Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Histochem Cytochem ; 69(7): 461-473, 2021 07.
Article in English | MEDLINE | ID: mdl-34126793

ABSTRACT

Breast cancers (BCa) with ERBB2 amplification show rapid tumor growth, increased disease progression, and lower survival rate. Deregulated intracellular trafficking and extracellular vesicle (EVs) release are mechanisms that support cancer progression and resistance to treatments. Neratinib (NE) is a Food and Drug Administration-approved pan-ERBB inhibitor employed for the treatment of ERBB2+ BCa that blocks signaling and causes survival inhibition. However, the effects of NE on ERBB2 internalization, its trafficking to multivesicular bodies (MVBs), and the release of EVs that originate from these organelles remain poorly studied. By confocal and electron microscopy, we observed that low nanomolar doses of NE induced a modest ERBB2 internalization along with an increase of clathrin-mediated endocytosis and of the CD63+ MVB compartment in SKBR-3 cells. Furthermore, we showed in the culture supernatant two distinct EV subsets, based on their size and ERBB2 positivity: small (30-100 nm) ERBB2- EVs and large (>100 nm) ERBB2+ EVs. In particular, we found that NE increased the overall release of EVs, which displayed a reduced ERBB2 positivity compared with controls. Taken together, these results provide novel insight into the effects of NE on ERBB2+ BCa cells that may lead to a reduction of ERBB2 potentially transferred to distant target cells by EVs.


Subject(s)
Breast Neoplasms/pathology , Endocytosis/drug effects , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Molecular Imaging , Quinolines/pharmacology , Receptor, ErbB-2/metabolism , Cell Line, Tumor , Female , Humans
2.
Membranes (Basel) ; 11(3)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809102

ABSTRACT

Cancers overexpressing the ERBB2 oncogene are aggressive and associated with a poor prognosis. Trastuzumab is an ERBB2 specific recombinant antibody employed for the treatment of these diseases since it blocks ERBB2 signaling causing growth arrest and survival inhibition. While the effects of Trastuzumab on ERBB2 cancer cells are well known, those on the extracellular vesicles (EVs) released from these cells are scarce. This study focused on ERBB2+ breast cancer cells and aimed to establish what type of EVs they release and whether Trastuzumab affects their morphology and molecular composition. To these aims, we performed immunoelectron microscopy, immunoblot, and high-resolution mass spectrometry analyses on EVs purified by differential centrifugation of culture supernatant. Here, we show that EVs released from ERBB2+ breast cancer cells are polymorphic in size and appearance and that ERBB2 is preferentially associated with large (120 nm) EVs. Moreover, we report that Trastuzumab (Tz) induces the expression of a specific glycosylated 50 kDa isoform of the CD63 tetraspanin and modulates the expression of 51 EVs proteins, including TOP1. Because these proteins are functionally associated with organelle organization, cytokinesis, and response to lipids, we suggest that Tz may influence these cellular processes in target cells at distant sites via modified EVs.

3.
Membranes (Basel) ; 10(5)2020 May 03.
Article in English | MEDLINE | ID: mdl-32375292

ABSTRACT

Colorectal cancer (CRC) is one of the main causes of cancer-related death in developed countries. Targeted therapies and conventional chemotherapeutics have been developed to help treat this type of aggressive cancer. Among these, the monoclonal antibodies cetuximab (Cxm) and panitumumab specifically target and inactivate the signaling of ERBB1 (EGF receptor), a key player in the development and progression of this cancer. Unfortunately, these antibodies are effective only on a small fraction of patients due to primary or secondary/acquired resistance. However, as ERBB1 cell surface expression is often maintained in resistant tumors, ERBB1 can be exploited as a target to deliver other drugs. Liposomes and immunoliposomes are under intensive investigation as pharmaceutical nanocarriers and can be functionalized with specific antibodies. In this study, we first investigated the anti-cancer activity of a cell permeable tripeptide, leucine-leucin-norleucinal (LLNle), an inhibitor of gamma-secretase and proteasome, in three different CRC cell lines that express ERBB1. We formulated LLNle-liposomes and Cxm-conjugated LLNle-loaded liposomes (LLNle-immunoliposomes) and evaluated their efficacy in inhibiting cell survival. Despite similar pro-apoptotic effects of free LLNle and LLNle-liposomes, immunoliposomes-LLNle were significantly less effective than their unconjugated counterparts. Indeed, immunoliposomes-LLNle were readily internalized and trafficked to lysosomes, where LLNle was likely trapped and/or inactivated. In conclusion, we demonstrated that LLNle was readily delivered to CRC cell lines by liposomes, but immunoliposomes-LLNle failed to show significant anti-cancer activity.

4.
Life Sci ; 232: 116610, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31254584

ABSTRACT

AIMS: The aim of this study was the characterization of the in vitro cytotoxic properties of a recently isolated diterpene compound, 7ß-acetoxy-20-hydroxy-19,20-epoxyroyleanone (compound 1), extracted from Salvia corrugata, versus human cell lines. MAIN METHODS: We used as model study immortalized breast epithelial cells MCF10A and two ERBB2+ breast cancer (BCa) cell lines, SKBR-3 and BT474. Compound 1 was isolated by methanolic extraction from regenerated shoots of Salvia corrugata Vahl, and purified by high pressure liquid chromatography (HPLC). Flow cytometry (FCM) was employed for cell cycle, apoptosis and reactive oxygen species (ROS) analysis. Cell morphology was assessed by immunofluorescence and transmission electron microscopy (TEM). KEY FINDINGS: Compound 1 inhibited cell survival of all breast cell lines. In particular, compound 1 promoted cell cycle arrest in the G0/G1 phase and apoptosis along with impairment of the mitochondrial function, which was reflected in a gross alteration of the mitochondrial network structure. Furthermore, we also detected a potent activation of the ERK1/2 kinase, which suggested the induction of reactive oxygen species (ROS). Partial rescue of survival obtained with n-acetylcysteine (NAC) when coadminstered with compound 1 further supported a contribution of ROS mediated mechanisms to the growth-arrest and proapoptotic activity of compound 1 in both BCa cell lines. ROS production was indeed confirmed in SKBR-3. SIGNIFICANCE: Our findings show that compound 1 has a cytotoxic activity against both human normal and cancer cell lines derived from breast epithelia, which is mediated by ROS generation and mitochondrial damage.


Subject(s)
Breast/drug effects , Diterpenes/pharmacology , Drugs, Chinese Herbal/pharmacology , Epithelial Cells/drug effects , Apoptosis/drug effects , Breast/cytology , Breast/metabolism , Camphanes , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/isolation & purification , Epithelial Cells/metabolism , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Panax notoginseng , Reactive Oxygen Species/metabolism , Resting Phase, Cell Cycle/drug effects , Salvia miltiorrhiza
5.
J Exp Clin Cancer Res ; 36(1): 154, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29100552

ABSTRACT

BACKGROUND: ERBB2 is overexpressed in up to 20-30% of human breast cancers (BCs), and it is associated with aggressive disease. Trastuzumab (Tz), a humanized monoclonal antibody, improves the prognosis associated with ERBB2-amplified BCs. However, the development of resistance remains a significant challenge. Carnosic acid (CA) is a diterpene found in rosemary and sage, endowed with anticancer properties. In this in vitro study, we have investigated whether Tz and CA have cooperative effects on cell survival of ERBB2 overexpressing (ERBB2+) cells and whether CA might restore Tz sensitivity in Tz-resistant cells. METHODS: We have studied BC cell migration and survival upon CA and Tz treatment. In particular, migration ability was assessed by transwell assay while cell survival was assessed by MTT assay. In addition, we have performed cell cycle and apoptosis analysis by high-resolution DNA flow cytometry and annexin-V, resazurin and sytox blue staining by flow cytometry, respectively. The expression of proteins involved in cell cycle progression, ERBB2 signaling pathway, and autophagy was evaluated by immunoblot and immunofluorescence analysis. Cellular structures relevant to the endosome/lysosome and autophagy pathways have been studied by immunofluorescence and transmission electron microscopy. RESULTS: We report that, in ERBB2+ BC cells, CA reversibly enhances Tz inhibition of cell survival, cooperatively inhibits cell migration and induces cell cycle arrest in G0/G1. These events are accompanied by ERBB2 down-regulation, deregulation of the PI3K/AKT/mTOR signaling pathway and up-regulation of both CDKN1A/p21WAF1 and CDKN1B/p27KIP1. Furthermore, we have demonstrated that CA impairs late autophagy and causes derangement of the lysosomal compartment as shown by up-regulation of SQSTM1/p62 and ultrastructural analysis. Accordingly, we have found that CA restores, at least in part, sensitivity to Tz in SKBR-3 Tz-resistant cell line. CONCLUSIONS: Our data demonstrate the cooperation between CA and Tz in inhibiting cell migration and survival of ERBB2+ BC cells that warrant further studies to establish if CA or CA derivatives may be useful in vivo in the treatment of ERBB2+ cancers.


Subject(s)
Abietanes/pharmacology , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Gene Regulatory Networks/drug effects , Humans , MCF-7 Cells , Up-Regulation/drug effects
6.
PLoS One ; 12(9): e0184425, 2017.
Article in English | MEDLINE | ID: mdl-28877236

ABSTRACT

The aim of this study was to investigate the relationship between tobacco smoke habit, patient age, DNA aneuploidy and genomic DNA copy number aberrations (CNAs) in oral potentially malignant disorder (OPMD) and oral squamous cell carcinoma (OSCC) patients. DNA aneuploidy was detected by high-resolution DNA flow cytometry (hr DNA-FCM) on DAPI stained nuclei obtained from multiple tissue samples from OPMDs/OSCCs in 220 consecutive patients. Nuclear genomic aberrations were determined in a subset of 65 patients by genome-wide array comparative genomic hybridization (aCGH) using DNA extracted from either diploid or aneuploid nuclei suspension sorted by FCM. DNA aneuploidy and mean nuclear genomic aberrations were associated with patients' age. In particular, DNA aneuploidy strongly associated with age in non-smoker OPMDs/OSCCs patients. OSCCs from smokers showed a lower prevalence of DNA aneuploidy compared to OSCCs from non-smokers. A higher occurrence of DNA aneuploidy (particularly in smokers' OPMDs) was observed in patients characterized by involvement of a single oral subsite. Our study suggests that: 1) DNA aneuploidy in non-smokers is mainly related to aging; 2) OPMDs/OSCCs involving multiple oral subsites in smokers are less likely to develop DNA aneuploidy compared to non-smokers; 3) OSCC development is characterized by both CIN and CIN-independent mechanisms and that the latter are more relevant in smokers. This study provides evidence that DNA diploid OPMDs may be considered at lower risk of cancerization than DNA aneuploid ones in non-smokers but not in smokers.


Subject(s)
Age Factors , Aneuploidy , Carcinoma, Squamous Cell/genetics , Mouth Neoplasms/genetics , Smoking , Adolescent , Adult , Aged , Aged, 80 and over , Cell Nucleus/metabolism , Chromosome Aberrations , Comparative Genomic Hybridization , Female , Genome, Human , Genome-Wide Association Study , Humans , Male , Middle Aged , Mouth Diseases/genetics , Mouth Mucosa , Precancerous Conditions/genetics , Risk Factors , Nicotiana , Young Adult
7.
Phytomedicine ; 23(7): 679-85, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27235706

ABSTRACT

BACKGROUND: Carnosic acid (CA) is a diterpenoid found in Rosmarinus officinalis L. and Salvia officinalis L. as well as in many other Lamiaceae. This compound is reported to have antioxidant and antimicrobial properties. In addition, a number of reports showed that CA has a cytotoxic activity toward several cancer cell lines. PURPOSE: The aim of this study was to establish whether CA has any specific antiproliferative effect toward human glioblastoma (GBM) cells and to analyze the molecular mechanisms involved. METHODS: We evaluated cell survival by MTT assay, apoptosis and DNA content by flow cytometry, protein expression and phosphorylation by immunoblot analyses. RESULTS: Our results showed that CA inhibited cell survival on both normal astrocytes and GBM cells. In GBM cells, in particular, CA caused an early G2 block, a reduction in the percentage of cells expressing Ki67, an enhanced expression of p21(WAF) and induced apoptosis. Furthermore, we showed that CA promoted proteasomal degradation of several substrate proteins, including Cyclin B1, retinoblastoma (RB), SOX2, and glial fibrillary acid protein (GFAP), whereas MYC levels were not modified. In addition, CA dramatically reduced the activity of CDKs. CONCLUSION: In conclusion, our findings strongly suggest that CA promotes a profound deregulation of cell cycle control and reduces the survival of GBM cells via proteasome-mediated degradation of Cyclin B1, RB and SOX2.


Subject(s)
Abietanes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cyclin B1/drug effects , Glioblastoma/pathology , Proteasome Endopeptidase Complex/drug effects , Retinoblastoma Protein/drug effects , SOXB1 Transcription Factors/drug effects , Astrocytes/drug effects , Cell Line, Tumor , Cell Proliferation , Cyclin B1/genetics , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , G2 Phase/drug effects , Humans , Proteasome Endopeptidase Complex/genetics , Retinoblastoma Protein/genetics , SOXB1 Transcription Factors/genetics
8.
PLoS One ; 10(11): e0142294, 2015.
Article in English | MEDLINE | ID: mdl-26540282

ABSTRACT

Oral potentially malignant disorders (OPMDs) characterized by the presence of dysplasia and DNA copy number aberrations (CNAs), may reflect chromosomal instability (CIN) and predispose to oral squamous cell carcinoma (OSCC). Early detection of OPMDs with such characteristics may play a crucial role in OSCC prevention. The aim of this study was to explore the relationship between CNAs, histological diagnosis, oral subsite and aneuploidy in OPMDs/OSCCs. Samples from OPMDs and OSCCs were processed by high-resolution DNA flow cytometry (hr DNA-FCM) to determine the relative nuclear DNA content. Additionally, CNAs were obtained for a subset of these samples by genome-wide array comparative genomic hybridization (aCGH) using DNA extracted from either diploid or aneuploid nuclei suspension sorted by FCM. Our study shows that: i) aneuploidy, global genomic imbalance (measured as the total number of CNAs) and specific focal CNAs occur early in the development of oral cancer and become more frequent at later stages; ii) OPMDs limited to tongue (TNG) mucosa display a higher frequency of aneuploidy compared to OPMDs confined to buccal mucosa (BM) as measured by DNA-FCM; iii) TNG OPMDs/OSCCs show peculiar features of CIN compared to BM OPMDs/OSCCs given the preferential association with total broad and specific focal CNA gains. Follow-up studies are warranted to establish whether the presence of DNA aneuploidy and specific focal or broad CNAs may predict cancer development in non-dysplastic OPMDs.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , DNA Copy Number Variations/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Aneuploidy , Chromosomal Instability/genetics , Chromosome Aberrations , DNA, Neoplasm/genetics , Diploidy , Female , Flow Cytometry/methods , Follow-Up Studies , Genomics , Humans , Male , Mouth Mucosa/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Tongue/pathology
9.
J Inorg Biochem ; 142: 101-8, 2015 01.
Article in English | MEDLINE | ID: mdl-25450024

ABSTRACT

8-Hydroxyquinoline derivatives and their metal complexes have recently awakened interest as promising therapeutic agents in cancer therapy. We have previously synthesized and evaluated glucoconjugated 8-hydroxyquinolines as copper ionophores activated by ß-glucosidases. In order to further evaluate the crucial role of the sugar, we designed and synthesized a series of new galactoconjugates of 8-hydroxyquinolines and investigated their biological properties in comparison with the 8-hydroxyquinoline analogs. The effect of copper(II) ions on their biological activities was evaluated. In particular, two compounds possess a pharmacologically relevant antiproliferative activity against specific tumor cells in the presence of copper(II) ions. Furthermore, the antiproliferative activity of the selected galactosides was successfully investigated in the presence of ß-galactosidase as a preliminary model of antibody directed enzyme prodrug therapy.


Subject(s)
Cell Proliferation/drug effects , Copper , Ionophores , Neoplasms/drug therapy , Oxyquinoline/analogs & derivatives , A549 Cells , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Ionophores/chemical synthesis , Ionophores/chemistry , Ionophores/pharmacology , Neoplasms/metabolism , Oxyquinoline/chemical synthesis , Oxyquinoline/chemistry , Oxyquinoline/pharmacology
10.
Arch Pharm (Weinheim) ; 347(6): 423-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24554280

ABSTRACT

Some new N-[6-indazolyl]arylsulfonamides and N-[alkoxy-6-indazolyl]arylsulfonamides were prepared by the reduction of 2-alkyl-6-nitroindazoles with SnCl2 in different alcohols, followed by coupling the corresponding amine with arylsulfonyl chlorides in pyridine. The newly synthesized compounds were evaluated for their antiproliferative and apoptotic activities against two human tumor cell lines: A2780 (ovarian carcinoma) and A549 (lung adenocarcinoma). Preliminary in vitro pharmacological studies revealed that N-(2-allyl-2H-indazol-6-yl)-4-methoxybenzenesulfonamide 4 and N-[7-ethoxy-2-(4-methyl-benzyl)-2H-indazol-6-yl]-4-methyl-benzenesulfonamide 9 exhibited significant antiproliferative activity against the A2780 and A549 cell lines with IC50 values in the range from 4.21 to 18.6 µM, and also that they trigger apoptosis in a dose-dependent manner. Furthermore, both active compounds were able to cause an arrest of cells in the G2/M phase of the cell cycle, typical but not exclusive of tubulin interacting agents, although only infrequent interactions with the microtubule network were observed by immunofluorescence microscopy, while docking analysis showed a possible different behavior between the two active compounds.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Indazoles/chemical synthesis , Indazoles/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Inhibitory Concentration 50 , Microscopy, Fluorescence , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
11.
Chemistry ; 19(41): 13946-55, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24038335

ABSTRACT

Recent investigations have rekindled interest in 8-hydroxyquinolines as therapeutic agents for cancer, Alzheimer's disease, and other neurodegenerative disorders. Three new ß-cyclodextrin conjugates of 8-hydroxyquinolines and their copper(II) and zinc(II) complexes have been synthesized and characterized spectroscopically. In addition to improving aqueous solubility, due to the presence of the cyclodextrin moiety, the hybrid systems have interesting characteristics including antioxidant activity, and their copper(II) complexes are efficient superoxide dismutase (SOD) mimics. The ligands and their copper(II) complexes show low cytotoxicity, attributed to the presence of the cyclodextrin moiety. These compounds have potential as therapeutic agents in diseases related both to metal dyshomeostasis and oxidative stress.


Subject(s)
Antioxidants/chemistry , Antioxidants/chemical synthesis , Chelating Agents/chemistry , Chelating Agents/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Cyclodextrins/chemistry , Glycoconjugates/chemistry , Metals/chemistry , Oxyquinoline/chemistry , Superoxide Dismutase/chemistry , Antioxidants/metabolism , Chelating Agents/metabolism , Coordination Complexes/metabolism , Copper/chemistry , Ligands , Metals/therapeutic use , Oxidative Stress , Solubility , Superoxide Dismutase/metabolism , Superoxide Dismutase/therapeutic use , Zinc/chemistry
12.
Pharmacol Rep ; 65(3): 717-23, 2013.
Article in English | MEDLINE | ID: mdl-23950595

ABSTRACT

BACKGROUND: In previous papers we demonstrated that the activity of short heteroretinoids as anti-proliferative and pro-apoptotic compounds was deeply linked to their heterocyclic moiety and that ionone-derived 1,5-pyrazoles had the highest anti-proliferative activity in our preliminary experiments. We then demonstrated the high and pharmacologically significant anti-proliferative and apoptotic activities of the pyrazole compounds 2-(1-(4-chlorophenyl)-1H-pyrazol-5-yl)-5-methoxyphenol (EN12-4), 5-methoxy-2-(1-(pyridin-2-yl)-1H-pyrazol-5-yl)phenol (EN12-2A) and 2-(5-(4-methoxyphenyl)-1H-pyrazol-1-yl)pyridine (EN7-2) establishing, especially for EN12-2A, a possible mechanism of action involving the cell microtubular system. METHODS: Here, the anti-proliferative activity of these pyrazole compounds was analyzed in vitro by the MTT assay in six drug-resistant cell lines, five of which were selected after exposure to increasing concentrations of cisplatin (L1210/DDP), doxorubicin (A2780/DX3), 5-fluorouracil (HCT-8/5FU), taxol (A549/T24) and etoposide (MCF-7/VP), and one was obtained by transfection of the ABCG2 membrane transporter (HEK-293/R2). RESULTS: Our data show that these compounds have a similar anti-proliferative activity in nearly all resistant and sensitive cell lines, demonstrating their ability to overcome the most common mechanisms of drug resistance with two exceptions regarding the MCF-7/VP cell line over-expressing the ABCC1 (MRP1) transporter, and the MDR1 over-expressing A2780/DX3 cells, with a calculated RI = 3.2 for EN12-2A, relative to their sensitive cellular counterpart. On the other hand, the taxol-resistant A549/T24 cell line showed a significantly increased sensitivity to our compounds. CONCLUSIONS: Our data suggest that our pyrazole compounds are able to overcome in vitro the most common drug-resistance mechanisms demonstrating a significant anti-proliferative activity and confirming a mechanism of action involving the depolymerization of microtubules.


Subject(s)
Cell Proliferation/drug effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Pyrazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , MCF-7 Cells , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Tumor Cells, Cultured
13.
Dalton Trans ; 42(6): 2023-34, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23174818

ABSTRACT

8-Hydroxyquinoline derivatives are metal-binding compounds that have recently attracted interest as therapeutic agents for cancer therapy. In this scenario, we designed and synthesized three new glucoconjugates, 5,7-dichloro-8-quinolinyl-ß-D-glucopyranoside, 5-chloro-8-quinolinyl-ß-D-glucopyranoside and 2-methyl-8-quinolinyl-ß-D-glucopyranoside and investigated their biological properties in comparison to the parent 8-hydroxyquinoline derivatives in the presence of Cu(2+). In vitro data show that 2 out of 3 glycosylated compounds possess a pharmacologically-relevant antiproliferative activity against tumor cells, similar to that of their parent compounds; this activity is associated with a relevant triggering of apoptosis. The pharmacological profile of the glucoconjugates depends on the cellular enzymatic ß-glucosidase activity, as demonstrated by the inhibition of antiproliferative activity in the presence of the 2,5-dideoxy-2,5-imino-D-mannitol.


Subject(s)
Antineoplastic Agents/chemistry , Copper/chemistry , Ionophores/chemistry , Prodrugs/chemistry , beta-Glucosidase/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Binding Sites , Caco-2 Cells , Catalytic Domain , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use , Coordination Complexes/toxicity , Enzyme Activation , Glycosylation , Humans , Hydroxyquinolines/chemistry , Molecular Docking Simulation , Neoplasms/drug therapy , Prodrugs/therapeutic use , Prodrugs/toxicity , Protein Stability , beta-Glucosidase/chemistry
14.
Eur J Med Chem ; 57: 240-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23072738

ABSTRACT

Recently, it has been reported that compounds bearing a sulfonamide moiety possess many types of biological activities, including anticancer activity. The present work reports the synthesis and antiproliferative evaluation of some N-(6(4)-indazolyl)benzenesulfonamides and 7-ethoxy-N-(6(4)-indazolyl)benzenesulfonamides. All compounds were evaluated for their in vitro antiproliferative activity against three tumor cell lines: A2780 (human ovarian carcinoma) A549 (human lung adenocarcinoma) and P388 (murine leukemia). The results indicated that sulfonamides 2c, 3c, 6d, 8, 13, 3b and 16 were endowed with a pharmacologically interesting antiproliferative activity with compounds 2c and 3c showing the lower IC(50) (from 0.50 ± 0.09 to 1.83 ± 0.52 µM and from 0.58 ± 0.17 to 5.83 ± 1.83 µM, respectively). Moreover, these indazoles were able to trigger apoptosis through the upregulation of the typical apoptosis markers p53 and bax. As regard to the hypothetic targets of these compounds, a preliminary docking analysis showed that all compounds seemed to interact with ß-tubulin, in particular compound 3b that showed the lower Ki. The cytofluorimetric analysis of the cell cycle phases indicates that all compounds, when administered at their IC(75), caused a block in the G2/M phase of the cell cycle with the generation of subpopulations of cells with a number of chromosome >4n. When the IC(50)s were applied we observed a prevalent block in the G0/G1 phase except for compounds 16 and 8 where a partial G2/M block was present with a concomitant decrease of cells in the G0/G1 and S phases of the cell cycle. Altogether these results suggest a possible, but not exclusive, interaction with microtubules.


Subject(s)
Antineoplastic Agents/chemical synthesis , Indazoles/chemical synthesis , Sulfonamides/chemical synthesis , Tubulin/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , G1 Phase/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Indazoles/pharmacology , Inhibitory Concentration 50 , Kinetics , Mice , Microtubules/drug effects , Molecular Docking Simulation , Polyploidy , Resting Phase, Cell Cycle/drug effects , Structure-Activity Relationship , Sulfonamides/pharmacology , Tubulin/metabolism
15.
Dalton Trans ; 41(15): 4530-5, 2012 Apr 21.
Article in English | MEDLINE | ID: mdl-22354329

ABSTRACT

8-Hydroxyquinolines are systems of great interest in the field of inorganic and bioinorganic chemistry. They are metal-binding compounds and are known to exhibit a variety of biological activities, such as antibacterial and anticancer activities. Among these systems, clioquinol has been the focus of a renewed interest in recent years. In this scenario, we synthesized and characterized the new clioquinol glucoconjugate, 5-chloro-7-iodo-8-quinolinyl-ß-D-glucopyranoside in order to compare this system to that of clioquinol. We also synthesized, 8-quinolinyl-ß-D-glucopyranoside, an 8-hydroxyquinoline glucoconjugate. The reason for the development of glucoconjugates is the glucose avidity, and the over-expression of glucose transporters in cancer cells. Here we demonstrate that glycoconjugates are cleaved in vitro by ß-glucosidase and these systems exhibit antiproliferative activity against different tumor cell lines in the presence of copper(II) ions.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glycoconjugates/chemistry , Glycoconjugates/pharmacology , Oxyquinoline/chemistry , Oxyquinoline/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Glycoconjugates/metabolism , Humans , Neoplasms/drug therapy , Oxyquinoline/metabolism , Prodrugs/metabolism , beta-Glucosidase/metabolism
16.
Eur J Med Chem ; 46(11): 5293-309, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21920636

ABSTRACT

We synthesized thirty-six novel pyrazole derivatives and studied their antiproliferative activity in human ovarian adenocarcinoma A2780 cells, human lung carcinoma A549 cells, and murine P388 leukemia cells. Four of these substances were selected because of their higher antiproliferative activity and further analyses showed that they were all able to induce apoptosis, although to a different extent. The expression of p53 and p21(waf1), which induce apoptosis and cell cycle arrest, was evaluated by western blot analysis in cells treated with compound 12d. The analysis of the cell cycle showed that all the selected compounds cause a partial G2/M block and the formation of polyploid cells. Furthermore, the four selected compounds were tested for their interaction with the microtubular cytoskeletal system by docking analysis, tubulin polymerization assay and immunofluorescence staining, demonstrating that the compound 12d, unlike the other active derivatives, was able to significantly bind dimers of α- and ß-tubulin, probably causing a molecular distortion resulting in the disassembly of microtubules.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Evaluation, Preclinical , Humans , Indoles/metabolism , Mice , Microtubules/drug effects , Microtubules/metabolism , Models, Molecular , Protein Multimerization/drug effects , Protein Structure, Quaternary , Pyrazoles/chemistry , Tubulin/chemistry , Tubulin/metabolism , Tumor Suppressor Protein p53/metabolism
17.
Invest New Drugs ; 29(1): 98-109, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19876599

ABSTRACT

We report herein the reversal of multidrug resistance-1 (MDR1) in A2780/DX3 cells by the two nifedipine-like compounds 1 and 2 that are part of a library of 1,4-dihydropyridines (1,4-DHPs) calcium-channel modulators bearing in C-4 a different substituted imidazo[2,1-b]thiazole system. By methylthiazol tetrazolium (MTT) assay, cytofluorimetry, and fluorescence microscopy we evaluated their ability to reverse MDR in our cell system. Moreover, together with compound 3 (the diltiazem-like 8-(4-chlorophenyl)-5-methyl-8-[(2Z)-pent-2-en-1-yloxy]-8H-[1,2,4]oxadiazolo[3,4-c][1,4]thiazin-3-one) we analyzed their ability to potentiate the triggering of apoptosis after exposure to doxorubicin, through the nuclear morphological analysis after 4',6-diamidino-2-phenylindole (DAPI), the fluorescein isothiocyanate (FITC)-Annexin-V/propidium iodide (PI) staining and the caspase activity determination. Our results demonstrate that compounds 1 and 2, at concentrations showing a very low (5%) or absent inhibition of cell proliferation, in combination with doxorubicin enhance its antiproliferative activity (from 30% to 54% IC(50) reduction) in A2780/DX3 cells through an increase of doxorubicin intracellular accumulation. These compounds together with compound 3, which has already been demonstrated to act as a potent inhibitor of MDR1 function, were also able to significantly potentiate the activation of the apoptosis machinery triggered by the exposure to doxorubicin. In conclusion, our results identify two new molecules structurally related to the calcium-channel blocker nifedipine, but characterized by a very low LTCC blockers activity, able to potentiate the antiproliferative and apoptotic activities of doxorubicin through an increase of its intracellular concentration likely caused by the inhibition of MDR1 function.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Apoptosis/drug effects , Diltiazem/analogs & derivatives , Diltiazem/pharmacology , Nifedipine/analogs & derivatives , Nifedipine/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Annexin A5/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Diltiazem/chemistry , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Activation/drug effects , Flow Cytometry , Humans , Indoles/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Nifedipine/chemistry , Propidium/metabolism , Staining and Labeling
18.
Invest New Drugs ; 29(3): 443-55, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20039098

ABSTRACT

In this work, nanoparticles with a positive surface charge were prepared through the electrostatic interaction of a new cisplatin-hyaluronate complex with N-trimethyl chitosan (substitution degree of 85%). Mean particle diameter was approximately 195 nm. Drug loading of nanoparticles, which had a zeta potential of about 27 mV, was equal to 6% w/w. After 24 h, while the cisplatin-hyaluronate complex released approximately 60% w/w drug in phosphate buffered saline at pH 7.4, approximately 40% w/w of total cisplatin was released from nanoparticles. The same cumulative amounts of released drug were found after 48 h. These nanoparticles, as well as the starting cisplatin-hyaluronate complex, were active on all cell lines tested (P388, A2780, A549), with an antiproliferative activity similar to that of cisplatin. Apoptosis was markedly induced in A2780 cells by nanoparticles. In a preliminary in vivo experiment, the antitumour activity against a murine tumour (P388 cells) subcutaneously implanted in mice, resulted similar to that of cisplatin for nanoparticles whereas the starting complex showed a non-significant activity at the cisplatin dose tested. Body weight change of treated mice suggested a significantly better tolerance of the nanoparticles compared to cisplatin, after an initial brief period of acute toxicity higher than the parent drug. These results indicate that such a particulate system could be useful as a carrier for cisplatin delivery.


Subject(s)
Antineoplastic Agents/pharmacology , Chitosan/pharmacology , Cisplatin/pharmacology , Hyaluronic Acid/pharmacology , Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/chemistry , Diffusion/drug effects , Drug Screening Assays, Antitumor , Humans , Hyaluronic Acid/chemistry , Mice , Particle Size , Regression Analysis , Tumor Burden/drug effects
19.
Org Biomol Chem ; 8(24): 5674-81, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-20936232

ABSTRACT

Different nitronaphthylbutadienes have been previously proved to have antitumour activity. The main drawback of these derivatives is their low water solubility. With the aim of facilitating the administration of these new drugs we have synthesized the hexyl (2Z,4E)-2-methylsulfanyl-5-(1-naphthyl)-4-nitro-2,4-pentadienoate analogue (1-Naph-NHCB) which is demonstrated to be easily included into cyclodextrins and/or entrapped into liposomes. Its antitumour activity was revealed to be almost comparable with that of the previously studied methyl analogue ester (1-Naph-NMCB). On the other hand, in vitro studies with different cancer cell lines showed that the cytotoxic activity of both 1-Naph-NMCB and 1-Naph-NHCB were fully preserved and in some cases also enhanced when entrapped into liposomal carriers.


Subject(s)
Antineoplastic Agents/chemistry , Butadienes/chemistry , Nitro Compounds/chemistry , Water/chemistry , Antineoplastic Agents/pharmacology , Butadienes/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Molecular Structure , Nitro Compounds/pharmacology , Solubility
20.
Am J Physiol Endocrinol Metab ; 297(5): E1078-88, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19706788

ABSTRACT

Somatostatin receptors (SSTR1-5) mediate antiproliferative effects. In C6 rat glioma cells, somatostatin is cytostatic in vitro via phosphotyrosine phosphatase-dependent inhibition of ERK1/2 activity mediated by SSTR1, -2, and -5. Here we analyzed the effects of SSTR activation on C6 glioma growth in vivo and the intracellular mechanisms involved, comparing somatostatin effects with selective agonists for SSTR1, -2, and -5 (BIM-23745, BIM-23120, BIM-23206) or receptor biselective compounds (SSTR1 and -2, BIM-23704; and SSTR2 and -5, BIM-23190). Nude mice subcutaneously xenografted with C6 cells were treated with somatostatin, SSTR agonists (50 µg, twice/day), or vehicle. Tumor growth was evaluated every 3 days for 19 days. The intracellular pathways responsible of SSTR effects in vivo were evaluated measuring Ki-67, phospho-ERK1/2, and p27(kip1) expression by immunohistochemistry in sections from explanted tumors. Somatostatin and SSTR1, -2, and -5 agonists strongly inhibited in vivo C6 tumor growth, intratumoral neovessel formation, Ki-67 expression, and ERK1/2 phosphorylation and induced upregulation of p27(Kip1), whereas only a modest activation of caspase-3 was observed. Somatostatin (acting on SSTR1, -2, and -5) displayed the highest efficacy; SSTR5 selective agonist showed a stronger effect than SSTR1 agonist, and SSTR2 agonist was less effective. On the other hand, SSTR1 and -2 agonists maximally reduced tumor neovascularization. The combined activation of SSTR1 and -2 showed a synergistic activity, reaching a higher efficacy than BIM-23206, whereas the simultaneous activation of SSTR2 and -5 resulted in a response resembling SSTR5 effects. Thus the simultaneous activation of different SSTRs inhibits glioma cell proliferation in vivo through both direct cytotostatic and antiangiogenic effects.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Receptors, Somatostatin/agonists , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Glioma/blood supply , Glioma/pathology , Immunohistochemistry , Mice , Mice, Nude , Microcirculation/drug effects , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Oligopeptides/pharmacology , Piperazines/pharmacology , Regional Blood Flow/drug effects , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...