14 results on '"de marco, C."'
Search Results
2. A gene expression-based classifier for HER2-low breast cancer.
- Author
-
Di Cosimo S, Pizzamiglio S, Ciniselli CM, Duroni V, Cappelletti V, De Cecco L, De Marco C, Silvestri M, De Santis MC, Vingiani A, Paolini B, Orlandi R, Iorio MV, Pruneri G, and Verderio P
- Subjects
- Humans, Female, Receptor, ErbB-2 metabolism, Genes, erbB-2, Immunohistochemistry, Gene Expression, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms genetics
- Abstract
In clinical trials evaluating antibody-conjugated drugs (ADCs), HER2-low breast cancer is defined through protein immunohistochemistry scoring (IHC) 1+ or 2+ without gene amplification. However, in daily practice, the accuracy of IHC is compromised by inter-observer variability. Herein, we aimed to identify HER2-low breast cancer primary tumors by leveraging gene expression profiling. A discovery approach was applied to gene expression profile of institutional INT1 (n = 125) and INT2 (n = 84) datasets. We identified differentially expressed genes (DEGs) in each specific HER2 IHC category 0, 1+, 2+ and 3+. Principal Component Analysis was used to generate a HER2-low signature whose performance was evaluated in the independent INT3 (n = 95), and in the publicly available TCGA and GSE81538 datasets. The association between the HER2-low signature and HER2 IHC categories was evaluated by Kruskal-Wallis test with post hoc pair-wise comparisons. The HER2-low signature discriminatory capability was assessed by estimating the area under the receiver operating characteristic curve (AUC). Gene Ontology and KEGG analyses were performed to evaluate the HER2-low signature genes functional enrichment. A HER2-low signature was computed based on HER2 IHC category-specific DEGs. The twenty genes included in the signature were significantly enriched with lipid and steroid metabolism pathways, peptidase regulation, and humoral immune response. The HER2-low signature values showed a bell-shaped distribution across IHC categories (low values in 0 and 3+; high values in 1+ and 2+), effectively distinguishing HER2-low from 0 (p < 0.001) to 3+ (p < 0.001). Notably, the signature values were higher in tumors scored with 1+ as compared to 0. The HER2-low signature association with IHC categories and its bell-shaped distribution was confirmed in the independent INT3, TCGA and GSE81538 datasets. In the combined INT1 and INT3 datasets, the HER2-low signature achieved an AUC value of 0.74 (95% confidence interval, CI 0.67-0.81) in distinguishing HER2-low vs. the other categories, outperforming the individual ERBB2 mRNA AUC value of 0.52 (95% CI 0.43-0.60). These results represent a proof-of-concept for an observer-independent gene-expression-based classifier of HER2-low status. The herein identified 20-gene signature shows promise in distinguishing between HER2 0 and HER2-low expressing tumors, including those scored as 1+ at IHC, and in developing a selection approach for ADCs candidates., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. Can we define breast cancer HER2 status by liquid biopsy?
- Author
-
Di Cosimo S, De Marco C, Silvestri M, Busico A, Vingiani A, Pruneri G, and Cappelletti V
- Subjects
- Female, Humans, Biological Evolution, Liquid Biopsy, Breast Neoplasms drug therapy
- Abstract
Human Epidermal growth factor Receptor 2 (HER2) assessment is crucial for breast cancer treatment. Therapeutic decisions for recurrent cases often rely on primary tumor status. However, mounting evidence suggests that tumors show dynamic changes and up to 10% of breast cancer modify their initial status during progression. It is still debated whether these changes reflect a biological evolution of the disease or are secondary to primary tumor heterogeneity. Certainly, repeating HER2 assessment during breast cancer trajectory is important for the increasing availability of effective anti-HER2 drugs. In response to this need, circulating biomarkers such as circulating tumor cells (CTCs) and cell-free circulating tumor DNA (ctDNA) offer the potential to safely and repeatedly assess HER2 status over time. This chapter outlines current methods for testing HER2 in CTCs and ctDNA, and reviews clinical trials evaluating its prognostic and predictive value in patients with breast cancer, as well as recent advances in the field., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
4. Polyurethane foam scaffold as in vitro model for breast cancer bone metastasis.
- Author
-
Angeloni V, Contessi N, De Marco C, Bertoldi S, Tanzi MC, Daidone MG, and Farè S
- Subjects
- Adipose Tissue cytology, Cadherins metabolism, Cell Death, Cell Differentiation, Cell Line, Tumor, Coculture Techniques, Compressive Strength, Female, Humans, Neoplastic Stem Cells pathology, Osteoblasts cytology, Polymers chemistry, Spectrometry, X-Ray Emission, Stem Cells cytology, Bone Neoplasms secondary, Breast Neoplasms pathology, Models, Biological, Polyurethanes chemistry, Tissue Scaffolds chemistry
- Abstract
Breast cancer (BC) represents the most incident cancer case in women (29%), with high mortality rate. Bone metastasis occurs in 20-50% cases and, despite advances in BC research, the interactions between tumor cells and the metastatic microenvironment are still poorly understood. In vitro 3D models gained great interest in cancer research, thanks to the reproducibility, the 3D spatial cues and associated low costs, compared to in vivo and 2D in vitro models. In this study, we investigated the suitability of a poly-ether-urethane (PU) foam as 3D in vitro model to study the interactions between BC tumor-initiating cells and the bone microenvironment. PU foam open porosity (>70%) appeared suitable to mimic trabecular bone structure. The PU foam showed good mechanical properties under cyclic compression (E=69-109kPa), even if lower than human trabecular bone. The scaffold supported osteoblast SAOS-2 cell line proliferation, with no cytotoxic effects. Human adipose derived stem cells (ADSC) were cultured and differentiated into osteoblast lineage on the PU foam, as shown by alizarin red staining and RT-PCR, thus offering a bone biomimetic microenvironment to the further co-culture with BC derived tumor-initiating cells (MCFS). Tumor aggregates were observed after three weeks of co-culture by e-cadherin staining and SEM; modification in CaP distribution was identified by SEM-EDX and associated to the presence of tumor cells. In conclusion, we demonstrated the suitability of the PU foam to reproduce a bone biomimetic microenvironment, useful for the co-culture of human osteoblasts/BC tumor-initiating cells and to investigate their interaction., Statement of Significance: 3D in vitro models represent an outstanding alternative in the study of tumor metastases development, compared to traditional 2D in vitro cultures, which oversimplify the 3D tissue microenvironment, and in vivo studies, affected by low reproducibility and ethical issues. Several scaffold-based 3D in vitro models have been proposed to recapitulate the development of metastases in different body sites but, still, the crucial challenge is to correctly mimic the tissue to be modelled in terms of physical, mechanical and biological properties. Here, we prove the suitability of a porous polyurethane foam, synthesized using an appropriate formulaton, in mimicking the bone tissue microenvironment and in reproducing the metastatic colonization derived from human breast cancer, particularly evidencing the devastating effects on the bone extracellular matrix caused by metastatic spreading., (Copyright © 2017 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
5. Induction of endoplasmic reticulum stress response by the indole-3-carbinol cyclic tetrameric derivative CTet in human breast cancer cell lines.
- Author
-
Galluzzi L, De Santi M, Crinelli R, De Marco C, Zaffaroni N, Duranti A, Brandi G, and Magnani M
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis, Autophagy, Cell Line, Tumor, Cell Proliferation, DNA-Binding Proteins metabolism, Endoplasmic Reticulum Chaperone BiP, Female, Gene Expression Regulation, Neoplastic, Gene Silencing, Humans, Macrolides pharmacology, Necrosis, Oligonucleotide Array Sequence Analysis, Proteasome Endopeptidase Complex metabolism, Reactive Oxygen Species, Regulatory Factor X Transcription Factors, Transcription Factors metabolism, X-Box Binding Protein 1, Breast Neoplasms metabolism, Endoplasmic Reticulum metabolism, Indoles pharmacology
- Abstract
Background: Indole-3-carbinol and its metabolic products are considered promising chemopreventive and anticancer agents. Previously we have shown that the indole-3-carbinol cyclic tetrameric derivative CTet induces autophagy and inhibits cell proliferation via inhibition of Akt activity and overexpression of p21/CDKN1A and GADD45A, in both estrogen receptor-positive (MCF-7) and triple negative (MDA-MB-231) breast cancer cell lines. In the present study, we further characterize the autophagic response and investigate the mechanism through which CTet regulates these events., Methodology/principal Findings: Analysis of gene expression microarray data and subsequent confirmation by quantitative real-time PCR, showed that CTet is able to induce up-regulation of key signaling molecules involved in endoplasmic reticulum (ER) stress response (e.g. DDIT3/CHOP, CHAC1, ATF3, HSPA5/BiP/GRP78, CEBPB, ASNS) and autophagy (e.g. MAP1LC3B), in both MCF-7 and MDA-MB-231 cell lines. Moreover, the monitoring of Xbp-1 splicing confirmed the activation of IRE1/Xbp-1 ER stress response branch after CTet treatment. The role of autophagic processes (known to be induced by ER stress) was investigated further through ATG5 gene silencing and pharmacological inhibition of AVOs formation. CTet was shown to induce an autophagy-related cell death. Moreover, CTet-treated cells stained with Hoechst/PI revealed the presence of necrotic processes without evidence of apoptosis., Conclusions/significance: The ER stress response was identified as the main upstream molecular mechanism through which CTet acts in both hormone-responsive and triple-negative breast cancer cells. Because of its important role in cancer development, ER stress is a potential target in cancer therapy. The abiltiy of CTet to induce ER stress response and subsequently activate a death program in tumor cells confirms this molecule as a promising anticancer agent.
- Published
- 2012
- Full Text
- View/download PDF
6. The indole-3-carbinol cyclic tetrameric derivative CTet inhibits cell proliferation via overexpression of p21/CDKN1A in both estrogen receptor-positive and triple-negative breast cancer cell lines.
- Author
-
De Santi M, Galluzzi L, Lucarini S, Paoletti MF, Fraternale A, Duranti A, De Marco C, Fanelli M, Zaffaroni N, Brandi G, and Magnani M
- Subjects
- Animals, Cell Cycle drug effects, Cell Cycle Proteins metabolism, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic drug effects, Genes, p53, Humans, Lysosomal-Associated Membrane Protein 2, Lysosomal Membrane Proteins metabolism, Mice, Mice, Nude, Nuclear Proteins metabolism, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, gamma-Cyclodextrins, Antineoplastic Agents pharmacology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Indoles pharmacology
- Abstract
Introduction: Indole-3-carbinol (I3C), an autolysis product of glucosinolates present in cruciferous vegetables, and its dimeric derivative (3,3'-DIM) have been indicated as promising agents in preventing the development and progression of breast cancer. We have recently shown that I3C cyclic tetrameric derivative CTet formulated in γ-cyclodextrin (γ-CD) efficiently inhibited cellular proliferation in breast cancer cell lines. This study aims to analyze the mechanisms involved in the in vitro inhibition of cell proliferation and to evaluate the in vivo antitumor activity of CTet in a xenograft study., Methods: Estrogen receptor-positive MCF-7 and triple-negative MDA-MB-231 breast cancer cell lines were exposed to CTet to evaluate cell cycle perturbation (propidium iodide staining and cytofluorimetric acquisition), induction of autophagic morphological features (co-localization of LC3b autophagosome marker and LAMP2a lysosome marker by immunofluorescence) and changes in protein expression (immunoblot and microarray-based gene expression analyses). To test the in vivo efficacy of CTet, female athymic nude mice inoculated with MCF-7 cells were i.p. treated with 5 mg/kg/day of CTet for five days/week for two weeks and the tumor mass was externally monitored., Results: CTet induced accumulation in G2/M phase without evidence of apoptotic response induction in both cell lines tested. In triple-negative MDA-MB-231 the autophagic lysosomal activity was significantly up-regulated after exposure to 4 μM of CTet for 8 hours, while the highest CTet concentration was necessary to observe autophagic features in MCF-7 cells. The inhibition of Akt activity and p53-independent p21/CDKN1A and GADD45A overexpression were identified as the main molecular events responsible for CTet activity in MCF-7 and p53-mutant MDA-MB-231 cells. In vivo, CTet administration was able to significantly inhibit the growth of MCF-7 xenotransplanted into nude mice, without adverse effect on body weight or on haematological parameters., Conclusions: Our data support CTet formulated with γ-CD as a promising and injectable anticancer agent for both hormone-responsive and triple-negative breast tumors.
- Published
- 2011
- Full Text
- View/download PDF
7. Cytotoxic activity of 2-Fluoro-ara-AMP and 2-Fluoro-ara-AMP-loaded erythrocytes against human breast carcinoma cell lines.
- Author
-
Pierigè F, De Marco C, Orlotti N, Dominici S, Biagiotti S, Serafini S, Zaffaroni N, Magnani M, and Rossi L
- Subjects
- Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Apoptosis drug effects, Arabinonucleotides metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carcinoma metabolism, Carcinoma pathology, Cell Line, Tumor, Cell Proliferation drug effects, Cytotoxins administration & dosage, Cytotoxins pharmacokinetics, Cytotoxins pharmacology, DNA, Neoplasm analysis, DNA, Neoplasm drug effects, Drug Delivery Systems methods, Drug Evaluation, Preclinical, Female, Flow Cytometry, Humans, Time Factors, Vidarabine Phosphate administration & dosage, Vidarabine Phosphate pharmacokinetics, Vidarabine Phosphate pharmacology, Breast Neoplasms drug therapy, Carcinoma drug therapy, Erythrocytes metabolism, Vidarabine Phosphate analogs & derivatives
- Abstract
Fludarabine phosphate (2-Fluoro-ara-AMP) is a purine analogue approved for the clinical treatment of haematological malignancies. This antimetabolite has also shown 'in vitro' antiproliferative activity against experimental models of solid mammary tumor. In this perspective, we have determined the cytotoxic effects of 2-Fluoro-ara-AMP against two human breast cancer cell lines (the ER-positive MCF-7 and the ER-negative MDA-MB-435), by adding the drug both in its free form and encapsulated into erythrocytes, as a strategy to modify the pharmacokinetic profile of the compound in order to increase its efficacy and decrease its toxicity. Similar antiproliferative activity of 2-Fluoro-ara-AMP in the two cell lines was obtained, reaching an almost complete abrogation of growth already after just 24 h of free drug exposure at all the tested doses. Meanwhile, encapsulated 2-Fluoro-ara-AMP was successfully released from erythrocytes into the culture media in a time-dependent manner with an efficacy comparable to that of the free drug treatment. This result suggests the possibility of administering 2-Fluoro-ara-AMP in patients with breast cancer using autologous erythrocytes as a system to slowly and constantly deliver 2-Fluoro-ara-A into circulation. In addition, possible mechanisms involved in the antiproliferative activity of 2-Fluoro-ara-AMP, such as the effects on cell cycle progression, p53 expression and STAT1 pathway activation in ER+ and ER- cancer cell lines, are proposed.
- Published
- 2010
- Full Text
- View/download PDF
8. Mechanisms of action and antiproliferative properties of Brassica oleracea juice in human breast cancer cell lines.
- Author
-
Brandi G, Schiavano GF, Zaffaroni N, De Marco C, Paiardini M, Cervasi B, and Magnani M
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis genetics, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Cycle drug effects, Cell Cycle genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cytotoxins pharmacology, Female, Gene Expression drug effects, Humans, Brassica chemistry, Breast Neoplasms pathology, Plant Extracts pharmacology
- Abstract
Cruciferous vegetables are an important source of compounds that may be useful for chemoprevention. In this study, we evaluated the antiproliferative activity of juice obtained from leaves of several varieties of Brassica oleracea on both estrogen receptor (ER)-positive (ER+; MCF-7 and BT474) and ER-negative (ER-; MDA-MB-231 and BT20) human breast cancer cell lines. The effect of juice on cell proliferation was evaluated on DNA synthesis and on cell cycle-related proteins. Juice markedly reduced DNA synthesis, evaluated by [3H]thymidine incorporation, starting from low concentrations (final concentration 5-15 mL/L), and this activity was independent of ER. All cauliflower varieties tested suppressed cell proliferation in a dose-dependent manner. Cell growth inhibition was accompanied by significant cell death at the higher juice concentrations, although no evidence of apoptosis was found. Interestingly, the juice displayed a preferential activity against breast cancer cells compared with other mammalian cell lines investigated (ECV304, VERO, Hep2, 3T3, and MCF-10A) (P < 0.01). At the molecular level, the inhibition of proliferation was associated with significantly reduced CDK6 expression and an increased level of p27 in ER+ cells but not in ER- cells, whereas a common feature in all cell lines was significantly decreased retinoblastoma protein phosphorylation. These results suggest that the edible part of Brassica oleracea contains substances that can markedly inhibit the growth of both ER+ and ER- human breast cancer cells, although through different mechanisms. These results suggest that the widely available cruciferous vegetables are potential chemopreventive agents.
- Published
- 2005
- Full Text
- View/download PDF
9. Akt-dependent T198 phosphorylation of cyclin-dependent kinase inhibitor p27kip1 in breast cancer.
- Author
-
Motti ML, De Marco C, Califano D, Fusco A, and Viglietto G
- Subjects
- Amino Acid Sequence, Breast cytology, Breast Neoplasms chemistry, Breast Neoplasms enzymology, Cell Cycle Proteins chemistry, Cell Cycle Proteins immunology, Cell Cycle Proteins physiology, Cell Line, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p27, Enzyme Activation physiology, Epithelial Cells chemistry, Epithelial Cells enzymology, Epithelial Cells metabolism, Humans, Kidney chemistry, Kidney embryology, Kidney enzymology, Kidney metabolism, Molecular Sequence Data, Phosphates immunology, Phosphorylation, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Threonine immunology, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins immunology, Tumor Suppressor Proteins physiology, Breast Neoplasms metabolism, Cell Cycle Proteins metabolism, Protein Serine-Threonine Kinases physiology, Proto-Oncogene Proteins physiology, Threonine metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The localization of the cyclin-dependent kinase inhibitor p27(kip1) is dependent on the phosphorylation of one of three key amino acid residues: S10, T157 and T198. However, it was unclear whether endogenous p27(kip1) is phosphorylated at T198 in the living cell. In the present work we describe the generation and characterization of a polyclonal antibody able to recognize recombinant, transfected as well as endogenous T198-phosphorylated p27(kip1). Using this antibody, we demonstrate that: (1) endogenous p27(kip1) is phosphorylated at T198 in 4 breast cancer cells lines (MCF7, MDA-MB231, MDA- MB436 and MDA-MB468); (2) T198 phosphorylation is increased in breast cancer cells compared with normal mammary epithelial cells (HMEC); (3) T198-phosphorylated p27(kip1) is exclusively cytoplasmic; (4) T198 phosphorylation is dependent on the activity of the PI3K-PKB/Akt pathway, being it drastically reduced by the pharmacological PI3K inhibitor LY294002 or stimulated by the constitutive activation of PKB/Akt. Finally, in primary human breast carcinomas, cytoplasmic accumulation of T198-phosphorylated p27(kip1) parallels Akt activation. We conclude that in breast cancer cells p27(kip1) is phosphorylated at T198 in a PI3K/Akt dependent manner and that this phosphorylation may contribute to p27(kip1) cytoplasmic mislocalization observed in breast cancer.
- Published
- 2004
10. A new indole-3-carbinol tetrameric derivative inhibits cyclin-dependent kinase 6 expression, and induces G1 cell cycle arrest in both estrogen-dependent and estrogen-independent breast cancer cell lines.
- Author
-
Brandi G, Paiardini M, Cervasi B, Fiorucci C, Filippone P, De Marco C, Zaffaroni N, and Magnani M
- Subjects
- Apoptosis drug effects, Apoptosis genetics, Breast Neoplasms enzymology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Cycle Proteins metabolism, Cyclin-Dependent Kinase 6, Cyclin-Dependent Kinase Inhibitor p27, Cyclin-Dependent Kinases biosynthesis, Enzyme Induction drug effects, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors pharmacology, Estrogen Receptor Modulators chemical synthesis, Estrogen Receptor Modulators pharmacology, Gene Expression Regulation, Neoplastic drug effects, Humans, Indoles chemical synthesis, Neoplasms, Hormone-Dependent enzymology, Neoplasms, Hormone-Dependent metabolism, Neoplasms, Hormone-Dependent pathology, Phosphorylation drug effects, Receptors, Estrogen biosynthesis, Retinoblastoma Protein metabolism, Tumor Suppressor Proteins metabolism, Up-Regulation drug effects, Breast Neoplasms drug therapy, Cyclin-Dependent Kinases antagonists & inhibitors, G1 Phase drug effects, Indoles pharmacology, Neoplasms, Hormone-Dependent drug therapy
- Abstract
Indole-3-carbinol (I3C), autolysis product of glucosinolates present in cruciferous vegetables, has been indicated as a promising agent in preventing the development and progression of breast cancer. I3C has been shown to inhibit the growth of human cancer cells in vitro and possesses anticarcinogenic activity in vivo. Because I3C is unstable and may be converted into many polymeric products in the digestive tract, it is not yet clear whether the biological activity observed can be attributed to I3C or some of its polymeric products. In this study we synthesized a stable I3C cyclic tetrameric derivative and investigated its effects on a panel of human breast cancer cell lines. The I3C tetramer suppressed the growth of both estrogen receptor (ER) -positive (MCF-7, 734B, and BT474) and ER-negative (BT20, MDA-MB-231, and BT539) human breast cancer cell lines, and it was found to induce G(1) cell cycle arrest in a dose-dependent manner without evidence of apoptosis, suggesting a growth arrest via a cytostatic mechanism. At the molecular level, the tetramer inhibited cyclin-dependent kinase (CDK) 6 expression and activity, induced an increase in the level of p27(kip1), and reduced the level of retinoblastoma protein expression. Contrarily to CDK6, the level of CDK4, the other kinase involved in the G(1) phase of the cell cycle, remains unchanged. Interestingly, the tetramer resulted about five times more active than I3C in suppressing the growth of human breast cancer cells. On the whole, our data suggest that the I3C tetrameric derivative is a novel lead inhibitor of breast cancer cell growth that may be a considered a new, promising therapeutic agent for both ER+ and ER- breast cancer.
- Published
- 2003
11. Involvement of bcl-2 and p21waf1 proteins in response of human breast cancer cell clones to Tomudex.
- Author
-
Orlandi L, Bearzatto A, Abolafio G, De Marco C, Daidone MG, and Zaffaroni N
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis drug effects, Breast Neoplasms pathology, Cell Cycle drug effects, Cell Division, Clone Cells, Cyclin-Dependent Kinase Inhibitor p21, Drug Resistance, Neoplasm, Flow Cytometry, Fluorescent Antibody Technique, Genes, p53, Humans, Proto-Oncogene Proteins c-bcl-2 genetics, Thymidylate Synthase metabolism, Transfection, Tumor Cells, Cultured, Antimetabolites, Antineoplastic pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cyclins metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Quinazolines pharmacology, Thiophenes pharmacology
- Abstract
Mechanisms of resistance to Tomudex include increased thymidylate synthase activity, as well as reduced intracellular drug uptake and polyglutamation. However, little is known about other mechanisms of resistance, such as a possible protection against Tomudex-induced apoptosis mediated by bcl-2. We transfected the MDA-MB-435 human breast cancer cell line, which is characterized by a mutated p53 gene, with cDNA of the bcl-2 gene and generated two clones (MDA-bcl4 and MDA-bcl7) characterized by bcl-2 expression twofold and fourfold that observed in the control cell clone (MDAneo). A concomitant overexpression of p21wafl was also detected in the MDA-bcl7 clone. The MDA-bcl4 clone was three times more resistant to a 24-h Tomudex exposure than the MDAneo clone, whereas the MDA-bcl7 clone was as sensitive to Tomudex as the control cell clone. A lower sensitivity of the MDA-bcl4 clone than MDAneo and MDA-bcl7 clones to 5-fluorouracil and gemcitabine was also observed. No significant difference was noted in the susceptibility of clones to fludarabine and methothrexate. Basal levels of thymidylate synthase activity were superimposable in the three clones. Tomudex induced a marked accumulation of cells in the S phase in all the clones. However, an apoptotic hypodiploid DNA peak and the characteristic nuclear morphology of apoptosis were observed only in the MDA-bcl7 clone after exposure to Tomudex. No difference in the treatment-induced modulation of proteins involved in cell cycle progression (cyclin A, cdk2, pRB, E2F-1) and apoptosis (bcl-2, bax) was observed in the three clones. The only exception was that the expression of p21wafl in the MDA-bcl4 clone was inducible at a Tomudex concentration much higher than that required to induce the protein in the other clones. Overall, the results indicate that bcl-2 and p21wafl proteins concur in determining the cellular profile of sensitivity/resistance to Tomudex.
- Published
- 1999
- Full Text
- View/download PDF
12. Modulation by lonidamine on the combined activity of cisplatin and epidoxorubicin in human breast cancer cells.
- Author
-
Silvestrini R, Gornati D, Zaffaroni N, Bearzatto A, and De Marco C
- Subjects
- Breast Neoplasms pathology, Cell Cycle drug effects, Cell Division drug effects, Cisplatin administration & dosage, Drug Administration Schedule, Drug Synergism, Epirubicin administration & dosage, Humans, Indazoles administration & dosage, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy
- Abstract
The ability of lonidamine (LND), an energolytic derivative of indazole-carboxylic acid, to modulate the cytotoxic activity of cisplatin (CDDP) and epidoxorubicin (EPI), singly or in combination, was investigated in two human breast cancer cell lines (MCF7 and T47D). A 72-hr post-incubation with a non-cytotoxic concentration of LND (75 microM) increased the activity of a 1-hr CDDP treatment as well as that of a 1 to 16-hr EPI treatment. A different pattern of interaction among the drugs and modulator was observed as a function of the sequence of drug treatment. Specifically, supra-additive or additive effects of the combination were obtained in the two cell lines according to the different treatment schemes. In particular, the maximum potentiation was observed in MCF7 cells simultaneously exposed to CDDP, EPI and LND for 1 hr and then post-incubated with LND for 72 hr, and in T47 first exposed to EPI and LND, then to CDDP and LND, and finally post-incubated with LND. Flow cytometric analysis of MCF7 cell distribution in the different cycle phases showed that combined treatment with EPI/CDDP/LND was able to stabilize cell cycle perturbations (mainly G2M accumulation) induced by individual agents. The ability of LND to potentiate CDDP and EPI cytotoxicity, and the consideration that LND causes side effects different from those caused by alkylating agents and anthracyclines, make this compound an attractive candidate for multidrug combination therapy in breast cancer.
- Published
- 1997
- Full Text
- View/download PDF
13. Activity of a chartreusin analog, elsamicin A, on breast cancer cells.
- Author
-
Silvestrini R, Sanfilippo O, Zaffaroni N, De Marco C, and Catania S
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma physiopathology, Animals, Benzopyrans pharmacology, Breast Neoplasms metabolism, Breast Neoplasms physiopathology, Cell Division drug effects, DNA, Neoplasm biosynthesis, Doxorubicin pharmacology, Drug Resistance, Female, Glycosides pharmacology, Humans, Mice, RNA, Neoplasm biosynthesis, Receptors, Estrogen drug effects, Tumor Cells, Cultured drug effects, Adenocarcinoma pathology, Aminoglycosides, Anti-Bacterial Agents pharmacology, Antibiotics, Antineoplastic pharmacology, Breast Neoplasms pathology
- Abstract
The in vitro activity of elsamicin A (ELS) was investigated compared with that of doxorubicin (DX) on two sensitive breast cancer cell lines: one estrogen receptor-positive (ER+, MCF7) and one estrogen receptor-negative (ER-, MDA-MB-231) line, and on a DX-resistant subline (MCF7DX). The activity of the two drugs was also investigated on 19 clinical breast cancer specimens from untreated patients. The drugs were tested at pharamcologically relevant concentrations, as calculated from the area under the curve for a 3 h exposure to the lethal dose producing 10% mortality (LD10) in mice, and at 10- and 100-fold concentrations. In DX-sensitive lines, a greater inhibition of RNA and DNA precursor incorporation, as well as of cell proliferation, was caused by ELS than by DX. Moreover, the antiproliferative effect was 10-fold higher in the ER+ MCF7 than in the ER- MDA-MB-231 cell line (IC50: 0.25 versus 0.21 micrograms/ml). ELS was cross-resistant to DX in the MCF7DX subline. In clinical specimens, effects on DNA precursor incorporation were more often observed for ELS than for DX at the same drug concentrations. The in vitro sensitivity to ELS was more pronounced for ER+ than for ER- tumors: minimal inhibiting concentrations of the drug were 0.1 and 3.5 micrograms/ml, respectively, in the two groups. If confirmed in a larger series of human breast tumors, these in vitro results would indicate a promising role for ELS in clinical treatment, mainly of ER+ breast cancer patients.
- Published
- 1992
- Full Text
- View/download PDF
14. Expression of P-glycoprotein in breast cancer tissue and in vitro resistance to doxorubicin and vincristine.
- Author
-
Sanfilippo O, Ronchi E, De Marco C, Di Fronzo G, and Silvestrini R
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1, Antibodies, Monoclonal, Breast chemistry, Cell Line, Drug Resistance physiology, Female, Humans, In Vitro Techniques, Tumor Cells, Cultured chemistry, Tumor Cells, Cultured drug effects, Breast Neoplasms chemistry, Doxorubicin pharmacology, Membrane Glycoproteins analysis, Neoplasm Proteins analysis, Vincristine pharmacology
- Abstract
Expression of P-glycoprotein was evaluated by C219 monoclonal antibody immunoblots in 34 previously untreated and 14 pretreated breast cancers and in benign breast lesions or histologically normal breast glands. P-glycoprotein was not detectable in the few cases of normal or benign tissue. P-glycoprotein was expressed in the 170 kD areas of 29% (10/34) of untreated and 64% (9/14) of previously treated tumours (P = 0.02). In treated tumours, high intensity expression was observed more frequently than in untreated breast cancer (40% vs. 9%). Moreover, there was a significant association between P-glycoprotein expression and in vitro resistance to doxorubicin and vincristine. Simultaneous resistance was observed in all of the P-glycoprotein positive and in only 56% of the P-glycoprotein negative tissues (P less than 0.01). Some aspects of the typical multidrug resistant phenotype, such as P-glycoprotein expression and simultaneous resistance to doxorubicin and vincristine, could be detected in small subsets of breast cancer patients. No relation between P-glycoprotein expression and the type of previous clinical treatment was observed.
- Published
- 1991
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.