Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 102
1.
Oncogene ; 30(27): 3011-23, 2011 Jul 07.
Article En | MEDLINE | ID: mdl-21339735

Papillary thyroid carcinoma (PTC) arises from the thyroid follicular epithelium and represents the most frequent thyroid malignancy. PTC is associated with gene rearrangements generating RET/PTC and TRK oncogenes, and to the BRAFV600E activating point mutation. A role of tumor-suppressor genes in the pathogenesis of PTC has not been assessed yet. The tissue inhibitor of metalloproteinase-3 (TIMP3) gene, encoding a metalloproteinases inhibitor and capable of inhibiting growth, angiogenesis, invasion and metastasis of several cancers, was found to be silenced by promoter methylation in a consistent fraction of PTCs, in association with tumor aggressiveness and BRAFV600E mutation, thus suggesting an oncosuppressor role. To explore this possibility, in this study we performed gene expression and functional studies. Analysis of gene expression data produced in our laboratory as well as meta-analysis of publicly available data sets confirmed the downregulation of TIMP3 gene expression in PTC with respect to normal thyroid. The functional consequences of TIMP3 downregulation were investigated in the PTC-derived NIM1 cell line, in which the expression of TIMP3 is silenced. Restoration of TIMP3 expression by exposure to soluble TIMP3 protein or by complementary DNA transfection had no effect on the growth rate of NIM1 cells. Instead, it affected the adhesive, migratory and invasive capabilities of NIM1 cells by modulating several proteins involved in these processes. A striking effect was observed in vivo, as TIMP3 reduced the tumorigenicity of NIM1 cells by repressing angiogenesis and macrophage infiltration. Our data indicate that the loss of TIMP3 expression exerts a functional role in the pathogenesis of PTC.


Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Thyroid Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-3/physiology , Cell Line , Cell Line, Tumor , DNA Methylation , Humans , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-3/genetics
2.
Ann Oncol ; 18(9): 1500-5, 2007 Sep.
Article En | MEDLINE | ID: mdl-17698835

BACKGROUND: The novel adamantyl retinoid ST1926 is a potent inducer of apoptosis in ovarian carcinoma cells. Since the pro-apoptotic effect is associated with activation of p53, in this study we have investigated the efficacy of combination of ST1926 with cisplatin, a DNA-damaging agent that is known to induce p53-dependent apoptosis. MATERIALS AND METHODS: The efficacy of ST1926 and its combination with cisplatin was evaluated in human ovarian carcinoma models, including resistant tumors. RESULTS: Oral treatment with ST1926 alone caused a marginal tumor growth inhibition (<50%), but the combination with cisplatin resulted in an improved efficacy, most evident in terms of tumor growth delay without a substantial increase of toxicity. The combination therapy achieved the best effects against the HOC18 ovarian carcinoma tumor, resulting in an appreciable number of animals without evidence of disease at the end of the experiment. In contrast to the marginal effect of ST1926 alone against the subcutaneous-growing tumors, loco-regional (intraperitoneal) treatment achieved a marked increase of survival of animals with ascitic IGROV-1 tumor. CONCLUSIONS: The present results document the efficacy of the combination of cisplatin with ST1926 and provide a rational basis for the design of novel, well-tolerated platinum-based treatment approaches in human ovarian carcinoma.


Adamantane/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cinnamates/administration & dosage , Cisplatin/administration & dosage , Ovarian Neoplasms/drug therapy , Adamantane/administration & dosage , Animals , Female , Humans , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
3.
Br J Cancer ; 90(7): 1464-8, 2004 Apr 05.
Article En | MEDLINE | ID: mdl-15054472

IDN 5390 is a novel C-seco taxane analogue selected for preclinical development on the basis of its antimotility activity on endothelial cells, antitumour efficacy in a large panel of human tumour xenografts and high tolerability in mouse. On the basis of oral availability, IDN 5390 is suitable for protracted administration schedules. Such a treatment schedule has been reported as the most appropriate to exploit the antiangiogenic effects of cytotoxic drugs. An ability to downregulate angiogenesis-related growth factors in tumour cells has been described for IDN 5390. The aim of the study was to investigate the antitumour and antiangiogenic potential of oral IDN 5390 on a human ovarian carcinoma xenograft, the INT.ACP/PTX, resistant to paclitaxel (PTX). Such tumour line was derived in vivo from a cisplatin-resistant tumour line, the A2780/DDP, which is sensitive to PTX. Compared to the parental cells, INT.ACP/PTX cells exhibited a high level of Pgp expression, resulting in a reduced in vitro sensitivity to both PTX and IDN 5390. The INT.ACP/PTX tumour xenograft was still resistant to PTX, but responsive to IDN 5390, when delivered per os, by a daily prolonged schedule. A direct effect on tumour cells, allowed by the high tolerability of the compound in mouse, cannot be excluded in vivo. Immunohistochemical analysis indicated a significant reduction of microvessel density in IDN 5390-treated tumours, lasting till 7 days after the last drug administration. Thus, a prolonged inhibitory effect on tumour angiogenesis is consistent with the persistent growth control of INT.ACP/PTX tumour achieved by IDN 5390. On the contrary, the low tolerability and the limited oral availability of conventional taxanes do not allow an easy feasibility of such treatment regimen. Thus, the tolerability profile of IDN 5390 in preclinical systems and its efficacy in PTX-resistant tumours support the therapeutic interest for its clinical development, with particular attention to oral daily prolonged schedules.


Angiogenesis Inhibitors/therapeutic use , Bridged-Ring Compounds/therapeutic use , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Taxoids/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous
4.
Cell Death Differ ; 11(3): 280-9, 2004 Mar.
Article En | MEDLINE | ID: mdl-14657960

To understand the molecular mechanisms mediating apoptosis induction by a novel atypical retinoid, ST1926, the cellular response to drug treatment was investigated in IGROV-1 ovarian carcinoma cells carrying wild-type p53 and a cisplatin-resistant p53 mutant subline (IGROV-1/Pt1). Despite a similar extent of drug-induced DNA strand breaks, the level of apoptosis was substantially higher in p53 wild-type cells. p53 activation and early upregulation of p53-target genes were consistent with p53-dependent apoptosis in IGROV-1 cells. Stress-activated protein kinases were activated in both cell lines in response to ST1926. This event and activation of AP-1 were more pronounced in IGROV-1/Pt1 cells, in which the modulation of DNA repair-associated genes suggests an increased ability to repair DNA damage. Inhibition of JNK or p38 stimulated ST1926-induced apoptosis only in IGROV-1 cells, whereas inhibition of ERKs enhanced apoptosis in both the cell lines. Such a pattern of cellular response and modulation of genes implicated in DNA damage response supports that the genotoxic stress is a critical event mediating drug-induced apoptosis. The results are consistent with apoptosis induction through p53-dependent and -independent pathways, regulated by MAP kinases, which likely play a protective role.


Adamantane/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma/drug therapy , Cinnamates/pharmacology , Ovarian Neoplasms/drug therapy , Stress, Physiological , Adamantane/analogs & derivatives , Adamantane/toxicity , Antineoplastic Agents/toxicity , Blotting, Western , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Caspases/drug effects , Caspases/metabolism , Cell Division/drug effects , Cell Line, Tumor , Cinnamates/toxicity , DNA Damage/drug effects , DNA Repair , DNA, Neoplasm/analysis , DNA, Neoplasm/drug effects , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Molecular Structure , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proliferating Cell Nuclear Antigen/metabolism , Transcription Factor AP-1/metabolism , Tumor Suppressor Protein p53/metabolism
5.
Cell Mol Life Sci ; 60(7): 1449-59, 2003 Jul.
Article En | MEDLINE | ID: mdl-12943231

Genetic alterations causing oncogenic activation of the RET gene are recognized as pathogenic events in papillary and medullary thyroid carcinomas. Inhibition of Ret oncoprotein functions could thereby represent a specific therapeutic approach. We previously described the inhibitory activity of the 2-indolinone derivative RPI-1 (formerly Cpdl) on the tyrosine kinase activity and transforming ability of the products of the RET/PTC1 oncogene exogenously expressed in murine cells. In the present study, we investigated the effects of RPI-1 in the human papillary thyroid carcinoma cell line TPC-1 spontaneously harboring the RET/PTC1 rearrangement. Treatment with RPI-1 inhibited cell proliferation and induced accumulation of cells at the G2 cell cycle phase. In treated cells, Ret/Ptc1 tyrosine phosphorylation was abolished along with its binding to Shc and phospholipase C(gamma), thereby indicating abrogation of constitutive signaling mediated by the oncoprotein. Activation of JNK2 and AKT was abolished, thus supporting the drug inhibitory efficacy on downstream pathways. In addition, cell growth inhibition was associated with a reduction in telomerase activity by nearly 85%. These findings in a cellular context relevant to the pathological function of RET oncogenes support the role of Ret oncoproteins as useful targets for therapeutic intervention, and suggest RPI-1 as a promising candidate for preclinical development in the treatment of thyroid tumors expressing RET oncogenes.


Carcinoma, Papillary/pathology , Cell Division/drug effects , Indoles/pharmacology , Oncogene Proteins/antagonists & inhibitors , Proteins , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thyroid Neoplasms/pathology , Animals , Cell Cycle/drug effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Humans , Intracellular Signaling Peptides and Proteins , Kinetics , Membrane Proteins , Mice , Oncogene Proteins, Fusion , Patched Receptors , Patched-1 Receptor , Protein-Tyrosine Kinases , Proto-Oncogene Proteins c-ret , Receptors, Cell Surface , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Telomerase/genetics , Tumor Cells, Cultured
6.
Br J Cancer ; 88(6): 965-72, 2003 Mar 24.
Article En | MEDLINE | ID: mdl-12644838

The recognition of the antiangiogenic properties of taxanes provides a basis for novel therapeutic approaches. A prolonged exposure to low drug concentrations has been proposed to be the most suitable approach to exploit the antiangiogenic potential of cytotoxic agents. Such schedule is required to target preferentially slowly dividing endothelial cells. The protracted use of taxanes could benefit from the availability of a taxane endowed with a favourable tolerability profile. Among compounds of a novel series of C-seco taxanes, IDN 5390 was originally selected on the basis of its potent antimotility activity and poor cytotoxicity on endothelial cells. The aim of the study was to investigate the preclinical pharmacologic profile of IDN 5390 in a variety of human tumour xenografts, including ovarian and colon carcinoma and a glioblastoma. IDN 5390, delivered by s.c. injection, daily for 5 days per week, exhibited a high activity against all tumours investigated (tumour growth inhibition was always >85%) in the range of well-tolerated doses. The maximum tolerated dose/injection (MTD), with no signs of systemic or local vesicant toxicity, was 120 mg kg(-1). In contrast, paclitaxel, delivered according to the same schedule, exhibited a variable antitumour efficacy associated with a substantial local toxicity (MTD=10 mg kg(-1)). Considering the remarkable efficacy of IDN 5390 delivered s.c. by protracted treatment schedule, the oral route of administration was further investigated, as the most suitable for daily treatment. Indeed, a good bioavailability of oral IDN 5390 was found. Oral IDN 5390 maintained a substantial efficacy against human tumour xenografts, including paclitaxel-resistant tumours, without loss of potency with respect to s.c. administration. In conclusion, the therapeutic advantages of IDN 5390, over paclitaxel, in protracted daily treatment schedules are represented by the oral efficacy and the high tolerability, which are favourable features to exploit the antiangiogenic potential and to design combinations with other effective agents.


Brain Neoplasms/drug therapy , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/pharmacology , Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Glioblastoma/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Administration, Oral , Animals , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Mice , Mice, Nude , Neoplasms, Experimental , Taxoids , Transplantation, Heterologous
7.
Cell Death Differ ; 9(12): 1352-9, 2002 Dec.
Article En | MEDLINE | ID: mdl-12478472

Mononuclear and multinuclear platinum complexes are known to induce distinct types of DNA lesions and exhibit different profiles of antitumor activity, in relation to p53 mutational status. In this study, we investigated the cellular effects of exposure to two platinum compounds (cisplatin and the multinuclear platinum complex BBR 3464), in the osteosarcoma cell line, U2-OS, carrying the wild-type p53 gene and capable of undergoing apoptosis or cell cycle arrest in response to diverse genotoxic stresses. In spite of the ability of both compounds to up-regulate p53 at cytotoxic concentrations, exposure to BBR 3464 resulted in cell cycle arrest but only cisplatin was capable of inducing significant levels of apoptosis and phosphorylation at the Ser15 residue of p53. The cisplatin-induced protein phosphorylation, not detectable in cells treated with BBR 3464, was associated with RPA phosphorylation, a specific up-regulation of Bax and down-regulation of p21(WAF1). Cells treated with BBR 3464 displayed a different cellular response with evidence of cytostasis associated with a high induction of p21(WAF1). The regulation of p21(WAF1) after cisplatin or BBR 3464 exposure required a p53 signal, as documented using stable transfectants expressing a dominant-negative form of p53 (175(his)). Taken together, these results indicate that cellular response to different genotoxic lesions (i.e. apoptosis or growth arrest) is associated with a specific recognition of DNA damage and a different p53-mediated signaling pathway. Multinuclear platinum complexes could be regarded as useful tools for investigating the p53-mediated process of cell cycle arrest in response to DNA damage.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Damage/drug effects , Neoplasms/drug therapy , Platinum Compounds/pharmacology , Tumor Suppressor Protein p53/drug effects , Amino Acid Sequence/drug effects , Amino Acid Sequence/physiology , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cisplatin/pharmacology , Cytotoxins/pharmacology , DNA Damage/physiology , Humans , Neoplasms/metabolism , Neoplasms/physiopathology , Phosphorylation/drug effects , Serine/drug effects , Serine/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Cell Mol Life Sci ; 59(8): 1406-12, 2002 Aug.
Article En | MEDLINE | ID: mdl-12363043

Stable transfection of human ovarian carcinoma cells with survivin cDNA caused a four- to sixfold increase in cell resistance to taxotere and taxol (two-sided Student's t test, p < 0.05), with a concomitant reduction in the apoptotic response to taxol, but did not affect cell sensitivity to cisplatin or oxaliplatin. Such findings were indirectly supported by similar observations obtained with clinical tumours. In fact, high levels of survivin protein expression (>30% positive cells), detected by immunohistochemistry in 90/124 (73%) advanced ovarian carcinomas, were significantly associated with clinical resistance to a taxol/platinum-based regimen but unrelated to tumour shrinkage following cisplatin-including combinations (non-taxol based). In the 95 patients receiving a taxol/platinum-based regimen, survivin overexpression correlated with a lower clinical or pathologic complete remission rate than absent/low protein expression (43 vs 75%, p = 0.0058 by logistic regression adjusted for tumour stage, histological grade and p53 expression). Conversely, in the 29 cases treated with cisplatin-containing regimens (not taxol based), survivin expression was unrelated to tumour response. Cellular studies and clinical data suggest a direct link between survivin expression and tumour cell susceptibility to taxol.


Antineoplastic Agents, Phytogenic/pharmacology , Chromosomal Proteins, Non-Histone/genetics , Drug Resistance, Neoplasm/genetics , Microtubule-Associated Proteins , Ovarian Neoplasms/genetics , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Taxoids , Apoptosis/physiology , Chromosomal Proteins, Non-Histone/physiology , Cisplatin/pharmacology , Docetaxel , Female , Humans , In Situ Nick-End Labeling , Inhibitor of Apoptosis Proteins , Mutation , Neoplasm Proteins , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Oxaliplatin , Phosphorylation/drug effects , Phosphotransferases/drug effects , Survivin , Transfection , Tumor Cells, Cultured
9.
Carcinogenesis ; 22(12): 1971-8, 2001 Dec.
Article En | MEDLINE | ID: mdl-11751427

The tumour suppressor gene TP53 plays an important role in the regulation of DNA repair, and particularly of nucleotide excision repair. The influence of p53 status on the efficiency of the principal steps of this repair pathway was investigated after UV-C irradiation in the human ovarian carcinoma cell line IGROV-1 (expressing wild-type p53) and in the derived clone IGROV-1/Pt1 (with p53 mutations at codons 270 and 282). Clonogenic survival after UV-C irradiation showed that IGROV-1/Pt1 cells were approximately 2-fold more resistant to DNA damage than parental cells. Modulation of p53 protein levels, cell cycle arrest and apoptosis were induced in UV-irradiated IGROV-1 cells, but not in the p53-mutant cell line. Exposure to UV or cisplatin induced down-regulation of p53-replication protein A (RPA) interaction in parental, but not in IGROV-1/Pt1 cells. However, persistent binding of p53 to RPA did not affect the early steps of DNA repair. In fact, both UV-induced DNA incision and the recruitment of proliferating cell nuclear antigen (PCNA) to DNA repair sites occurred to a comparable extent in p53-wild type and -mutant cell lines, although PCNA remained associated with chromatin for a longer period of time in IGROV-1/Pt1 cells. Global genome repair, as detected by immunoblot analysis of cyclobutane pyrimidine dimers, was not significantly different in the two cell lines at 3 h after UV irradiation. In contrast, lesion removal at 24 h was markedly reduced in IGROV-1/Pt1 cells, being approximately 25% of the initial amount of damage, as compared with approximately 50% repair in parental cells. These results indicate that the presence of mutant p53 protein and its persistent interaction with RPA do not affect the early steps of nucleotide excision repair in IGROV-1/Pt1 cells. Thus, repair defects in p53-mutant ovarian carcinoma cells may be attributed to late events, possibly related to a reduced removal/recycling of PCNA at repair sites.


DNA Damage/radiation effects , DNA Repair , DNA-Binding Proteins/metabolism , Ovarian Neoplasms/genetics , Proliferating Cell Nuclear Antigen/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Blotting, Western , Cell Survival/radiation effects , DNA Repair/genetics , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Point Mutation/genetics , Replication Protein A , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
10.
Eur J Cancer ; 37(17): 2247-56, 2001 Nov.
Article En | MEDLINE | ID: mdl-11677115

Based on the role of p53 in the control of apoptosis following DNA damage, the status of the TP53 gene has been implicated as a major determinant of tumour responsiveness to cytotoxic therapies. In spite of the high frequency of TP53 mutations, small-cell lung cancer (SCLC) is recognised as one of the most chemoresponsive solid tumours. Since the relevance of the TP53 gene status in the modulation of tumour responsiveness is dependent on the molecular/biological context, in the present study, we have examined the relationship between chemosensitivity and susceptibility to apoptosis of a TP53-mutant human SCLC cell line. The cell line, in spite of TP53 mutation, retained an efficient response to genotoxic stress as documented by cells ability to modulate the p53 protein, arrest in the G1 and G2 phases of the cell cycle and its marked susceptibility to apoptosis following treatment with DNA damaging agents. Exposure to DNA-damaging agents caused an increase of c-Myc, a DNA damage-responsive transcription factor. An analysis of damage-induced apoptosis in the presence of an anti-Fas/CD95 inhibitory antibody indicated that Fas/CD95 was not required for the apoptotic response. The results support an implication of c-myc in sensitising cells to apoptosis, since inhibition of c-Myc expression with an antisense oligodeoxynucleotide (AS-ODN) almost abolished the drug-induced apoptotic response. In conclusion, the present results support a role for c-myc in the induction of apoptosis by genotoxic stress in the absence of a functional p53 and provide new insights into the mechanisms that may influence apoptosis in TP53-mutant cells. Elucidation of this pathway and of the possible cooperation with p53-dependent mechanisms may provide a basis for therapeutic intervention.


Apoptosis/genetics , Carcinoma, Small Cell/genetics , DNA Damage/genetics , Genes, p53 , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Small Cell/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Doxorubicin/pharmacology , Gamma Rays , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , Mutation , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
11.
Oncology ; 61(3): 234-42, 2001.
Article En | MEDLINE | ID: mdl-11574780

OBJECTIVES: Systematic investigation of a novel series of intercalating agents, 9-aza-anthrapyrazoles, has led to the identification of a promising analogue, BBR 3438. This study describes the antitumour efficacy of the novel compound in human prostate carcinoma models and the molecular/cellular basis of its activity. METHODS AND RESULTS: The novel 9-aza-anthrapyrazole BBR 3438 was significantly more effective than doxorubicin and losoxantrone (DuP-941) in two of the three tested prostate carcinoma models. The superior activity was more evident in PC3 tumour, since BBR 3438 produced an appreciable rate of complete tumour regressions. Under these conditions, the drug-induced antiproliferative activity paralleled delayed apoptosis. Tumour response to in vivo drug treatment was associated with an early down-regulation of Bcl-2, which was somewhat more marked for the aza compound. In fact, the 9-aza-anthrapyrazole induced DNA cleavage in vitro with isolated DNA topoisomerase II (isoform alpha) and DNA strand breaks in prostatic carcinoma cells. Although the molecular effects of losoxantrone and the 9-aza analogue on the enzyme target were comparable, the cytotoxic effects of BBR 3438 could be enhanced by long-term exposure as a consequence of favourable cellular accumulation and prominent DNA-binding affinity. In addition, a lower reduction potential of the 9-aza-anthrapyrazole in comparison with classical anthrapyrazoles suggests an increased ability of the drug to induce oxidative stress following free radical production, which may be a contributing factor in determining the long-term response (i.e. delayed cell death) to genotoxic damage. CONCLUSIONS: BBR 3438 exhibited a unique profile of preclinical activity with a superior efficacy against prostatic carcinoma models compared to reference compounds (doxorubicin and losoxantrone). The antitumour efficacy of BBR 3438 against prostatic carcinoma could be the result of a combination of favourable events, including enhanced intracellular accumulation and an increased DNA-binding affinity favouring the accumulation of multiple sublethal or lethal damage. In spite of its enhanced cytotoxic potency, the 9-aza compound was better tolerated in vivo than losoxantrone, thus improving the therapeutic index. The preclinical profile of efficacy against prostatic carcinoma, a tumour resistant to conventional antitumour drugs, makes the novel 9-aza-anthrapyrazole BBR 3438 a promising candidate for clinical evaluation.


Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Ethanolamines/therapeutic use , Intercalating Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyrazolones , Animals , Anthraquinones/therapeutic use , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , DNA Damage , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/drug effects , DNA-Binding Proteins , Doxorubicin/therapeutic use , Ethanolamines/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, bcl-2 , Humans , Intercalating Agents/pharmacology , Male , Mice , Mice, Nude , Molecular Structure , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Pyrazoles/pharmacology , Remission Induction , Xenograft Model Antitumor Assays
12.
Prostate ; 48(4): 254-64, 2001 Sep 15.
Article En | MEDLINE | ID: mdl-11536305

BACKGROUND: Defects in the cell cycle machinery of prostate cancer cells might impair the efficiency of cell cycle checkpoints and affect the cell response to chemotherapeutic drugs. We examined the relationship between the status of microtubule damage-activated checkpoints and the response of hormone-refractory prostate cancer cells to paclitaxel. METHODS: The two cell lines DU145 and PC3 harboring defects at proteins involved in the regulation of checkpoints activated by microtubule damage were examined for cell sensitivity, apoptotic response, and efficiency of checkpoints in response to paclitaxel. RESULTS: In spite of a comparable sensitivity to the antiproliferative effects of paclitaxel, DU145 and PC3 cells exhibited different cell cycle control at checkpoints activated by microtubule damage. A transient mitotic arrest was induced by the taxane in both cell lines. However, PC3 cells underwent a rapid mitotic slippage and displayed a defective postmitotic checkpoint as evidenced by the appearance of polyploid cells. In this cell line, paclitaxel-induced cell death was a slow and delayed event, occurring also after S-phase re-entry. The mitotic checkpoint appeared to be more stringent in DU145 cells compared to PC3 cells. Moreover, despite the expression of mutated proteins involved in the prevention of DNA endoreduplication (p53, pRb, and p16(INK4A)), these cells did not progress into the cell cycle but efficiently underwent apoptosis by 24 hr. Such a response of DU145 cells was associated with phosphorylation of the p21(WAF1) protein. CONCLUSIONS: These observations evidence that activation of checkpoints following microtubule damage in prostate cancer may be regulated through complex mechanisms possibly involving p21(WAF1). Our findings support that the status of cell cycle checkpoints might affect the modality of cell death. However, the relevance of the mode of cell death for the sensitivity to taxanes remains to be determined.


Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Apoptosis/drug effects , Blotting, Western , CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/metabolism , DNA, Neoplasm/analysis , Flow Cytometry , Humans , Male , Microtubules/drug effects , Microtubules/physiology , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
13.
Histol Histopathol ; 16(3): 719-26, 2001 07.
Article En | MEDLINE | ID: mdl-11510961

The effects of electromagnetic fields on several processes related to cell physiology and proliferation are currently being investigated. Although the results are still not conclusive and even conflicting, there seems to be a fairly good agreement on the early effects of electromagnetic fields on the generation of free radicals and on Ca++-intracellular concentration and transport. To evaluate the long-lasting consequences of these precocious events, we examined the effects of short- and long-term magnetic field exposure on structural organization (cytokeratin or actin detection), proliferation (bromodeoxyuridine incorporation and propidium iodide staining), colony forming ability and viability (trypan blue exclusion test) of highly proliferating MCF-7 cells (from human breast carcinoma) and on slowly proliferating normal human fibroblasts (from healthy donors). Cells were exposed to either 20 or 500 microT sinusoidally oscillating (50Hz) magnetic fields for different lengths of time (1 to 4 days). Short (1 day)- and long (4 days)-time exposure to the two intensities did not affect MCF-7 growth and viability, colony number and size, or cellular distribution along the cell cycle; neither were the cell morphology and the intracellular distribution and amount of cytokeratin modified. Similarly, no modifications in the actin distribution and proliferative potential were observed in normal human fibroblasts. These findings suggest that under our experimental conditions, continuous exposure to magnetic fields does not result in any appreciable effect in both normal and tumor cells in vitro.


Cell Division , Magnetics/adverse effects , Actins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival , DNA, Neoplasm/biosynthesis , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Infant , Keratins/metabolism , Ki-67 Antigen/metabolism , Tumor Cells, Cultured , Tumor Stem Cell Assay
14.
J Cell Biochem ; 82(2): 271-6, 2001.
Article En | MEDLINE | ID: mdl-11527152

The piperidine nitroxide TEMPOL induces apoptosis in a number of tumor cell lines through free radical-dependent mechanisms. As mitochondria play a major role in apoptosis as both source and target for free radicals, the present study focuses on mitochondrial effects of TEMPOL in a human promyelocytic leukemic cell line (HL-60). On 24-h exposure to TEMPOL, the following alterations were observed: 1) decrease in both the intracellular and mitochondrial glutathione pools; 2) impairment of oxidative phosphorylation; and 3) decrease in mitochondrial membrane potential. In addition, TEMPOL was found to specifically target complex I of the respiratory chain, with minor effects on complexes II and IV, suggesting that mitochondrial effects might play a role in TEMPOL-induced oxidative stress and apoptosis, and that TEMPOL might sensitize tumor cells to the pro-apoptotic effects of cytotoxic agents.


Antioxidants/pharmacology , Apoptosis/drug effects , Cyclic N-Oxides/pharmacology , HL-60 Cells/drug effects , Mitochondria/drug effects , Chromatography, High Pressure Liquid , Electron Transport Complex II , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex IV/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , HL-60 Cells/cytology , Humans , Intracellular Membranes/drug effects , Membrane Potentials/drug effects , Multienzyme Complexes/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Oxidoreductases/antagonists & inhibitors , Rotenone/pharmacology , Sodium Azide/pharmacology , Spin Labels , Succinate Dehydrogenase/antagonists & inhibitors
15.
Int J Cancer ; 92(5): 738-47, 2001 Jun 01.
Article En | MEDLINE | ID: mdl-11340581

Loss of p53 function has been linked to increased responsiveness to taxane treatment of ovarian carcinoma in clinical studies. We recently reported that the acquisition of cisplatin resistance in an ovarian carcinoma cell line (IGROV-1) was associated with mutation of p53 and collateral sensitivity to paclitaxel. The increased sensitivity to paclitaxel of the cisplatin-resistant subline appeared to be pharmacologically relevant since it was reflected in an in vivo sensitization to taxanes. To investigate the cellular and molecular basis of this phenomenon, we performed a comparative study of cellular response to taxanes (paclitaxel and the novel analog IDN 5109) in the parental cell line, containing wild-type p53 and its cisplatin-resistant p53 mutant subline (IGROV-1/Pt1). IDN 5109 was included in this study because of its higher potency and efficacy compared with paclitaxel on both tumor systems. The pattern of cellular response of the two ovarian cell lines was different. In IGROV-1 cells, apoptosis was an early event consequent to a transient mitotic arrest. The cell death of IGROV-1/Pt1 cells was a somewhat slow and delayed event, following mitotic arrest and appearance of hyperploid cells. The increased cytotoxic effect of IDN 5109, compared with paclitaxel, was associated with more marked p34(cdc2) dephosphorylation in IGROV-1 cells and higher Bcl-2 phosphorylation in IGROV-1/Pt1 cells after 24 hr of treatment. In each cell line, these biochemical events were not correlated with parallel levels of mitotic cells. Attempts to reintroduce wild-type p53 in IGROV-1/Pt1 were unsuccessful. However, in other p53-deficient cells (osteosarcoma SAOS), taxane treatment was associated with hyperploid progression and the introduction of wild-type p53 resulted in a reduced sensivity. Although our approach does not allow definitive conclusions, these results suggest that loss of p53-dependent post-mitotic checkpoint results in a different time-course of taxane-induced cell death following DNA reduplication. These events, more evident after exposure to the potent analog IDN 5109, support the notion that the enhanced sensitivity of p53 mutant cells is closely related to the different mode of cell death.


Antineoplastic Agents, Phytogenic/pharmacology , Bridged-Ring Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Taxoids , Tumor Suppressor Protein p53/physiology , Apoptosis/drug effects , Cell Cycle/drug effects , DNA, Neoplasm/analysis , Female , Humans , Ovarian Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin/metabolism , Tumor Cells, Cultured
16.
Curr Med Chem ; 8(1): 31-7, 2001 Jan.
Article En | MEDLINE | ID: mdl-11172690

Cellular resistance to anthracyclines is a major limitation of their clinical use in the treatment of human tumors. Resistance to doxorubicin is described as a multifactorial phenomenon involving the overexpression of defense factors and alterations in drug-target interactions. Such changes do not account for all manifestations of drug resistance, in particular intrinsic resistance of solid tumors. Since anthracyclines can induce apoptotic cell death, an alternative promising approach to drug resistance has focused on the study of cellular response to drug-induced DNA damage, with particular reference to the relationship between cytotoxicity/antitumor efficacy and apoptotic response. The evidence that a novel disaccharide analog (MEN 10755), endowed with an improved preclinical activity over doxorubicin, was also more effective as an inducer of apoptosis provided additional insights to better understand the cellular processes that confer sensitivity to anthracyclines. Although the presence or alteration of a single apoptosis-related factor (e.g., p53, bcl-2) is not predictive of the sensitivity/resistance status, the complex interplay among DNA damage-activated pathways is likely an important determinant of tumor cell sensitivity to anthracyclines


Antibiotics, Antineoplastic/therapeutic use , Apoptosis/genetics , Apoptosis/physiology , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Humans , Neoplasms/pathology
17.
Free Radic Biol Med ; 29(7): 633-41, 2000 Oct 01.
Article En | MEDLINE | ID: mdl-11033415

The antiproliferative effect of Tempol, a stable nitroxide free radical, was investigated on the p53-negative human leukemia cell line HL60. A concentration- and time-dependent inhibition of cell growth was observed that appears to be due to induction of apoptosis. Involvement of oxidative stress is indicated by a concentration-dependent increase in intracellular peroxides and a parallel decrease in total cellular glutathione; in addition, increased survival rates were observed in cells simultaneously treated with Tempol and the antioxidant N-acetylcysteine. Tempol did not affect the relative levels of Bax and Bcl2, whereas p21(WAF1/CIP1) was enhanced in a concentration- and time-dependent fashion; this effect was partially inhibited by N-acetylcysteine, was maintained for up to 8 h after Tempol removal, and seemed to depend on continuing protein synthesis. The increase in p21(WAF1/CIP1) was accompanied by a parallel accumulation of cells in the G(1) phase of the cycle and by a decrease in the 110 kDa form of pRb. Our results suggest that p53-independent induction of p21(WAF1/CIP1) mediates the antiproliferative effect of Tempol; on the basis of this observation, the nitroxide could be proposed as an useful adjunct to the treatment of p53-deficient tumors, which are often refractory to standard chemotherapy.


Apoptosis/drug effects , Cell Cycle/drug effects , Cyclic N-Oxides/toxicity , Cyclins/metabolism , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Glutathione/metabolism , HL-60 Cells , Humans , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Spin Labels , bcl-2-Associated X Protein
18.
J Cell Sci ; 113 ( Pt 15): 2671-8, 2000 Aug.
Article En | MEDLINE | ID: mdl-10893182

The metabolism of glutathione by membrane-bound &ggr;-glutamyl transpeptidase (GGT) has been recently recognized as a basal source of hydrogen peroxide in the extracellular space. Significant levels of GGT activity are expressed by malignant tumours, and in melanoma cell lines they were found to correlate with the malignant behaviour. As hydrogen peroxide and other oxidants can affect signal transduction pathways at several levels, the present study was aimed to verify: (i) the occurrence of GGT-dependent production of hydrogen peroxide in melanoma cells; (ii) the effects of GGT-dependent prooxidant reactions on known redox-sensitive cellular targets, i.e. protein thiols, the nuclear transcription factor NF-kappa B and p53. Two melanoma Me665/2 cell clones, exhibiting traces of (clone 2/21) or high (clone 2/60) GGT activity, were studied. The occurrence of GGT-dependent production of hydrogen peroxide was apparent in 2/60 cells, in which it was accompanied by lower levels of cell surface protein thiols. In 2/60 cells, GGT expression was also associated with higher levels of NF-kappa B activation, as compared to GGT-poor 2/21 cell clone. Indeed, stimulation or inhibition of GGT activity in 2/60 cells resulted in progressive activation or inactivation of NF-kappa B, respectively. An analysis of the p53 gene product indicated lack of protein expression in 2/60 cells, whereas a mutant protein was highly expressed in 2/21 cells. Taken together, these results indicate that the expression of GGT activity can provide melanoma cells with an additional source of hydrogen peroxide, and that such prooxidant reactions are capable to modify protein thiols at the cell surface level. In addition, GGT expression results in an up-regulation of the transcription factor NF-kappa B, which could explain the higher metastatic behaviour reported for GGT-rich melanoma cells as compared to their GGT-poor counterparts.


Hydrogen Peroxide/metabolism , Melanoma , NF-kappa B/metabolism , Sulfhydryl Compounds/metabolism , gamma-Glutamyltransferase/metabolism , Blotting, Western , Cell Membrane/enzymology , Clone Cells , Enzyme-Linked Immunosorbent Assay , Humans , Membrane Proteins/analysis , Membrane Proteins/metabolism , Oxidation-Reduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , gamma-Glutamyltransferase/analysis
19.
Clin Cancer Res ; 6(7): 2626-34, 2000 Jul.
Article En | MEDLINE | ID: mdl-10914703

Multinuclear platinum complexes represent a new class of anticancer agents, distinct in terms of DNA binding features and the profile of antitumor activity from their mononuclear counterparts, in particular cisplatin. Among complexes of this class, BBR 3464, a trinuclear platinum compound has been selected for preclinical development. In the present study, we describe the preclinical evaluation of BBR 3464 in a series of human tumor cell lines and tumor xenografts, with special emphasis on tumor types known to be resistant to cisplatin. In a panel of seven human tumor cell lines naturally resistant to cisplatin (three ovarian and four melanomas), BBR 3464 was extremely potent with IC50 values at least 20-fold lower than cisplatin. Against eight human tumor xenografts including four tumors refractory to cisplatin, BBR 3464 was confirmed to be very active with a tumor weight inhibition >80% in seven of them. The efficacy of BBR 3464 against cisplatin-resistant tumors was consistent with the ability of the drug to completely overcome resistance in three cell systems characterized by acquired resistance to cisplatin. Moreover, BBR 3464 caused a more prolonged effect than cisplatin, which was reflected by higher specific growth delay values. This prolonged effect is likely to be related to a more persistent perturbation of the cell cycle induced by BBR 3464 than by cisplatin, as shown in one ovarian tumor cell line. Finally, the profile of sensitivity to BBR 3464 within the 60-cell-lines screening panel of the National Cancer Institute, NIH (Bethesda, MD) differed from those of established drugs, thus supporting the hypothesis of a distinct mechanism of cytotoxic activity of BBR 3464. The novel trinuclear platinum complex, in light of its innovative antitumor activity profile, has the potential to become a useful clinical agent for the treatment of unresponsive tumors.


Antineoplastic Agents/toxicity , Cell Survival/drug effects , Cisplatin/toxicity , Organoplatinum Compounds/toxicity , Ovarian Neoplasms/drug therapy , Animals , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/pathology , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Int J Oncol ; 16(5): 995-1002, 2000 May.
Article En | MEDLINE | ID: mdl-10762636

Since telomerase plays a role in cellular resistance to apoptosis, which is the primary mode of cell death induced by several drugs, telomerase could be involved in determining the chemosensitivity profile of tumor cells. Thus, we investigated the relationship between telomerase activity, telomere length and chemosensitivity to effective antitumor agents in a panel of human melanoma and ovarian cancer cell lines. Telomerase activity, as detected by the telomeric repeat amplification protocol, ranged from 0.58 to 1.10 arbitrary units in individual cell lines, with similar median values for melanoma and ovarian carcinoma cell lines (0.80 vs. 0.90). Telomeres were generally longer in melanoma than in ovarian carcinoma cell lines, with a more than 2-fold median telomere restriction fragment length (7.74 vs. 3.82 kb). No significant correlation was evidenced between the two telomere-related parameters and cell population doubling time, DNA index or TP53 gene status. No precise relation was found between telomerase activity and cellular sensitivity to different DNA damaging agents including doxorubicin, cisplatin and the multinuclear platinum compound BBR 3464. In contrast, longer telomeres were associated to resistance to the drugs, even though the association reached statistical significance only for cisplatin. Since platinum compounds may have affinity for telomere sequences, it is conceivable that the interaction is relevant for drug sensitivity/resistance status depending on telomere length.


Antineoplastic Agents/pharmacology , Melanoma/enzymology , Melanoma/ultrastructure , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/ultrastructure , Telomerase/metabolism , Telomere/ultrastructure , Cell Division , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Female , Humans , Tumor Cells, Cultured
...