tomentosin was found to inhibit the growth of SiHa and HeLa cell lines in dose and time-dependent manner (IC50 values of 7.10 ± 0.78 µM and 5.87 ± 0.36 µM, respectively after 96 h of treatment). As evidenced by TTAGGG telomere length assay, tomentosin target specifically the telomeric overhang lengthening. This was confirmed by the evaluation of the cytotoxic effects of tomentosin in the foetal fibroblast Wi38 and JW10 cells which were derived from Wi38 and express hTERT, the telomerase catalytic subunit. We found that JW10 cells are 4.7-fold more sensitive to tomentosin which argues for telomere as its specific target. Furthermore, we found that tomentosin mediate this cytotoxic effect by inducing apoptosis and cell cycle arrest at G2/M phase. Morphological features of treated cells, as evidenced by Hoechst 33324 staining, revealed that the cytotoxic effect was due to induction of apoptosis. This was accompanied by pro-caspase-3 cleavage, an increase in caspase-3 activity and a cleavage of poly (ADP-ribose) polymerase (PARP). Moreover, tomentosin induced a decrease in mitochondrial membrane potential (ΔΨm) and an increase in reactive oxygen species (ROS), accompanied by a decrease in Bcl-2 expression. This indicates that tomentosin-induced apoptosis may involve a mitochondria-mediated signaling pathway. This study provides the first evidence that tomentosin targets telomere machinery and induces apoptosis in cervical cancer cells. The molecular mechanism underlying tomentosin-induced apoptosis may involve a mitochondria-mediated signaling pathway. J. Cell. Biochem. 118: 1689-1698, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Lactones/pharmacology , Sesquiterpenes/pharmacology , Telomere/genetics , Uterine Cervical Neoplasms/genetics , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , G2 Phase/drug effects , G2 Phase/genetics , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Reactive Oxygen Species/metabolism , Telomere/drug effects
2.
Nutr Cancer ; 68(1): 131-43, 2016.
Article in English | MEDLINE | ID: mdl-26771897

ABSTRACT

Telomerase is activated in human papillomavirus (HPV) positive cervical cancer and targeting telomeres offers a novel anticancer therapeutic strategy. In this study, the telomere targeting properties, the cytotoxic as well as the pro-apoptotic effects of hexane (IV-HE) and dichloromethane (IV-DF) fractions from Inula viscosa L. extracts were investigated on human cervical HeLa and SiHa cancer cells. Our data demonstrate that IV-HE and IV-DF extracts were able to inhibit cell growth in HeLa and SiHa cells in a dose-dependent manner and studied resistant cell lines exhibited a resistance factor less than 2 when treated with the extracts. IV-HE and IV-DF extracts were able to inhibit telomerase activity and to induce telomere shortening as shown by telomeric repeat amplification protocol and TTAGGG telomere length assay, respectively. The sensitivity of fibroblasts to the extracts was increased when telomerase was expressed. Finally, IV-HE and IV-DF were able to induce apoptosis as evidenced by an increase in annexin-V labeling and caspase-3 activity. This study provides the first evidence that the IV-HE and IV-DF extracts from Inula viscosa L. target telomeres induce apoptosis and overcome drug resistance in tumor cells. Future studies will focus on the identification of the molecules involved in the anticancer activity.


Subject(s)
Apoptosis/drug effects , Inula , Plant Extracts/pharmacology , Telomere Shortening/drug effects , Annexin A5/analysis , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Telomerase/metabolism
3.
Phytomedicine ; 22(13): 1186-94, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26598918

ABSTRACT

BACKGROUND: High consumption of flavonoids has been associated with a decrease risk of cancer. Alfalfa (Medicago sativa) leaves have been widely used in traditional medicine and is currently used as a dietary supplement because of their high nutrient content. We previously reported the cytotoxic activity of alfalfa leaf extracts against several sensitive and multidrug resistant tumor cell lines. HYPOTHESIS/PURPOSE: We aimed to determine whether medicarpin and millepurpan, two isoflavonoids isolated from alfalfa leaves, may have pro-apoptotic effects against drug-sensitive (P388) and multidrug resistant P388 leukemia cells (P388/DOX). STUDY DESIGN/METHODS: Cells were incubated with medicarpin or millepurpan for the appropriate time. Cell viability was assessed by the MTT assay. DNA fragmentation was analyzed by agarose gel electrophoresis. Cell cycle analysis was realized by flow cytometry technics. Caspases 3 and 9 activities were measured using Promega caspACE assay kits. Proteins and genes expression were visualized respectively by western-blot using specific antibodies and RT-PCR assay. RESULTS: P-glycoprotein-expressing P388/DOX cells did not show resistance to medicarpin (IC50 ≈ 90 µM for P388 and P388/DOX cells) and millepurpan (IC50 = 54 µM and 69 µM for P388 and P388/DOX cells, respectively). Treatment with medicarpin or millepurpan triggered apoptosis in sensitive as well as multidrug resistant P388 cells. These effects were mediated through the mitochondrial pathway by modifying the balance pro/anti-apoptotic proteins. While 3 µM doxorubicin alone could not induce cell death in P388/DOX cells, concomitant treatment with doxorubicin and subtoxic concentration of medicarpin or millepurpan restored the pro-apoptotic cascade. Each compound increased sensitivity of P388/DOX cells to doxorubicin whereas they had no effect in sensitive P388 cells. Vinblastine cytotoxicity was also enhanced in P388/DOX cells (IC50 = 210 nM to 23 and 25 nM with medicarpin and millepurpan, respectively). This improved sensitivity was mediated by an increased uptake of doxorubicin in P388/DOX cells expressing P-gp. P-gp expression was not altered by exposure to medicarpin and millepurpan. CONCLUSION: These data indicate that medicarpin and millepurpan possess pro-apoptotic properties and potentiate the cytotoxicity of chemotherapy drugs in multidrug resistant P388 leukemia cells by modulating P-gp-mediated efflux of drugs. These flavonoids may be used as chemopreventive agents or as sensitizer to enhance cytotoxicity of chemotherapy drugs in multidrug resistant cancer cells.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , Leukemia P388/metabolism , Medicago sativa/chemistry , Pterocarpans/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Caspases/metabolism , Cell Line, Tumor/drug effects , Doxorubicin , Mice , Molecular Structure , Plant Leaves/chemistry
4.
Phytochemistry ; 109: 49-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25468712

ABSTRACT

Seven triterpenoid glycosides, named meliosmosides A-G, were isolated from the leaves of Meliosma henryi Diels (Sabiaceae). Their structures were elucidated by different spectroscopic methods including 1D and 2D NMR experiments as well as HRESIMS analysis. Isolated compounds were evaluated for their cytotoxic activity against KB cell line.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Glycosides/chemistry , Magnoliopsida/chemistry , Saponins/chemistry , Triterpenes/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Line, Tumor , Glycosides/isolation & purification , Humans , Molecular Structure , Plant Leaves/chemistry , Saponins/isolation & purification , Triterpenes/isolation & purification
5.
Nutr Cancer ; 66(3): 483-91, 2014.
Article in English | MEDLINE | ID: mdl-24628411

ABSTRACT

Alfalfa (Medicago sativa) has been used to cure a wide variety of ailments. However, only a few studies have reported its anticancer effects. In this study, extracts were obtained from alfalfa leaves and their cytotoxic effects were assessed on several sensitive and multidrug-resistant tumor cells lines. Using the mouse leukaemia P388 cell line and its doxorubicin-resistant counterpart (P388/DOX), we showed that the inhibition of cell growth induced by alfalfa leaf extracts was mediated through the induction of apoptosis, as evidenced by DNA fragmentation analysis. The execution of programmed cell death was achieved via the activation of caspase-3, leading to PARP cleavage. Fractionation of toluene extract (To-1), the most active extract obtained from crude extract, led to the identification of 3 terpene derivatives and 5 flavonoids. Among them, (-)-medicarpin, (-)-melilotocarpan E, millepurpan, tricin, and chrysoeriol showed cytotoxic effects in P388 as well as P388/DOX cells. These results demonstrate that alfalfa leaf extract may have interesting potential in cancer chemoprevention and therapy.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Leukemia P388/drug therapy , Medicago sativa/chemistry , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor/drug effects , DNA Fragmentation/drug effects , Doxorubicin/pharmacology , Drug Resistance, Multiple , Humans , Leukemia P388/pathology , Mice , Plant Extracts/analysis , Plant Leaves/chemistry
6.
Oncol Rep ; 27(1): 232-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21935580

ABSTRACT

Resistance to etoposide has been associated with the overexpression of P-glycoprotein and MRP1 in human tumor cells. However, the role of BCRP in resistance to etoposide has not been clearly established, especially the significance of arginine 482 mutations in drug transport (cellular uptake and efflux). Different levels of resistance to etoposide have been recently observed in cells expressing BCRP in terms of cytotoxicity. The aim of this work was to study the effects of these mutations on the functional involvement of BCRP in etoposide transport. HEK293 cells were transfected with an empty vector (HEK/V), the vector bearing the wild-type BCRP (HEK/R482), the mutant arginine-482-glycine (HEK/R482G) or the mutant arginine-482-threonine (HEK/R482T). MTT assay was used to study the cytotoxic effect of etoposide and [3H]-etoposide was used to determine cellular drug uptake and efflux. Data show that HEK/R482G cells displayed the highest levels of resistance to etoposide. Cellular [3H]-etoposide uptake was lower in HEK/R482, HEK/R482G and HEK/R482T cells compared to HEK/V cells. In addition, cellular [3H]-etoposide uptake in HEK/R482G was the lowest. Drug efflux measurements showed that fumitremorgin C was able to increase the residual cellular [3H]-etoposide uptake in BCRP-transfected cells and especially in HEK/R482G ones. Our data show that the R482G mutation in BCRP is able to increase efflux of etoposide and that mutation analysis at codon 482 may be of clinical importance in cancers treated with etoposide.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Antineoplastic Agents, Phytogenic/metabolism , Drug Resistance, Neoplasm/genetics , Etoposide/metabolism , Neoplasm Proteins/genetics , Point Mutation , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Arginine/genetics , Biological Transport/genetics , Flow Cytometry , Glycine/genetics , HEK293 Cells , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
7.
Oncol Rep ; 25(4): 1161-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21318225

ABSTRACT

The selection pressure for resistance to chemotherapy is accompanied by the enhanced expression of ABC proteins and increased cellular glycosphingolipid content. Thus, a possible connection between glycosphingolipid metabolism and ABC proteins in drug resistance has been suggested. In the present study, we established two human multidrug-resistant (MDR) cell lines derived from MESSA sarcoma cells by culturing with increasing concentrations of doxorubicin (DX5 cells) or doxorubicin together with cyclosporin A (GARF cells). Both resistant cell lines overexpressed the MDR1 gene and the wild-type P-glycoprotein at the same level. The cyclosporin derivative PSC833, a potent inhibitor of P-glycoprotein, sensitized DX5 but not GARF cells to the cytotoxic effects of daunorubicin. Moreover, PSC833 increased the nuclear accumulation of daunorubicin and the cellular accumulation of [3H]vinblastine in the DX5 but not in the GARF cells. The cellular incorporation of [3H]-cyclosporin A was lower in DX5 cells compared to MESSA and GARF cells, which incorporated the same level of [3H]-cyclosporin A. Sphingolipid analysis showed that the lactosylceramide level was 2.5- and 5-fold higher in DX5 and GARF cells, respectively, than in MESSA cells. Whereas the pharmacological inhibition of lactosylceramide synthesis was able to reverse only partially the resistance of GARF cells to daunorubicin without significant increase in nuclear accumulation of the drug, the same treatment before the co-treatment with PSC833 and daunorubicin increased the cytotoxic effect of daunorubicin and its nuclear accumulation. These data suggest a possible relationship between lactosylceramide levels and the resistance of P-glycoprotein to modulation by MDR modulators.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antigens, CD/metabolism , Cyclosporins/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Lactosylceramides/metabolism , Sarcoma/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Cell Proliferation , Daunorubicin/pharmacology , Doxorubicin/pharmacology , Flow Cytometry , Humans , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma/metabolism , Tumor Cells, Cultured
8.
J Cell Biochem ; 110(4): 893-902, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20564188

ABSTRACT

Resveratrol, a naturally occurring polyphenol, has been shown to possess chemopreventive activities. In this study, we show that resveratrol (0-500 microM) inhibits the growth of a doxorubicin-resistant B16 melanoma cell subline (B16/DOX) (IC(50) = 25 microM after 72 h, P < 0.05). This was accomplished by imposing an artificial checkpoint at the G(1)-S phase transition, as demonstrated by cell-cycle analysis and down-regulation of cyclin D1/cdk4 and increased of p53 expression level. The G(1)-phase arrest of cell cycle in resveratrol-treated (10-100 microM) B16/DOX cells was followed by the induction of apoptosis, which was revealed by pyknotic nuclei and fragmented DNA. Resveratrol also potentiated at subtoxic dose (25 microM for 24 h) doxorubicin cytotoxicity in the chemoresistant B16 melanoma (P < 0.01). When administered to mice, resveratrol (12.5 mg/kg) reduced the growth of an established B16/DOX melanoma and prolonged survival (32% compared to untreated mice). All these data support a potential use of resveratrol alone or in combination with other chemotherapeutic agents in the management of chemoresistant tumors.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Melanoma, Experimental/pathology , Stilbenes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Mice , Resveratrol
9.
Methods Mol Biol ; 596: 433-46, 2010.
Article in English | MEDLINE | ID: mdl-19949935

ABSTRACT

Multidrug-resistance (MDR) is the major reason for failure of cancer therapy. ATP-binding cassette (ABC) transporters contribute to drug resistance via ATP-dependent drug efflux. P-glycoprotein (Pgp), which is encoded by MDR1 gene, confers resistance to certain anticancer agents. The development of agents able to modulate MDR mediated by Pgp and other ABC transporters remained a major goal for the past 20 years. The calcium blocker verapamil was the first drug shown to be a modulator of Pgp, and since many different chemical compounds have been shown to exert the same effect in vitro by blocking Pgp activity. These included particularly immunosuppressors. Cyclosporin A (CSA) was the first immunosuppressor that have been shown to modulate Pgp activity in laboratory models and entered very early into clinical trials for reversal of MDR. The proof of reversing activity of CSA was found in phase II studies with myeloma and acute leukemia. In phase III studies, the results were less convincing regarding the response rate, progression-free survival, and overall survival, which were detected in advanced refractory myeloma. The non-immunosuppressive derivative PSC833 (valspodar) was subsequently developed. This compound showed tenfold higher potency in reversal of MDR mediated by Pgp. However, pharmacokinetic interactions required reductions in the dose of the concurrently administered anticancer agents. The pharmacokinetic interactions were likely because of decreased clearance of the anticancer agents, possibly as a result of Pgp inhibition in organs such as the gastrointestinal tract and kidney, as well as inhibition of cytochrome P450. Finally, CSA and PSC833 have been shown also to modulate the ceramide metabolism which stands as second messenger of anticancer agent-induced apoptosis. In fact, CSA and PSC833 are also able to respectively inhibit ceramide glycosylation and stimulate de novo ceramide synthesis. This could enhance the cellular level of ceramide and potentiate apoptosis induced by some anticancer agents.


Subject(s)
Drug Resistance, Multiple/drug effects , Immunosuppressive Agents , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/therapeutic use , Ceramides/metabolism , Clinical Trials as Topic , Cyclosporine/chemistry , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Molecular Structure , Neoplasms/drug therapy , Neoplasms/metabolism
10.
Vaccine ; 27(44): 6107-9, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19686694

ABSTRACT

Malignant melanoma causes significant health problems. The identification of tumour-associated antigens has led to novel approaches to increase T cell mediated anti-tumour immune response. Melan-A/MART-1 has been use as target antigen for several T cell based immunotherapeutic treatments. More recently, the critical role of CD4+ T cells in inducing and maintaining anti-tumour immunity has been increasingly recognized. In order to optimize tumour immunotherapy, greater efforts have been concentrated on the identification of tumour antigens presented by MHC class II molecules to CD4+ T cells. In a publication, Tiwari et al. (2004) [1] have identified by a computational approach the 15-mer amino-acid sequence 101-115 (PPAYEKLSAEQSPPP) of the Melan-A/MART-1 as a good target for a vigorous and safe immunotherapy. Therefore, we have investigated the in vivo anti-tumour activity of this peptide in a murine melanoma model. For the prophylactic treatment, 20microg or 50microg peptide was subcutaneously injected in mice once a week during 3 weeks before tumour induction. Treatment with 50microg peptide significantly affected tumour development. Thus, our preliminary data demonstrate potential in vivo prophylactic activity of the 101-115 peptide-based vaccine to control melanoma growth.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Melanoma/therapy , Neoplasm Proteins/immunology , Amino Acid Sequence , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Female , Immunotherapy , MART-1 Antigen , Melanoma/immunology , Mice , Molecular Sequence Data , Peptides/immunology
11.
Int J Pharm ; 353(1-2): 131-8, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18162346

ABSTRACT

Microparticles and nanoparticles represent promising carriers for the in vivo delivery of peptides, proteins or deoxyribonucleic acid (DNA). In this study, new hydroxyethylstarch (HES) microparticles were obtained by interfacial cross-linking with terephtaloyl chloride. These microparticles exhibit the characteristics required to improve antigen release and presentation to antigen presentating cells compared to free antigens. The adjuvant activity of HES microparticles as vaccine carrier was investigated in mice using bovine serum albumin (BSA) as model antigen. We showed HES microparticles were phagocyted by peritoneal mononuclear cells. The immunization with BSA-microparticles induced antibody synthesis that was predominantly immunoglobulin G1 (IgG1). Aluminium hydroxide remained more efficient to induce IgG synthesis. The analysis of the cytokine profile from spleen cells revealed that BSA-microparticles induced the secretion of both interferon-gamma (IFN-gamma) and interleukin-4 (IL-4). However, the immune responses induced by BSA-microparticles were qualitatively and quantitatively affected by the route of injection. Taken together, these results demonstrate that HES microparticles induce a mixed T helper 1/T helper 2 response against BSA and may be a suitable delivery and presentation system in the field of vaccine development.


Subject(s)
Hydroxyethyl Starch Derivatives/administration & dosage , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/immunology , Animals , Cytokines/biosynthesis , Female , Immunization , Immunoglobulin G/blood , Lymphocyte Activation , Mice , Phagocytosis , Th1 Cells/immunology , Th2 Cells/immunology
12.
Cancer Lett ; 257(2): 165-71, 2007 Nov 18.
Article in English | MEDLINE | ID: mdl-17517470

ABSTRACT

The clinical use of chemotherapy in cancer treatment is limited by the occurrence of multidrug resistance (MDR) associated with the overexpression of membrane transporters, one of the best known is P-glycoprotein (Pgp), that actively expels drugs out of tumor cells. To overcome Pgp-mediated MDR, synthetic peptides corresponding to fragments from extracellular loops 1, 2 and 4 of the murine Pgp were coupled to polyethylene glycol-distearoylphosphatidylethanolamine and inserted into empty or monophosphoryl lipid A-containing liposomes. This formulation elicited specific antibodies which blocked Pgp-mediated efflux of doxorubicin, resulting in increased intracellular drug accumulation and subsequent potentiation of the cytotoxic effect of doxorubicin on multidrug-resistant P388 (P388R) cells. Previous immunizations with MDR1 peptides improved the efficiency of chemotherapy against P388R cells in vivo, with an increase of 83% of mice survival time. Overall, these results suggest that this approach can modulate Pgp activity by blocking drug efflux and may have clinical relevance as an alternative strategy to toxic chemosensitizers in drug-resistant cancer therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , Doxorubicin/pharmacology , Immunization/methods , Peptides/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amino Acid Sequence , Animals , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Antibodies/administration & dosage , Antibodies/immunology , Antibodies/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Drug Resistance, Bacterial , Female , Flow Cytometry , Liposomes/chemistry , Mice , Mice, Inbred Strains , Molecular Sequence Data , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Peptides/chemistry , Polyethylene Glycols/chemistry , Survival Analysis , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology
13.
Biochem Biophys Res Commun ; 358(1): 325-30, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17477907

ABSTRACT

Overexpression of the 170 kDa plasma membrane P-glycoprotein (P-gp) represents the most common MDR mechanism in chemotherapy. In this work, specific autoantibodies to fragments from extracellular loops 1, 2, and 4 of the murine MDR1 P-gp were elicited in mice using synthetic palmitoylated peptides reconstituted in liposomes and alum. The highest IgG level was observed after the third immunization and the immune response against lipopeptides was still detected more than 200 days after immunizations. Immunocytochemichal studies revealed that these antibodies were specific for P-gp. When incubated with P-gp-expressing MDR cell lines, serum from immunized mice restored sensitivity to either doxorubicin or vinblastine, or had no effect in a cell type specific manner, suggesting that several mechanisms may occur in the establishment of the MDR phenotype. The expression of mdr1 and mdr3 genes was unchanged in organs from mice immunized with palmitoylpeptides grafted on liposomes. These results suggest that the induction of autoantibodies to P-gp is a safe strategy to overcome MDR in cancer chemotherapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , Autoantibodies/biosynthesis , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Genes, MDR , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP-Binding Cassette Transporters/biosynthesis , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Female , Liposomes , Mice , Organ Specificity , Peptides/immunology , Vinblastine/pharmacology
14.
Anticancer Res ; 27(6B): 3865-74, 2007.
Article in English | MEDLINE | ID: mdl-18225544

ABSTRACT

BACKGROUND: Peripheral blood mononuclear cells (PBMCs) present an antitumor activity in vitro on doxorubicin-resistant B16 melanoma (B16R) spheroids, but do not inhibit tumor growth in vivo. This study aimed to improve in vivo the antitumor immune response by using antigen presenting cells. MATERIALS AND METHODS: After injection of B16R cells, mice received either tumor cell lysate-pulsed PBMCs, or naive or cytostatic tumor cell-pulsed bone marrow-derived dendritic cells (DCs). Tumor development and mouse survival were followed and spleen cell cytotoxic activity against B16R was estimated in vitro by Lactate dehydrogenase activity measurement. RESULTS: The best results were obtained with peritumoral injections of cytostatic tumor cell-pulsed DCs which induced tumor regression, increased mouse survival and exhibited a higher splenocyte cytotoxic activity against B16R cells. When injected at a distant site, the efficacy was reduced. Furthermore, preventative injections of pulsed DCs induced protection of mice against B16R cells. CONCLUSION: These results show the important role of cytostatic tumor cell-pulsed DCs in a specific antitumor immunity establishment. Consequently, they could be used combined with other treatments to improve clinical outcomes.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , Cell Growth Processes/immunology , Female , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , T-Lymphocytes/immunology
15.
Melanoma Res ; 16(4): 325-33, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16845328

ABSTRACT

As tumour cells use multiple mechanisms to escape from chemotherapeutic drugs, the anti-tumoural activity of naive mouse peripheral blood mononuclear cells was examined in this study, using a mouse melanoma cell subline resistant to doxorubicin (B16R). Multicellular spheroids are known to be the most adapted in-vitro model to mimic solid tumours in vivo and are used to investigate many aspects of tumour biology. For in-vitro studies, murine peripheral blood mononuclear cells recovered by Ficoll gradient centrifugation after caudal puncture were co-cultured with multicellular tumour spheroids of B16R cells. Morphological investigations show that peripheral blood mononuclear cells were gathered and focused around the spheroids after 14 h of co-culture and contacts were established within 32 h. Between 38 and 62 h of co-culture, the size of the spheroids decreased significantly. The peripheral blood mononuclear cells exerted cytolytic effects that correlated with the induction of cell death in spheroids of B16R melanoma cells. Immunological investigations to localize and identify peripheral blood mononuclear cells that exerted anti-tumoural effects have shown that spheroids were deeply infiltrated by monocytes/macrophages at a stage in which a significant cytolytic activity and a strong cell death rate were observed. For in-vivo studies, intratumoural injections of syngeneic naive peripheral blood mononuclear cells were administered. A weak potential in-vivo anti-tumoural effect of these cells was observed (inhibition of B16R melanoma growth by 20-25%) but the median survival time of mice treated with peripheral blood mononuclear cells did not increase compared with untreated control mice. Thus, despite anti-tumoural activities of peripheral blood mononuclear cells against the poorly immunogenic and highly metastatic chemoresistant B16 melanoma cells in vitro, a potential anti-melanoma effect in vivo, if present, did not increase the life span of B16R melanoma-bearing mice.


Subject(s)
Leukocytes, Mononuclear/physiology , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Survival , Coculture Techniques , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Immunohistochemistry , In Situ Hybridization , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Spheroids, Cellular , Tumor Cells, Cultured
16.
Int J Cancer ; 111(3): 381-7, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15221965

ABSTRACT

Curcumin, the active ingredient from the spice turmeric (Curcuma longa Linn), is known to be an anti-oxidant and an anti-inflammatory agent. It has been demonstrated recently to possess anti-angiogenic effects and pro-apoptotic activities against Ehrlich ascites tumor cells. In the current study, curcumin was found to be cytotoxic in vitro for B16-R melanoma cells resistant to doxorubicin either cultivated as monolayers or grown in three-dimensional (3-D) cultures (spheroids). We have demonstrated that the cytotoxic effect observed in the 2 culture types can be related to the induction of programmed cell death. In our in vivo studies, we examined the effectiveness of a prophylactic immune preparation of soluble proteins from B16-R cells, or a treatment with curcumin as soon as tumoral appearance, alone or in combination, on the murine melanoma B16-R. The combination treatment resulted in substantial inhibition of growth of B16-R melanoma, whereas each treatment by itself showed little effect. Moreover, animals receiving the combination therapy exhibited an enhancement of their humoral anti-soluble B16-R protein immune response and a significant increase in their median survival time (> 82.8% vs. 48.6% and 45.7% respectively for the immunized group and the curcumin-treated group). Our study shows that curcumin may provide a valuable tool for the development of a therapeutic combination against the melanoma.


Subject(s)
Antineoplastic Agents/toxicity , Cell Division/drug effects , Curcumin/toxicity , Melanoma, Experimental/pathology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Curcumin/therapeutic use , DNA, Neoplasm/drug effects , DNA, Neoplasm/isolation & purification , Female , Melanoma, Experimental/drug therapy , Mice , Phytotherapy
17.
Anticancer Res ; 24(2B): 935-41, 2004.
Article in English | MEDLINE | ID: mdl-15161047

ABSTRACT

BACKGROUND: Multicellular spheroids are known to be the most adapted model to keep the in vitro resistance properties of cells. This in vivo-like tissue-culture representation was applied to investigate the immune reactivity of MCF-7 cells by monocytes. MATERIALS AND METHODS: Human blood monocytes, obtained by elutriation, were co-cultured with multicellular tumor spheroids of drug-sensitive (MCF-7S) and doxorubicin-resistant (MCF-7DXR) MCF-7 breast cancer cells. RESULTS: Tumor cells, according to their phenotype, induced differential recruitment and behavior of the immune cells towards the two types of spheroids. The secretion of various cytokines and the expression of several adhesion molecules were analysed. The MCF-7DXR/monocytes co-culture supernatant showed higher levels of IL-6 and IL-8 than the MCF-7S/monocytes co-culture supernatant. Cells from the MCF-7DXR spheroids expressed some adhesion molecules, CD-44 and CD-54, leading to a strong cellular cohesion in comparison with the sensitive spheroids. CONCLUSION: The two spheroid phenotypes represented an excellent model system for determining the precise tumor microenvironment in which cells move, the crucial molecular requirements and the mechanisms by which immunotherapeutic strategies could be developed to eradicate chemo-resistant tumors.


Subject(s)
Breast Neoplasms/immunology , Cell Communication/immunology , Monocytes/immunology , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Coculture Techniques , Cytokines/biosynthesis , Drug Resistance, Multiple , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Monocytes/cytology , Monocytes/metabolism , Spheroids, Cellular
18.
Crit Rev Oncol Hematol ; 42(3): 283-96, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12050020

ABSTRACT

Gallium (Ga) is the second metal ion, after platinum, to be used in cancer treatment. Its activities are numerous and various. It modifies three-dimensional structure of DNA and inhibits its synthesis, modulates protein synthesis, inhibits the activity of a number of enzymes, such as ATPases, DNA polymerases, ribonucleotide reductase and tyrosine-specific protein phosphatase. Ga alters plasma membrane permeability and mitochondrial functions. Ga salts are taken up more efficiently and more specifically by tumour cells when orally administered. New compounds have been prepared: Ga maltolate, doxorubicin-Ga-transferrin conjugate and Tris(8-quinolinolato)Ga(III), which show interesting activities. Ga toxicity is well documented in vitro and in vivo in animals. In humans, the oral administration Ga is less toxic, and allows a chronic treatment, allowing an improvement of its bioavailability in tumours, by comparison with the parenteral use. The anticancer activity of Ga salts has been demonstrated but other effects have also been noted such as many bone effects that could be useful in bone metastatic patients. Its has also been shown that a long period of administration could induce tumour fibrosis. Ga is synergistic with other anticancer drugs. Although not as potent as platinum in vitro, the anticancer activity of Ga should not be ignored, but the schedule of administration still needs to be optimised and new compounds are now under clinical investigations.


Subject(s)
Gallium/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Drug Evaluation, Preclinical , Gallium/pharmacokinetics , Gallium/pharmacology , Humans , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/pharmacology , Organometallic Compounds/therapeutic use
19.
Crit Rev Oncol Hematol ; 42(3): 317-25, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12050023

ABSTRACT

The history of platinum in cancer treatment began 150 years ago with the first synthesis of cisplatin; but it was not used in the clinic before 30 years ago. Then 3000 derivatives were synthesised and tested, with poor successes: three other derivatives only are available today. Clearly they are not more active, but they are less toxic than cisplatin, although two, carboplatin and nedaplatin, yield a cross-resistance, while one, oxaliplatin, does not. Their mechanisms of action are similar: these four pro-drugs form adducts with DNA, impairing DNA synthesis and repair then. Their pharmacokinetics are complicated since we always measure two overlapping pharmacokinetics: those of the parent compound and of the bound platinum. Cisplatin is now recommended for few cancers, it is replaced by less-toxic carboplatin, and therefore more easily used in combination. Oxaliplatin give interesting results in a number of cancers. The official recommendation in Japan for nedaplatin is head and neck, testicular, lung, oesophageal, ovarian, and cervical cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Platinum Compounds/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Resistance , Humans , Molecular Structure , Neoplasms/pathology , Pharmacokinetics , Platinum Compounds/chemistry , Platinum Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...