Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Front Oncol ; 12: 849127, 2022.
Article in English | MEDLINE | ID: mdl-35252017
2.
Oncotarget ; 10(28): 2709-2721, 2019 Apr 12.
Article in English | MEDLINE | ID: mdl-31105871

ABSTRACT

Despite recent progress in its treatment, Multiple Myeloma (MM) remains incurable and its associated bone disease persists even after complete remission. Thus, identification of new therapeutic agents that simultaneously suppress MM growth and protect bone is an unmet need. Herein, we examined the effects of Aplidin, a novel anti-cancer marine-derived compound, on MM and bone cells. In vitro, Aplidin potently inhibited MM cell growth and induced apoptosis, effects that were enhanced by dexamethasone (Dex) and bortezomib (Btz). Aplidin modestly reduced osteocyte/osteoblast viability and decreased osteoblast mineralization, effects that were enhanced by Dex and partially prevented by Btz. Further, Aplidin markedly decreased osteoclast precursor numbers and differentiation, and reduced mature osteoclast number and resorption activity. Moreover, Aplidin reduced Dex-induced osteoclast differentiation and further decreased osteoclast number when combined with Btz. Lastly, Aplidin alone, or suboptimal doses of Aplidin combined with Dex or Btz, decreased tumor growth and bone resorption in ex vivo bone organ cultures that reproduce the 3D-organization and the cellular diversity of the MM/bone marrow niche. These results demonstrate that Aplidin has potent anti-myeloma and anti-resorptive properties, and enhances proteasome inhibitors blockade of MM growth and bone destruction.

3.
Sci Rep ; 8(1): 8616, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29872155

ABSTRACT

Plocabulin (PM060184) is a microtubule depolymerizing agent with potent antiproliferative activity undergoing phase II clinical trials for the treatment of solid tumors. Plocabulin shows antifungal activity virtually abolishing growth of the filamentous fungus Aspergillus nidulans. A. nidulans hyphae depend both on mitotic and interphase microtubules, as human cells. Here, we exploited the A. nidulans genetic amenability to gain insight into the mechanism of action of plocabulin. By combining mutations in the two A. nidulans ß-tubulin isotypes we obtained a plocabulin-insensitive strain, showing that ß-tubulin is the only molecular target of plocabulin in fungal cells. From a genetic screen, we recovered five mutants that show plocabulin resistance but do not carry mutations in ß-tubulin. Resistance mutations resulted in amino acid substitutions in (1) two subunits of the eukaryotic translation initiation factor eIF2B activating the General Amino Acid Control, (2) TIM44, an essential component of the inner mitochondrial membrane translocase, (3) two transcription factors of the binuclear zinc cluster family potentially interfering with the uptake or efflux of plocabulin. Given the conservation of some of the identified proteins and their respective cellular functions in the tumor environment, our results pinpoint candidates to be tested as potential biomarkers for determination of drug efficiency.


Subject(s)
Antineoplastic Agents/pharmacology , Aspergillus nidulans/drug effects , Drug Resistance, Neoplasm , Microtubules/drug effects , Polyketides/pharmacology , Pyrones/pharmacology , Drug Resistance, Fungal , Fungal Proteins/genetics , Mutation, Missense , Tubulin/genetics
5.
Cancer Res ; 76(22): 6657-6668, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27697767

ABSTRACT

There is a great need to develop novel approaches to target oncogenic transcription factors with small molecules. Ewing sarcoma is emblematic of this need, as it depends on the continued activity of the EWS-FLI1 transcription factor to maintain the malignant phenotype. We have previously shown that the small molecule trabectedin interferes with EWS-FLI1. Here, we report important mechanistic advances and a second-generation inhibitor to provide insight into the therapeutic targeting of EWS-FLI1. We discovered that trabectedin functionally inactivated EWS-FLI1 by redistributing the protein within the nucleus to the nucleolus. This effect was rooted in the wild-type functions of the EWSR1, compromising the N-terminal half of the chimeric oncoprotein, which is known to be similarly redistributed within the nucleus in the presence of UV light damage. A second-generation trabectedin analogue lurbinectedin (PM01183) caused the same nuclear redistribution of EWS-FLI1, leading to a loss of activity at the promoter, mRNA, and protein levels of expression. Tumor xenograft studies confirmed this effect, and it was increased in combination with irinotecan, leading to tumor regression and replacement of Ewing sarcoma cells with benign fat cells. The net result of combined lurbinectedin and irinotecan treatment was a complete reversal of EWS-FLI1 activity and elimination of established tumors in 30% to 70% of mice after only 11 days of therapy. Our results illustrate the preclinical safety and efficacy of a disease-specific therapy targeting the central oncogenic driver in Ewing sarcoma. Cancer Res; 76(22); 6657-68. ©2016 AACR.


Subject(s)
Camptothecin/analogs & derivatives , Oncogene Proteins, Fusion/genetics , Oncogene Proteins/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/drug therapy , Animals , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Female , Humans , Irinotecan , Mice , Mice, Nude , Sarcoma, Ewing/pathology
6.
Dis Model Mech ; 9(12): 1461-1471, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27780828

ABSTRACT

We explored whether the combination of lurbinectedin (PM01183) with the antimetabolite gemcitabine could result in a synergistic antitumor effect in pancreatic ductal adenocarcinoma (PDA) mouse models. We also studied the contribution of lurbinectedin to this synergism. This drug presents a dual pharmacological effect that contributes to its in vivo antitumor activity: (i) specific binding to DNA minor grooves, inhibiting active transcription and DNA repair; and (ii) specific depletion of tumor-associated macrophages (TAMs). We evaluated the in vivo antitumor activity of lurbinectedin and gemcitabine as single agents and in combination in SW-1990 and MIA PaCa-2 cell-line xenografts and in patient-derived PDA models (AVATAR). Lurbinectedin-gemcitabine combination induced a synergistic effect on both MIA PaCa-2 [combination index (CI)=0.66] and SW-1990 (CI=0.80) tumor xenografts. It also induced complete tumor remissions in four out of six patient-derived PDA xenografts. This synergism was associated with enhanced DNA damage (anti-γ-H2AX), cell cycle blockage, caspase-3 activation and apoptosis. In addition to the enhanced DNA damage, which is a consequence of the interaction of the two drugs with the DNA, lurbinectedin induced TAM depletion leading to cytidine deaminase (CDA) downregulation in PDA tumors. This effect could, in turn, induce an increase of gemcitabine-mediated DNA damage that was especially relevant in high-density TAM tumors. These results show that lurbinectedin can be used to develop 'molecularly targeted' combination strategies.


Subject(s)
Adenocarcinoma/drug therapy , Carbolines/therapeutic use , Deoxycytidine/analogs & derivatives , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Macrophages/pathology , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Carbolines/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cytidine Deaminase/metabolism , DNA Damage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Models, Animal , Down-Regulation/drug effects , Drug Synergism , Enzyme Activation/drug effects , Female , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice, Nude , Pancreatic Neoplasms/pathology , Treatment Outcome , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
7.
Mol Cancer Ther ; 15(10): 2399-2412, 2016 10.
Article in English | MEDLINE | ID: mdl-27630271

ABSTRACT

We have defined the mechanism of action of lurbinectedin, a marine-derived drug exhibiting a potent antitumor activity across several cancer cell lines and tumor xenografts. This drug, currently undergoing clinical evaluation in ovarian, breast, and small cell lung cancer patients, inhibits the transcription process through (i) its binding to CG-rich sequences, mainly located around promoters of protein-coding genes; (ii) the irreversible stalling of elongating RNA polymerase II (Pol II) on the DNA template and its specific degradation by the ubiquitin/proteasome machinery; and (iii) the generation of DNA breaks and subsequent apoptosis. The finding that inhibition of Pol II phosphorylation prevents its degradation and the formation of DNA breaks after drug treatment underscores the connection between transcription elongation and DNA repair. Our results not only help to better understand the high specificity of this drug in cancer therapy but also improve our understanding of an important transcription regulation mechanism. Mol Cancer Ther; 15(10); 2399-412. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Aquatic Organisms/chemistry , Biological Products/pharmacology , DNA Breaks , RNA Polymerase II/metabolism , Animals , Antineoplastic Agents/chemistry , Biological Products/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Transcription, Genetic , Transcriptional Activation , Ubiquitin/metabolism , Xenograft Model Antitumor Assays
8.
Oncotarget ; 6(22): 18875-90, 2015 08 07.
Article in English | MEDLINE | ID: mdl-26056084

ABSTRACT

Recent preclinical evidence has suggested that Ewing Sarcoma (ES) bearing EWSR1-ETS fusions could be particularly sensitive to PARP inhibitors (PARPinh) in combination with DNA damage repair (DDR) agents. Trabectedin is an antitumoral agent that modulates EWSR1-FLI1 transcriptional functions, causing DNA damage. Interestingly, PARP1 is also a transcriptional regulator of EWSR1-FLI1, and PARPinh disrupts the DDR machinery. Thus, given the impact and apparent specificity of both agents with regard to the DNA damage/DDR system and EWSR1-FLI1 activity in ES, we decided to explore the activity of combining PARPinh and Trabectedin in in vitro and in vivo experiments. The combination of Olaparib and Trabectedin was found to be highly synergistic, inhibiting cell proliferation, inducing apoptosis, and the accumulation of G2/M. The drug combination also enhanced γH2AX intranuclear accumulation as a result of DNA damage induction, DNA fragmentation and global DDR deregulation, while EWSR1-FLI1 target expression remained unaffected. The effect of the drug combination was corroborated in a mouse xenograft model of ES and, more importantly, in two ES patient-derived xenograft (PDX) models in which the tumors showed complete regression. In conclusion, the combination of the two agents leads to a biologically significant deregulation of the DDR machinery that elicits relevant antitumor activity in preclinical models and might represent a promising therapeutic tool that should be further explored for translation to the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dioxoles/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Sarcoma, Ewing/drug therapy , Tetrahydroisoquinolines/pharmacology , Animals , Cell Line, Tumor , Child , DNA Damage , Dioxoles/administration & dosage , Drug Synergism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Phthalazines/administration & dosage , Piperazines/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Random Allocation , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Tetrahydroisoquinolines/administration & dosage , Trabectedin , Xenograft Model Antitumor Assays
9.
Mar Drugs ; 11(3): 944-59, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23519149

ABSTRACT

Elisidepsin (PM02734, Irvalec®) is a synthetic marine-derived cyclic peptide of the Kahalalide F family currently in phase II clinical development. Elisidepsin was shown to induce rapid oncosis in ErbB3-expressing cells. Other predictive factors of elisidepsin sensitivity remained unknown. A panel of 23 cancer cell lines of different origin was assessed for elisidepsin cytotoxicity and correlated with mutational state, mRNA and protein expression of selected genes. Elisidepsin showed potent and broad cytotoxic effects in our cancer cell line panel, being active at concentrations ranging from 0.4 to 2 µM that may be relevant for clinical settings. We have shown that elisidepsin is more active in cells harboring epithelial phenotype with high E-cadherin and low vimentin expression. In addition, high ErbB3 and Muc1 expression was correlated with sensitivity to elisidepsin, whereas the presence of KRAS activating mutations was associated with resistance. In DU-PM cells with acquired resistance to elisidepsin, ErbB3 expression was decreased, while Bcl2 was increased. DU-PM cells displayed higher sensitivity to ErbB1-inhibitors suggesting possible cross-talk of ErbB1 and ErbB3 signaling pathways. Combinations of elisidepsin with lapatinib and several chemotherapies including 5-FU and oxaliplatin resulted in synergistic effects that offer the potential of clinical use of elisidepsin in combination settings.


Subject(s)
Antineoplastic Agents/pharmacology , Depsipeptides/pharmacology , Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Depsipeptides/administration & dosage , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/pathology , RNA, Messenger/metabolism , Receptor Cross-Talk , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
10.
Eur J Cell Biol ; 90(10): 872-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21820201

ABSTRACT

Numerous works have questioned the pertinence of using ßII- and/or ßIII-tubulin expression as markers of prognosis and/or prediction of breast cancer response to chemotherapy containing microtubule-targeting agents. The rationale of such studies was essentially based on microtubule dynamics analysis using purified tubulin in vitro and cancer cell lines. Nonetheless, the significance of ßII- and ßIII-tubulin expression in the control of microtubule dynamics in normal mammary epithelium has never been addressed. Here we investigate the expression and the consequences of ßII- and/or ßIII-tubulin depletion in interphase microtubule dynamics in non-tumor human mammary epithelial cells. We find that both isoforms contribute to the tubulin isotype composition in primary and immortalized human mammary epithelial cells. Moreover, while ßII-tubulin depletion has limited effects on interphase microtubule behavior, ßIII-tubulin depletion causes a strong exclusion of microtubules from lamella and a severe suppression of dynamic instability. These results demonstrate that, while ßII-tubulin is dispensable, ßIII-tubulin is required for interphase microtubule dynamics in untransformed mammary epithelial cells. This strongly suggests that ßIII-tubulin is an essential regulator of interphase microtubule functions in normal breast epithelium cells.


Subject(s)
Interphase , Mammary Glands, Human/cytology , Microtubules/metabolism , Tubulin/metabolism , Cells, Cultured , Female , Gene Knockdown Techniques , Humans , Primary Cell Culture , RNA Interference , Single-Cell Analysis , Tubulin/genetics
11.
Eur J Cell Biol ; 90(8): 631-41, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21561680

ABSTRACT

Despite its frequent inactivation in human breast cancers, the role of p21(Cip1) (p21) in morphological plasticity of normal mammary epithelial cells is still poorly understood. To address this question, we have investigated the consequences of p21 silencing in two-dimensional (2D) morphogenesis of untransformed human mammary epithelial cells. Here we show that p21 inactivation causes a reduction of 2D cell spreading and suppresses focal adhesion. In order to investigate the cytoskeletal modifications associated with this altered morphology, we have analyzed the microtubule dynamics in interphase p21-depleted cells. Our results demonstrate that interphase microtubule dynamic instability is strongly increased by p21 silencing. This alteration correlates with severe microtubule hypoacetylation. Next, we show that these microtubule defects in p21-depleted cells can be reversed by the use of the small molecule tubacin, a specific inhibitor of the α-tubulin deacetylase HDAC6. Tubacin-induced microtubule dynamics decrease also correlates with a partial recovery of cell spreading and focal adhesion in those cells. Collectively, these data indicate that p21 regulates the morphological plasticity of normal mammary epithelial cells by modulating dynamics of key cytoskeletal components.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/metabolism , Focal Adhesions/metabolism , Interphase , Mammary Glands, Human/metabolism , Microtubules/metabolism , Anilides/pharmacology , Cell Adhesion , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cytoskeleton/metabolism , DNA/biosynthesis , Epithelial Cells/cytology , Gene Knockdown Techniques , Gene Silencing , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Mammary Glands, Human/cytology , rho GTP-Binding Proteins/antagonists & inhibitors
12.
Cancer Res ; 70(6): 2235-44, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215499

ABSTRACT

Inflammatory mediators present in the tumor milieu may promote cancer progression and are considered promising targets of novel biological therapies. We previously reported that the marine antitumor agent trabectedin, approved in Europe in 2007 for soft tissue sarcomas and in 2009 for ovarian cancer, was able to downmodulate the production of selected cytokines/chemokines in immune cells. Patients with myxoid liposarcoma (MLS), a subtype characterized by the expression of the oncogenic transcript FUS-CHOP, are highly responsive to trabectedin. The drug had marked antiproliferative effects on MLS cell lines at low nanomolar concentrations. We tested the hypothesis that trabectedin could also affect the inflammatory mediators produced by cancer cells. Here, we show that MLS express several cytokines, chemokines, and growth factors (CCL2, CCL3, CCL5, CXCL8, CXCL12, MIF, VEGF, SPARC) and the inflammatory and matrix-binder protein pentraxin 3 (PTX3), which build up a prominent inflammatory environment. In vitro treatment with noncytotoxic concentrations of trabectedin selectively inhibited the production of CCL2, CXCL8, IL-6, VEGF, and PTX3 by MLS primary tumor cultures and/or cell lines. A xenograft mouse model of human MLS showed marked reduction of CCL2, CXCL8, CD68+ infiltrating macrophages, CD31+ tumor vessels, and partial decrease of PTX3 after trabectedin treatment. Similar findings were observed in a patient tumor sample excised after several cycles of therapy, indicating that the results observed in vitro might have in vivo relevance. In conclusion, trabectedin has dual effects in liposarcoma: in addition to direct growth inhibition, it affects the tumor microenvironment by reducing the production of key inflammatory mediators.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dioxoles/pharmacology , Inflammation Mediators/metabolism , Liposarcoma, Myxoid/drug therapy , Tetrahydroisoquinolines/pharmacology , Animals , Antigens, CD/biosynthesis , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/immunology , C-Reactive Protein/biosynthesis , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Chemokine CCL2/biosynthesis , Humans , Immunohistochemistry , Inflammation Mediators/immunology , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Liposarcoma, Myxoid/immunology , Liposarcoma, Myxoid/metabolism , Liposarcoma, Myxoid/pathology , Macrophages/immunology , Mice , Serum Amyloid P-Component/biosynthesis , Trabectedin , Vascular Endothelial Growth Factor A/biosynthesis , Xenograft Model Antitumor Assays
13.
Biochem Pharmacol ; 78(2): 162-70, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19427997

ABSTRACT

Zalypsis is a new synthetic alkaloid tetrahydroisoquinoline antibiotic that has a reactive carbinolamine group. This functionality can lead to the formation of a covalent bond with the amino group of selected guanines in the DNA double helix, both in the absence and in the presence of methylated cytosines. The resulting complex is additionally stabilized by the establishment of one or more hydrogen bonds with adjacent nucleotides in the opposite strand as well as by van der Waals interactions within the minor groove. Fluorescence-based thermal denaturation experiments demonstrated that the most favorable DNA triplets for covalent adduct formation are AGG, GGC, AGC, CGG and TGG, and these preferences could be rationalized on the basis of molecular modeling results. Zalypsis-DNA adducts eventually give rise to double-strand breaks, triggering S-phase accumulation and apoptotic cell death. The potent cytotoxic activity of Zalypsis was ascertained in a 24 cell line panel. The mean IC(50) value was 7nM and leukemia and stomach tumor cell lines were amongst the most sensitive. Zalypsis administration in four murine xenograft models of human cancer demonstrates significant tumor growth inhibition that is highest in the Hs746t gastric cancer cell line with no weight loss of treated animals. Taken together, these results indicate that the potent antitumor activity of Zalypsis supports its current development in the clinic as an anticancer agent.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Mice, Nude , Neoplasms/drug therapy , Tetrahydroisoquinolines/pharmacology , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Dioxoles/chemistry , Dioxoles/pharmacology , Dioxoles/therapeutic use , Drug Screening Assays, Antitumor/methods , Humans , Mice , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/therapeutic use , Trabectedin , Xenograft Model Antitumor Assays/methods
14.
Biochem Pharmacol ; 75(4): 857-65, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18078872

ABSTRACT

Nucleoside transporters (NTs) are essential for the uptake of therapeutic nucleoside analogs, broadly used in cancer treatment. The mechanisms responsible for NT regulation are largely unknown. IL-4 is a pro-survival signal for chronic lymphocytic leukemia (CLL) cells and has been shown to confer resistance to nucleoside analogs. The aim of this study was to investigate whether IL-4 is able to modulate the expression and function of the human equilibrative NT1 (hENT1) in primary cultures of CLL cells and, consequently, to affect cytotoxicity induced by therapeutic nucleosides analogs. We found that treatment with IL-4 (20 ng/ml for 24 h) increased mRNA hENT1 expression in CLL cells without affecting that of normal B cells. Given that the enhanced mRNA levels of hENT1 in CLL cells did not result in increased transport activity, we examined the possibility that hENT1 induced by IL-4 may require post-translational modifications to become active. We found that the acute stimulation of PKC in IL-4-treated CLL cells by short-term incubation with PMA significantly increased hENT1 transport activity and favoured fludarabine-induced apoptosis. By contrast, and in line with previous reports, IL-4 plus PMA protected CLL cells from a variety of cytotoxic agents. Our findings indicate that the combined treatment with IL-4 and PMA enhances hENT1 activity and specifically sensitizes CLL cells to undergo apoptosis induced by fludarabine.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes , Equilibrative Nucleoside Transporter 1/metabolism , Interleukin-4/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell , Tetradecanoylphorbol Acetate/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Humans , Interleukin-4/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Vidarabine/analogs & derivatives , Vidarabine/pharmacokinetics , Vidarabine/pharmacology
15.
Leuk Res ; 31(12): 1649-58, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17475325

ABSTRACT

p16(ink4a) is known to be a major inhibitor of cyclin-dependent kinases of G1-phase. Its accumulation is associated with replicative senescence. We analyzed to what extent the number of cell doublings may participate to p16(ink4a) expression in normal and malignant lymphocytes. p16(ink4a) expression, not found in normal quiescent B or T-lymphocytes, was observed after stimulation of B-lymphocytes (72 h) and T-lymphocytes (2 weeks) before the occurrence of replicative senescence markers such as senescence-associated-beta-galactosidase activity. Afterwards, in lymphocyte long-term cultures, the increase in p16(ink4a) followed the expression of features of cell ageing. In acute lymphoblastic leukemia, the analysis of the individual differences between peripheral blood and blood compartments (34 cases) showed a decrease in cell proliferation (p<0.005), in telomerase activity (p<0.0005), and in hTERT expression (p<0.04), associated with an increase of p16(ink4a) (p<0.035) in blood leukemic cells. These results support the hypothesis that (i) an increase in p16(ink4a) expression in normal lymphocytes is linked, in part, to the number of cell doublings before the occurrence of replicative senescence and (ii) this process is maintained in leukemic cell populations of numerous patients.


Subject(s)
Cell Division , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation/physiology , Lymphocytes/cytology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adolescent , Cell Proliferation , Cellular Senescence , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Telomerase/genetics , Telomerase/metabolism
16.
Crit Rev Oncol Hematol ; 58(3): 190-207, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16690321

ABSTRACT

Loss of p53 function compromises genetic homeostasis in cells exhibiting deregulated DNA replication and/or DNA damage, and prevents normal cytotoxic responses to cancer therapies. Genetic and pharmacological approaches are being developed with the ultimate goal of restoring or controlling p53 functions in cancer patients. Progress has recently been made in the clinical use of replication-deficient virus carrying wt-TP53 (Ad5CMV-p53) and/or cancer-selective oncolytic adenoviruses (ONYX-015). These strategies demonstrated clinical activity as monotherapy and were synergistic with traditional chemotherapy agents in the treatment of some types of cancer. In addition, pharmacological methods are under development to either stimulate wild-type p53 protein function, or induce p53 mutant proteins to resume wild-type functions. These methods are based on small chemicals (CP-31388, PRIMA-1), peptides (CDB3) or single-chain Fv antibody fragments corresponding to defined p53 domains. Here, we discuss the mechanisms underlying these approaches and their perspectives for cancer therapy.


Subject(s)
Adenoviridae , Antineoplastic Agents/therapeutic use , Genetic Therapy , Neoplasms/metabolism , Neoplasms/therapy , Tumor Suppressor Protein p53/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E1B Proteins , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , DNA Damage/genetics , DNA Replication/genetics , Drug Design , Humans , Neoplasms/genetics , Protein Binding/genetics , Protein Structure, Tertiary , Tumor Suppressor Protein p53/genetics , Viral Vaccines
17.
Fam Cancer ; 5(1): 29-34, 2006.
Article in English | MEDLINE | ID: mdl-16528606

ABSTRACT

Cancers arise from the sequential acquisition of genetic alterations in specific genes. The high number of mutations in cancer cells led to the hypothesis that an early step in tumor progression is the generation of a genetic instability. The potent role of genetic instability in initiation and progression of colorectal cancers has been well defined in hereditary nonpolyposis colon cancer (HNPCC) syndrome. HNPCC is a common hereditary disorder caused by germline mutations of DNA mismatch repair (MMR) genes. Somatic loss of the normal allele of the predisposition gene leads to a strong "mutator phenotype", characterized by a high rate of mutations in repetitive sequences. Nevertheless, the observation of frequent alterations of key growth regulatory genes in MMR-deficient cells such as NF1, APC, p53, K-Ras, with no significant excess of frameshift mutations and changes at short coding repeats, suggest that even in the presence of an inherited tendency to genomic instability, tumor progression is mainly driven by a process of natural selection.


Subject(s)
Base Pair Mismatch/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Genetic Predisposition to Disease/epidemiology , Germ-Line Mutation , Neoplasm Proteins/genetics , Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Chromosomal Instability/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , DNA Mutational Analysis , Female , France/epidemiology , Gene Expression Regulation, Neoplastic , Humans , Incidence , Male , Microsatellite Repeats , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Prognosis , Sensitivity and Specificity , Tumor Cells, Cultured
18.
Int J Cancer ; 119(1): 60-6, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16432835

ABSTRACT

We investigated the mechanisms responsible for paclitaxel resistance in HME-1 cells (human mammary epithelial cells immortalized with hTERT). These cells were exposed to paclitaxel (10 pM for 7 days) and 20 cellular surviving populations (PSP) were obtained. PSP demonstrated high levels of resistance to paclitaxel cytotoxicity as compared with HME-1 cells. Activation of mdr-1 gene expression was observed in 2 PSP. Protein expression analysis using a C-terminal targeted antibody showed that 13 PSP were negative for p21/WAF1 expression after ionizing radiation (6 Gy) or doxorubicin (100 nM) treatment. Sequencing of the 3 exons of the CDKN1A gene revealed that 13 PSP contained a point mutation in exon 2. This mutation consisted in a T insertion at codon 104 leading to a premature STOP codon appearance. Mismatch amplification mutation assay and RFLP-PCR confirmed the presence of the mutation in 16 PSP. Western blot using an N-terminal targeted antibody demonstrated that the C-terminal-truncated p21/WAF1 protein (14 kDa) was indeed expressed in the 13 PSP. Our data suggest that p21/WAF1 inactivation may confer a strong resistance to paclitaxel in noncancerous breast epithelial cells harboring a p21/WAF1 mutant.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Mammary Glands, Human/drug effects , Paclitaxel/pharmacology , Point Mutation , Amino Acid Sequence , Base Pair Mismatch , Blotting, Western , Codon , Cyclin-Dependent Kinase Inhibitor p21/radiation effects , Drug Resistance/drug effects , Drug Resistance/radiation effects , Epithelial Cells/drug effects , Female , Gene Expression Regulation , Gene Silencing , Genes, MDR/drug effects , Genes, MDR/radiation effects , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/radiation effects , Molecular Sequence Data , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Radiation Dosage , Radiation, Ionizing , Threonine
19.
Haematologica ; 90(12): 1699-701, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16330448

ABSTRACT

We analyzed the expression of deoxycytidine kinase (dCK), UMP/CMP-kinase (UMP/CMP-K), nucleotide diphosphokinase (NDPK-B) and 5'-nucleotidases cN-II, cN-III, cdN and mdN by quantitative polymerase chain reaction at diagnosis in leukemic blasts from 96 patients with acute myeloid leukemia (AML) treated with ara-C. Our results show that high mRNA levels of cN-II and low mRNA levels of cN-III are correlated with a worse clinical outcome and suggest that these enzymes may have a role in sensitivity to ara-C in AML patients.


Subject(s)
5'-Nucleotidase/genetics , Antimetabolites, Antineoplastic/pharmacokinetics , Cytarabine/pharmacokinetics , Glycoproteins/genetics , Leukemia, Myeloid/enzymology , Neoplasm Proteins/genetics , Neoplastic Stem Cells/enzymology , RNA, Messenger/analysis , RNA, Neoplasm/analysis , 5'-Nucleotidase/biosynthesis , Acute Disease , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Cells/enzymology , Cytarabine/administration & dosage , Drug Resistance, Neoplasm/genetics , Female , Glycoproteins/biosynthesis , Humans , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/genetics , Leukemia, Myeloid/mortality , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Prognosis , Proportional Hazards Models , Pyrimidine Nucleotides/metabolism , RNA, Messenger/blood , RNA, Neoplasm/blood , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...