79 results on '"Claudia Fumarola"'
Search Results
2. Targeting glucosylceramide synthase induces antiproliferative and proapoptotic effects in osimertinib-resistant NSCLC cell models
- Author
-
Silvia La Monica, Federica Vacondio, Kamal Eltayeb, Alessio Lodola, Francesco Volta, Martina Viglioli, Francesca Ferlenghi, Francesca Galvani, Maricla Galetti, Mara Bonelli, Claudia Fumarola, Andrea Cavazzoni, Lisa Flammini, Michela Verzè, Roberta Minari, Pier Giorgio Petronini, Marcello Tiseo, Marco Mor, and Roberta Alfieri
- Subjects
Medicine ,Science - Abstract
Abstract The EGFR tyrosine kinase inhibitor osimertinib has been approved for the first-line treatment of EGFR-mutated Non-Small Cell Lung Cancer (NSCLC) patients. Despite its efficacy, patients develop resistance. Mechanisms of resistance are heterogeneous and not fully understood, and their characterization is essential to find new strategies to overcome resistance. Ceramides are well-known regulators of apoptosis and are converted into glucosylceramides (GlcCer) by glucosylceramide synthase (GCS). A higher content of GlcCers was observed in lung pleural effusions from NSCLC patients and their role in osimertinib-resistance has not been documented. The aim of this study was to determine the therapeutic potential of inhibiting GCS in NSCLC EGFR-mutant models resistant to osimertinib in vitro and in vivo. Lipidomic analysis showed a significant increase in the intracellular levels of glycosylceramides, including GlcCers in osimertinib resistant clones compared to sensitive cells. In resistant cells, the GCS inhibitor PDMP caused cell cycle arrest, inhibition of 2D and 3D cell proliferation, colony formation and migration capability, and apoptosis induction. The intratumoral injection of PDMP completely suppressed the growth of OR xenograft models. This study demonstrated that dysregulation of ceramide metabolism is involved in osimertinib-resistance and targeting GCS may be a promising therapeutic strategy for patients progressed to osimertinib.
- Published
- 2024
- Full Text
- View/download PDF
3. A Photoactive Supramolecular Complex Targeting PD-L1 Reveals a Weak Correlation between Photoactivation Efficiency and Receptor Expression Levels in Non-Small-Cell Lung Cancer Tumor Models
- Author
-
Pietro Delcanale, Manuela Maria Alampi, Andrea Mussini, Claudia Fumarola, Maricla Galetti, Pier Giorgio Petronini, Cristiano Viappiani, Stefano Bruno, and Stefania Abbruzzetti
- Subjects
targeted photodynamic therapy ,photo-immunotherapy ,PD-L1 ,dSTORM ,eosin ,photosensitizer ,Pharmacy and materia medica ,RS1-441 - Abstract
Photo-immunotherapy uses antibodies conjugated to photosensitizers to produce nanostructured constructs endowed with targeting properties and photo-inactivation capabilities towards tumor cells. The superficial receptor density on cancer cells is considered a determining factor for the efficacy of the photodynamic treatment. In this work, we propose the use of a photoactive conjugate that consists of the clinical grade PD-L1-binding monoclonal antibody Atezolizumab, covalently linked to either the well-known photosensitizer eosin or the fluorescent probe Alexa647. Using single-molecule localization microscopy (direct stochastic optical reconstruction microscopy, dSTORM), and an anti-PD-L1 monoclonal antibody labelled with Alexa647, we quantified the density of PD-L1 receptors exposed on the cell surface in two human non-small-cell lung cancer lines (H322 and A549) expressing PD-L1 to a different level. We then investigated if this value correlates with the effectiveness of the photodynamic treatment. The photodynamic treatment of H322 and A549 with the photo-immunoconjugate demonstrated its potential for PDT treatments, but the efficacy did not correlate with the PD-L1 expression levels. Our results provide additional evidence that receptor density does not determine a priori the level of photo-induced cell death.
- Published
- 2023
- Full Text
- View/download PDF
4. CDK4/6 inhibitors improve the anti-tumor efficacy of lenvatinib in hepatocarcinoma cells
- Author
-
Graziana Digiacomo, Claudia Fumarola, Silvia La Monica, Mara Bonelli, Andrea Cavazzoni, Maricla Galetti, Rita Terenziani, Kamal Eltayeb, Francesco Volta, Silvia Zoppi, Patrizia Bertolini, Gabriele Missale, Roberta Alfieri, and Pier Giorgio Petronini
- Subjects
hepatocarcinoma (HCC) ,CDK4/6 inhibition ,abemaciclib ,lenvatinib ,senescence ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer with a poor prognosis and limited treatment options. Considering that alterations of the CDK4/6-cyclin D-Rb pathway occur frequently in HCC, we tested the efficacy of two CDK4/6 inhibitors, abemaciclib and ribociclib, in combination with lenvatinib, a multi-kinase inhibitor approved as first-line therapy for advanced HCC, in a panel of HCC Rb-expressing cell lines. The simultaneous drug combinations showed a superior anti-proliferative activity as compared with single agents or sequential schedules of treatment, either in short or in long-term experiments. In addition, the simultaneous combination of abemaciclib with lenvatinib reduced 3D cell growth, and impaired colony formation and cell migration. Mechanistically, these growth-inhibitory effects were associated with a stronger down-regulation of c-myc protein expression. Depending on the HCC cell model, reduced activation of MAPK, mTORC1/p70S6K or src/FAK signaling was also observed. Abemaciclib combined with lenvatinib arrested the cells in the G1 cell cycle phase, induced p21 accumulation, and promoted a stronger increase of cellular senescence, associated with elevation of β-galactosidase activity and accumulation of ROS, as compared with single treatments. After drug withdrawal, the capacity of forming colonies was significantly impaired, suggesting that the anti-tumor efficacy of abemaciclib and lenvatinib combination was persistent.Our pre-clinical results demonstrate the effectiveness of the simultaneous combination of CDK4/6 inhibitors with lenvatinib in HCC cell models, suggesting that this combination may be worthy of further investigation as a therapeutic approach for the treatment of advanced HCC.
- Published
- 2022
- Full Text
- View/download PDF
5. Third generation EGFR inhibitor osimertinib combined with pemetrexed or cisplatin exerts long-lasting anti-tumor effect in EGFR-mutated pre-clinical models of NSCLC
- Author
-
Silvia La Monica, Roberta Minari, Daniele Cretella, Lisa Flammini, Claudia Fumarola, Mara Bonelli, Andrea Cavazzoni, Graziana Digiacomo, Maricla Galetti, Denise Madeddu, Angela Falco, Costanza Annamaria Lagrasta, Anna Squadrilli, Elisabetta Barocelli, Alessandro Romanel, Federico Quaini, Pier Giorgio Petronini, Marcello Tiseo, and Roberta Alfieri
- Subjects
Non-Small Cell Lung Cancer ,Osimertinib ,Resistance ,Epidermal Growth Factor Receptor ,Pemetrexed ,Cisplatin ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The third generation Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitor (TKI) osimertinib has been initially approved for T790M positive Non-Small Cell Lung Cancer (NSCLC) and more recently for first-line treatment of EGFR-mutant T790M negative NSCLC patients. Similarly to previous generation TKIs, despite the high response rate, disease progression eventually occurs and current clinical research is focused on novel strategies to delay the emergence of osimertinib resistance. In this study we investigated the combination of osimertinib with pemetrexed or cisplatin in EGFR-mutated NSCLC cell lines and xenografts. Methods Tumor growth was evaluated in a PC9T790M xenograft model and tissue composition was morphometrically determined. PC9, PC9T790M and HCC827 cell lines were employed to test the efficacy of osimertinib and chemotherapy combination in vitro. Cell viability and cell death were evaluated by MTT assay and fluorescence microscopy. Protein expression and gene status were analysed by Western blotting, fluorescence in situ hybridization analysis, next-generation sequencing and digital droplet PCR. Results In xenograft models, osimertinib significantly inhibited tumor growth, however, as expected, in 50% of mice drug-resistance developed. A combination of osimertinib with pemetrexed or cisplatin prevented or at least delayed the onset of resistance. Interestingly, such combinations increased the fraction of fibrotic tissue and exerted a long-lasting activity after stopping therapy. In vitro studies demonstrated the stronger efficacy of the combination over the single treatments in inhibiting cell proliferation and inducing cell death in PC9T790M cells as well as in T790M negative PC9 and HCC827 cell lines, suggesting the potential role of this strategy also as first-line treatment. Finally, we demonstrated that osimertinib resistant clones, either derived from resistant tumors or generated in vitro, were less sensitive to pemetrexed prompting to use a chemotherapy regimen non-containing pemetrexed in patients after progression to osimertinib treatment. Conclusions Our results identify a combination between osimertinib and pemetrexed or cisplatin potentially useful in the treatment of EGFR-mutated NSCLC patients, which might delay the appearance of osimertinib resistance with long-lasting effects.
- Published
- 2019
- Full Text
- View/download PDF
6. Simultaneous Combination of the CDK4/6 Inhibitor Palbociclib With Regorafenib Induces Enhanced Anti-tumor Effects in Hepatocarcinoma Cell Lines
- Author
-
Graziana Digiacomo, Claudia Fumarola, Silvia La Monica, Mara A. Bonelli, Daniele Cretella, Roberta Alfieri, Andrea Cavazzoni, Maricla Galetti, Patrizia Bertolini, Gabriele Missale, and Pier Giorgio Petronini
- Subjects
hepatocarcinoma (HCC) ,CDK4/6 inhibition ,palbociclib (PD-0332991) ,regorafenib ,metabolism ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Advanced hepatocarcinoma (HCC) is an aggressive malignancy with poor prognosis and limited treatment options. Alterations of the cyclin D-CDK4/6-Rb pathway occur frequently in HCC, providing the rationale for its targeting at least in a molecular subset of HCC. In a panel of HCC cell lines, we investigated whether the CDK4/6 inhibitor palbociclib might improve the efficacy of regorafenib, a powerful multi-kinase inhibitor approved as second-line treatment for advanced HCC after sorafenib failure and currently under clinical investigation as first-line therapy in combination with immunotherapy. In Rb-proficient cells, the simultaneous drug combination, but not the sequential schedules, inhibited cell proliferation, either in short or in long-term experiments, and induced cell death more strongly than individual treatments. Moreover, the combination significantly reduced spheroid cell growth and inhibited cell migration/invasion. The superior efficacy of palbociclib plus regorafenib emerged also under hypoxia and was associated with a significant down-regulation of CDK4/6-Rb-myc and mTORC1/p70S6K signaling. Moreover, regorafenib suppressed palbociclib-induced expression of cyclin D1 contributing to the cytotoxic effects of the combination. Besides these inhibitory effects on cell viability/proliferation, palbociclib and regorafenib reduced glucose uptake, although this effect was dependent on the cell model and on the oxygen availability (normoxia or hypoxia). Palbociclib and regorafenib combination impaired glucose uptake and utilization, down-regulating basal and hypoxia-induced expression of HIF-1α, HIF-2α, GLUT-1, and MCT4 proteins as well as the activity/expression of glycolytic enzymes (HK2, PFKP, aldolase A, PKM2). In addition, regorafenib alone reduced mitochondrial respiration. The combined treatment impaired glucose metabolism and respiration without enhancing the effects of the single agents. Our findings provide pre-clinical evidence for the effectiveness of palbociclib and regorafenib combination in HCC cell models.
- Published
- 2020
- Full Text
- View/download PDF
7. The anti-tumor efficacy of CDK4/6 inhibition is enhanced by the combination with PI3K/AKT/mTOR inhibitors through impairment of glucose metabolism in TNBC cells
- Author
-
Daniele Cretella, Andrea Ravelli, Claudia Fumarola, Silvia La Monica, Graziana Digiacomo, Andrea Cavazzoni, Roberta Alfieri, Alessandra Biondi, Daniele Generali, Mara Bonelli, and Pier Giorgio Petronini
- Subjects
Palbociclib ,Triple-negative breast cancer ,CDK4/6 inhibition ,PI3K/mTOR inhibitors ,Glucose metabolism ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Cell cycle regulators have gain attention as potential targets for anticancer therapy. Palbociclib is a selective inhibitor of the cyclin-dependent kinases 4 and 6 (CDK4/6), which coordinate the G1-S transition. Palbociclib is currently approved for the treatment of hormone receptor positive, HER2-negative advanced breast cancer (BC) in association with letrozole or fulvestrant. In contrast, its efficacy in triple negative BC (TNBC), either alone or in combined therapies, has not been fully investigated to date. Methods Here we evaluated the potential of combining palbociclib with PI3K/mTOR inhibitors in Rb-proficient TNBC cells comparing different schedules of treatment: simultaneous, sequential, or sequential combined treatment (pre-incubation with palbociclib followed by exposure to both palbociclib and PI3K/mTOR inhibitors). We assessed the effects on cell proliferation, cell death, and cell cycle distribution, and looked at the impact of such treatments on glucose metabolism. Results Palbociclib exerted cytostatic effects in Rb-positive TNBC cells, inducing a reversible blockade in G0/G1 cell cycle phase associated with down-regulation of CDK6, Rb, and c-myc expression and/or activity. Palbociclib treatment induced AKT signaling, providing a rationale for its combination with PI3K/mTOR inhibitors. The simultaneous or sequential treatment resulted in an additive inhibition of cell proliferation. On the other hand, the sequential combined treatment in which palbociclib was maintained also during exposure to PI3K/mTOR inhibitors gave rise to synergistic anti-proliferative and pro-apoptotic effects, by inhibiting both CDK4/6/Rb/myc and PI3K/mTOR signaling. Interestingly, the inhibition of the Rb/E2F/myc axis mediated by palbociclib resulted in a significant down-regulation of glucose metabolism; most importantly, these inhibitory effects were enhanced by the combination of palbociclib with BYL719 (specific inhibitor of the p110α PI3K-subunit), which promoted a stronger inhibition of GLUT-1 glucose transporter expression, glucose uptake and consumption in comparison with individual treatments, under both normoxic and hypoxic conditions. Conclusions Combination of palbociclib with PI3K/mTOR inhibitors may represent a promising therapeutic option for the treatment of Rb-proficient TNBC, with the sequential combined schedule showing a superior efficacy over the other schedules. In addition our results demonstrate that the impairment of glucose metabolism may contribute to the anti-tumor activity of such drug combinations.
- Published
- 2018
- Full Text
- View/download PDF
8. Reprogramming of Lipid Metabolism in Lung Cancer: An Overview with Focus on EGFR-Mutated Non-Small Cell Lung Cancer
- Author
-
Kamal Eltayeb, Silvia La Monica, Marcello Tiseo, Roberta Alfieri, and Claudia Fumarola
- Subjects
lipid metabolism ,lung cancer ,EGFR ,EGFR-TKI resistance ,Cytology ,QH573-671 - Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Most of lung cancer cases are classified as non-small cell lung cancers (NSCLC). EGFR has become an important therapeutic target for the treatment of NSCLC patients, and inhibitors targeting the kinase domain of EGFR are currently used in clinical settings. Recently, an increasing interest has emerged toward understanding the mechanisms and biological consequences associated with lipid reprogramming in cancer. Increased uptake, synthesis, oxidation, or storage of lipids has been demonstrated to contribute to the growth of many types of cancer, including lung cancer. In this review, we provide an overview of metabolism in cancer and then explore in more detail the role of lipid metabolic reprogramming in lung cancer development and progression and in resistance to therapies, emphasizing its connection with EGFR signaling. In addition, we summarize the potential therapeutic approaches targeting lipid metabolism for lung cancer treatment.
- Published
- 2022
- Full Text
- View/download PDF
9. Trastuzumab emtansine delays and overcomes resistance to the third-generation EGFR-TKI osimertinib in NSCLC EGFR mutated cell lines
- Author
-
Silvia La Monica, Daniele Cretella, Mara Bonelli, Claudia Fumarola, Andrea Cavazzoni, Graziana Digiacomo, Lisa Flammini, Elisabetta Barocelli, Roberta Minari, Nadia Naldi, Pier Giorgio Petronini, Marcello Tiseo, and Roberta Alfieri
- Subjects
NSCLC ,EGFR ,Osimertinib ,T-DM1 ,TKI-resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Osimertinib is a third-generation EGFR-TKI with a high selective potency against T790M-mutant NSCLC patients. Considering that osimertinib can lead to enhanced HER-2 expression on cell surface and HER-2 overexpression is a mechanism of resistance to osimertinib, this study was addressed to investigate the potential of combining osimertinib with trastuzumab emtansine (T-DM1) in order to improve the efficacy of osimertinib and delay or overcome resistance in NSCLC cell lines with EGFR activating mutation and with T790M mutation or HER-2 amplification. Methods The effects of osimertinib combined with T-DM1 on cell proliferation, cell cycle, cell death, antibody-dependent cell-mediated cytotoxicity (ADCC), and acquisition of osimertinib resistance was investigated in PC9, PC9-T790M and H1975 cell lines. The potential of overcoming osimertinib resistance with T-DM1 was tested in a PC9/HER2c1 xenograft model. Results T-DM1 exerted an additive effect when combined with osimertinib in terms of inhibition of cell proliferation, cell death and ADCC induction in PC9, PC9-T790M and H1975 cell lines. Combining osimertinib and T-DM1 using different schedules in long-term growth experiments revealed that the appearance of osimertinib-resistance was prevented in PC9-T790M and delayed in H1975 cells when the two drugs were given together. By contrast, when osimertinib was followed by T-DM1 an antagonistic effect was observed on cell proliferation, cell death and resistance acquisition. In xenograft models, we demonstrated that HER-2 amplification was associated with osimertinib-resistance and that T-DM1 co-administration is a potential strategy to overcome this resistance. Conclusions Our data suggest that concomitant treatment with osimertinib and T-DM1 may be a promising therapeutic strategy for EGFR-mutant NSCLC.
- Published
- 2017
- Full Text
- View/download PDF
10. Combined Inhibition of CDK4/6 and PI3K/AKT/mTOR Pathways Induces a Synergistic Anti-Tumor Effect in Malignant Pleural Mesothelioma Cells
- Author
-
Mara A. Bonelli, Graziana Digiacomo, Claudia Fumarola, Roberta Alfieri, Federico Quaini, Angela Falco, Denise Madeddu, Silvia La Monica, Daniele Cretella, Andrea Ravelli, Paola Ulivi, Michela Tebaldi, Daniele Calistri, Angelo Delmonte, Luca Ampollini, Paolo Carbognani, Marcello Tiseo, Andrea Cavazzoni, and Pier Giorgio Petronini
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Malignant pleural mesothelioma (MPM) is a progressive malignancy associated to the exposure of asbestos fibers. The most frequently inactivated tumor suppressor gene in MPM is CDKN2A/ARF, encoding for the cell cycle inhibitors p16INK4a and p14ARF, deleted in about 70% of MPM cases. Considering the high frequency of alterations of this gene, we tested in MPM cells the efficacy of palbociclib (PD-0332991), a highly selective inhibitor of cyclin-dependent kinase (CDK) 4/6. The analyses were performed on a panel of MPM cell lines and on two primary culture cells from pleural effusion of patients with MPM. All the MPM cell lines, as well as the primary cultures, were sensitive to palbociclib with a significant blockade in G0/G1 phase of the cell cycle and with the acquisition of a senescent phenotype. Palbociclib reduced the phosphorylation levels of CDK6 and Rb, the expression of myc with a concomitant increased phosphorylation of AKT. Based on these results, we tested the efficacy of the combination of palbociclib with the PI3K inhibitors NVP-BEZ235 or NVP-BYL719. After palbociclib treatment, the sequential association with PI3K inhibitors synergistically hampered cell proliferation and strongly increased the percentage of senescent cells. In addition, AKT activation was repressed while p53 and p21 were up-regulated. Interestingly, two cycles of sequential drug administration produced irreversible growth arrest and senescent phenotype that were maintained even after drug withdrawal. These findings suggest that the sequential association of palbociclib with PI3K inhibitors may represent a valuable therapeutic option for the treatment of MPM.
- Published
- 2017
- Full Text
- View/download PDF
11. Expanding the Arsenal of FGFR Inhibitors: A Novel Chloroacetamide Derivative as a New Irreversible Agent With Anti-proliferative Activity Against FGFR1-Amplified Lung Cancer Cell Lines
- Author
-
Claudia Fumarola, Nicole Bozza, Riccardo Castelli, Francesca Ferlenghi, Giuseppe Marseglia, Alessio Lodola, Mara Bonelli, Silvia La Monica, Daniele Cretella, Roberta Alfieri, Roberta Minari, Maricla Galetti, Marcello Tiseo, Andrea Ardizzoni, Marco Mor, and Pier Giorgio Petronini
- Subjects
FGFR ,irreversible inhibitors ,drug design ,lung cancer ,SQCLC ,cancer drug resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Fibroblast Growth Factor Receptors (FGFR1–4) have a critical role in the progression of several human cancers, including Squamous Non-Small-Cell Lung Cancer (SQCLC). Both non-selective and selective reversible FGFR inhibitors are under clinical investigation for the treatment of patients with tumors harboring FGFR alterations. Despite their potential efficacy, the clinical development of these drugs has encountered several challenges, including toxicity, and the appearance of drug resistance. Recent efforts have been directed at development of irreversible FGFR inhibitors, which have the potential to exert superior anti-proliferative activity in tumors carrying FGFR alterations. With this in mind, we synthetized, and investigated a set of novel inhibitors possessing a warhead potentially able to covalently bind a cysteine in the P-loop of FGFR. Among them, the chloroacetamide UPR1376 resulted able to irreversible inhibit FGFR1 phosphorylation in FGFR1 over-expressing cells generated from SQCLC SKMES-1 cells. In addition, this compound inhibited cell proliferation in FGFR1-amplified H1581 cells with a potency higher than the reversible inhibitor BGJ398 (infigratinib), while sparing FGFR1 low-expressing cells. The anti-proliferative effects of UPR1376 were demonstrated in both 2D and 3D systems and were associated with the inhibition of MAPK and AKT/mTOR signaling pathways. UPR1376 inhibited cell proliferation also in two BGJ398-resistant cell clones generated from H1581 by chronic exposure to BGJ398, although at concentrations higher than those effective in the parental cells, likely due to the persistent activation of the MAPK pathway associated to NRAS amplification. Combined blockade of FGFR1 and MAPK signaling, by UPR1376 and trametinib respectively, significantly enhanced the efficacy of UPR1376, providing a means of circumventing resistance to FGFR1 inhibition. Our findings suggest that the insertion of a chloroacetamide warhead on a suitable scaffold, as exemplified by UPR1376, is a valuable strategy to develop a novel generation of FGFR inhibitors for the treatment of SQCLC patients with FGFR alterations.
- Published
- 2019
- Full Text
- View/download PDF
12. Novel Activity of a Synthetic Decapeptide Against Toxoplasma gondii Tachyzoites
- Author
-
Laura Giovati, Claudia Santinoli, Carlo Mangia, Alice Vismarra, Silvana Belletti, Tiziana D’Adda, Claudia Fumarola, Tecla Ciociola, Cristina Bacci, Walter Magliani, Luciano Polonelli, Stefania Conti, and Laura H. Kramer
- Subjects
anti-Toxoplasma agents ,killer peptide KP ,apoptosis ,TUNEL ,mitochondrial potential ,transmission electron microscopy ,Microbiology ,QR1-502 - Abstract
The killer peptide KP is a synthetic decapeptide derived from the sequence of the variable region of a recombinant yeast killer toxin-like microbicidal single-chain antibody. KP proved to exert significant activities against diverse microbial and viral pathogens through different mechanisms of action, but little is known of its effect on apicomplexan protozoa. The aim of the present study was to evaluate the in vitro activity of KP against Toxoplasma gondii, a globally widespread protozoan parasite of great medical interest. The effect of KP treatment and its potential mechanism of action on T. gondii were evaluated by various methods, including light microscopy, quantitative PCR, flow cytometry, confocal microscopy, and transmission electron microscopy. In the presence of KP, the number of T. gondii tachyzoites able to invade Vero cells and the parasite intracellular proliferation were significantly reduced. Morphological observation and analysis of apoptotic markers suggested that KP is able to trigger an apoptosis-like cell death in T. gondii. Overall, our results indicate that KP could be a promising candidate for the development of new anti-Toxoplasma drugs with a novel mechanism of action.
- Published
- 2018
- Full Text
- View/download PDF
13. Effect of ABCG2/BCRP Expression on Efflux and Uptake of Gefitinib in NSCLC Cell Lines.
- Author
-
Maricla Galetti, Pier Giorgio Petronini, Claudia Fumarola, Daniele Cretella, Silvia La Monica, Mara Bonelli, Andrea Cavazzoni, Francesca Saccani, Cristina Caffarra, Roberta Andreoli, Antonio Mutti, Marcello Tiseo, Andrea Ardizzoni, and Roberta R Alfieri
- Subjects
Medicine ,Science - Abstract
BCRP/ABCG2 emerged as an important multidrug resistance protein, because it confers resistance to several classes of cancer chemotherapeutic agents and to a number of novel molecularly-targeted therapeutics such as tyrosine kinase inhibitors. Gefitinib is an orally active, selective EGFR tyrosine kinase inhibitor used in the treatment of patients with advanced non small cell lung cancer (NSCLC) carrying activating EGFR mutations. Membrane transporters may affect the distribution and accumulation of gefitinib in tumour cells; in particular a reduced intracellular level of the drug may result from poor uptake, enhanced efflux or increased metabolism.The present study, performed in a panel of NSCLC cell lines expressing different ABCG2 plasma membrane levels, was designed to investigate the effect of the efflux transporter ABCG2 on intracellular gefitinib accumulation, by dissecting the contribution of uptake and efflux processes.Our findings indicate that gefitinib, in lung cancer cells, inhibits ABCG2 activity, as previously reported. In addition, we suggest that ABCG2 silencing or overexpression affects intracellular gefitinib content by modulating the uptake rather than the efflux. Similarly, overexpression of ABCG2 affected the expression of a number of drug transporters, altering the functional activities of nutrient and drug transport systems, in particular inhibiting MPP, glucose and glutamine uptake.Therefore, we conclude that gefitinib is an inhibitor but not a substrate for ABCG2 and that ABCG2 overexpression may modulate the expression and activity of other transporters involved in the uptake of different substrates into the cells.
- Published
- 2015
- Full Text
- View/download PDF
14. Gefitinib inhibits invasive phenotype and epithelial-mesenchymal transition in drug-resistant NSCLC cells with MET amplification.
- Author
-
Silvia La Monica, Cristina Caffarra, Francesca Saccani, Elena Galvani, Maricla Galetti, Claudia Fumarola, Mara Bonelli, Andrea Cavazzoni, Daniele Cretella, Rita Sirangelo, Rita Gatti, Marcello Tiseo, Andrea Ardizzoni, Elisa Giovannetti, Pier Giorgio Petronini, and Roberta R Alfieri
- Subjects
Medicine ,Science - Abstract
Despite the initial response, all patients with epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) eventually develop acquired resistance to EGFR tyrosine kinase inhibitors (TKIs). The EGFR-T790M secondary mutation is responsible for half of acquired resistance cases, while MET amplification has been associated with acquired resistance in about 5-15% of NSCLCs. Clinical findings indicate the retained addiction of resistant tumors on EGFR signaling. Therefore, we evaluated the molecular mechanisms supporting the therapeutic potential of gefitinib maintenance in the HCC827 GR5 NSCLC cell line harbouring MET amplification as acquired resistance mechanism. We demonstrated that resistant cells can proliferate and survive regardless of the presence of gefitinib, whereas the absence of the drug significantly enhanced cell migration and invasion. Moreover, the continuous exposure to gefitinib prevented the epithelial-mesenchymal transition (EMT) with increased E-cadherin expression and down-regulation of vimentin and N-cadherin. Importantly, the inhibition of cellular migration was correlated with the suppression of EGFR-dependent Src, STAT5 and p38 signaling as assessed by a specific kinase array, western blot analysis and silencing functional studies. On the contrary, the lack of effect of gefitinib on EGFR phosphorylation in the H1975 cells (EGFR-T790M) correlated with the absence of effects on cell migration and invasion. In conclusion, our findings suggest that certain EGFR-mutated patients may still benefit from a second-line therapy including gefitinib based on the specific mechanism underlying tumor cell resistance.
- Published
- 2013
- Full Text
- View/download PDF
15. Supplementary Tables S1-S5 from Inhibition of PI3K Pathway Reduces Invasiveness and Epithelial-to-Mesenchymal Transition in Squamous Lung Cancer Cell Lines Harboring PIK3CA Gene Alterations
- Author
-
Andrea Ardizzoni, Pier Giorgio Petronini, Marcello Tiseo, Claudia Fumarola, Pietro Rossetti, Angela Falco, Costanza Annamaria Lagrasta, Caterina Frati, Denise Madeddu, Cristina Caffarra, Daniele Cretella, Maricla Galetti, Silvia La Monica, Federico Quaini, Roberta R. Alfieri, Francesca Saccani, Andrea Cavazzoni, and Mara A. Bonelli
- Abstract
Supplementary Tables S1-S5. Table S1: Antiproliferative effects of PI3K inhibitors on SKMES-1 and PIK3CA amplified/mutated clones Tables S2-S4: densitometric analysis of Western blotting of Figure 1D Table S5: Antiproliferative effects of PI3K inhibitors on H596 and HCC2450 cells
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figures S1-S5 from Inhibition of PI3K Pathway Reduces Invasiveness and Epithelial-to-Mesenchymal Transition in Squamous Lung Cancer Cell Lines Harboring PIK3CA Gene Alterations
- Author
-
Andrea Ardizzoni, Pier Giorgio Petronini, Marcello Tiseo, Claudia Fumarola, Pietro Rossetti, Angela Falco, Costanza Annamaria Lagrasta, Caterina Frati, Denise Madeddu, Cristina Caffarra, Daniele Cretella, Maricla Galetti, Silvia La Monica, Federico Quaini, Roberta R. Alfieri, Francesca Saccani, Andrea Cavazzoni, and Mara A. Bonelli
- Abstract
Supplementary Figures S1-S5. Figure S1: p-AKT levels in PIK3CA transfected clones Figure S2: Induction of cell death after treatment with PI3K inhibitors Figure S3: Time course evaluation of PI3K/AKT/mTOR pathway activity after exposure to PI3K inhibitors Figure S4: Evaluation of cell migration after treatment with PI3K inhibitors Figure S5: CD133 expression in tumor spheres
- Published
- 2023
- Full Text
- View/download PDF
17. Synergic activity of FGFR2 and MEK inhibitors in the treatment of FGFR2-amplified cancers of unknown primary
- Author
-
Andrea Cavazzoni, Irene Salamon, Claudia Fumarola, Giulia Gallerani, Noemi Laprovitera, Francesco Gelsomino, Mattia Riefolo, Karim Rihawi, Elisa Porcellini, Tania Rossi, Martina Mazzeschi, Maria Naddeo, Salvatore Serravalle, Elisabetta Broseghini, Federico Agostinis, Gabriele Sales, Olivier Deas, Giorgio Durante, Mattia Lauriola, Ingrid Garajova, George A. Calin, Massimiliano Bonafè, Antonia D’Errico, Pier Giorgio Petronini, Stefano Cairo, Andrea Ardizzoni, and Manuela Ferracin
- Abstract
SummaryPatients with cancer of unknown primary (CUP) carry the double burden of an aggressive disease and reduced access to therapies. Experimental models summing up CUP features are pivotal for CUP biology investigation and drug testing. We derived two CUP cell lines (CUP#55 and #96), and corresponding patient-derived xenografts (PDXs), from ascites tumor cells. CUP cell lines and PDXs underwent histological, immune-phenotypical, molecular, and genomic characterization confirming the features of the original tumor. Genetic testing and FISH analysis identified FGFR2 amplification as therapeutic target in tumor tissues and patient-derived models. Drug-screening assays were performed to test the activity of FGFR2 targeting drug BGJ-398 (infigratinib) and the combination treatment with the MEK inhibitor trametinib, which proved to be synergic and exceptionally active, bothin vitroandin vivo. This study brings personalized therapy closer to CUP patients and paves the way to future applications of personalized medicine for metastatic patients with adverse prognosis.
- Published
- 2023
- Full Text
- View/download PDF
18. CDK4/6 Inhibition Enhances the Efficacy of Standard Chemotherapy Treatment in Malignant Pleural Mesothelioma Cells
- Author
-
Rita Terenziani, Maricla Galetti, Silvia La Monica, Claudia Fumarola, Silvia Zoppi, Roberta Alfieri, Graziana Digiacomo, Andrea Cavazzoni, Delia Cavallo, Massimo Corradi, Marcello Tiseo, Pier Giorgio Petronini, and Mara Bonelli
- Subjects
Cancer Research ,Oncology ,malignant pleural mesothelioma ,CDK4/6 inhibitors ,abemaciclib ,chemotherapy ,senescence - Abstract
Background: The loss of the CDKN2A/ARF (cyclin-dependent kinase inhibitor 2A/alternative reading frame) gene is the most common alteration in malignant pleural mesothelioma (MPM), with an incidence of about 70%, thus representing a novel target for mesothelioma treatment. In the present study, we evaluated the antitumor potential of combining the standard chemotherapy regimen used for unresectable MPM with the CDK4/6 (cyclin-dependent kinase 4 or 6) inhibitor abemaciclib. Methods: Cell viability, cell death, senescence, and autophagy induction were evaluated in two MPM cell lines and in a primary MPM cell culture. Results: The simultaneous treatment of abemaciclib with cisplatin and pemetrexed showed a greater antiproliferative effect than chemotherapy alone, both in MPM cell lines and in primary cells. This combined treatment induced cellular senescence or autophagic cell death, depending on the cell type. More in detail, the induction of cellular senescence was related to the increased expression of p21, whereas autophagy induction was due to the impairment of the AKT/mTOR signaling. Notably, the effect of the combination was irreversible and no resumption in tumor cell proliferation was observed after drug withdrawal. Conclusion: Our results demonstrated the therapeutic potential of CDK4/6 inhibitors in combination with chemotherapy for the treatment of MPM and are consistent with the recent positive results in the MiST2 arm in abemaciclib-treated patients.
- Published
- 2022
- Full Text
- View/download PDF
19. Acquired BRAF G469A Mutation as a Resistance Mechanism to First-Line Osimertinib Treatment in NSCLC Cell Lines Harboring an EGFR Exon 19 Deletion
- Author
-
Federica Riccardi, Maricla Galetti, Roberta Alfieri, Mara Bonelli, Claudia Fumarola, Roberta Minari, Graziana Digiacomo, Pier Giorgio Petronini, Silvia La Monica, Daniele Cretella, Andrea Cavazzoni, and Marcello Tiseo
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Genotype ,Antineoplastic Agents ,Apoptosis ,03 medical and health sciences ,T790M ,0302 clinical medicine ,Gefitinib ,Epidermal growth factor ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Humans ,Pharmacology (medical) ,Osimertinib ,Epidermal growth factor receptor ,Cell Proliferation ,Sequence Deletion ,Trametinib ,Acrylamides ,Aniline Compounds ,biology ,business.industry ,High-Throughput Nucleotide Sequencing ,Dabrafenib ,Exons ,ErbB Receptors ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Selumetinib ,Cancer research ,biology.protein ,business ,medicine.drug - Abstract
Osimertinib is a new third-generation, epidermal growth factor receptor-tyrosine kinase inhibitor highly selective for the epidermal growth factor receptor with both activating and T790M mutations. A recent phase III trial showed a statistically significant progression-free survival benefit with osimertinib vs. gefitinib or erlotinib as first-line treatment for EGFR-mutated non-small cell lung cancer, and preliminary data are available on resistance mechanisms to first-line osimertinib therapy. The objective of this study was to examine potential in vitro mechanisms of acquired resistance to osimertinib in a cell model carrying an EGFR exon 19 deletion. PC9 cells were cultured in the presence of increasing concentrations of osimertinib (ranging from 10 to 500 nM) to generate resistant cells. Three clones resistant to osimertinib (half maximal inhibitory concentration > 1 μM) were isolated, genotyped by next-generation sequencing and tested for drug sensitivity. Cell proliferation and migration, cell death, and signaling transduction pathways were analyzed. Our study revealed that all the three resistant clones developed acquired resistance via the BRAF G469A mutation maintaining a constitutive activation of the ERK pathway. Stable transfection of PC9 and HCC827 cells with a plasmid containing BRAF G469A rendered the cells resistant to osimertinib. Treatment with selumetinib and trametinib, but not dabrafenib, restored the sensitivity to osimertinib and enhanced cell death in the resistant clones with the BRAF G469A mutation. Our in vitro studies revealed the BRAF G469A-activating mutation as a potential mechanism of acquired resistance to first-line osimertinib treatment, and provide a strategy of intervention to overcome this mechanism of resistance.
- Published
- 2019
- Full Text
- View/download PDF
20. Dual Inhibition of CDK4/6 and PI3K/AKT/mTOR Signaling Impairs Energy Metabolism in MPM Cancer Cells
- Author
-
Maricla Galetti, Rita Terenziani, Pier Giorgio Petronini, Silvia Zoppi, Roberta Alfieri, Silvia La Monica, Graziana Digiacomo, Mara Bonelli, Claudia Fumarola, Daniele Cretella, and Andrea Cavazzoni
- Subjects
Pyridines ,Pleural Neoplasms ,Glucose uptake ,Cell ,Palbociclib ,Article ,Piperazines ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,medicine ,Humans ,malignant pleural mesothelioma ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,Spectroscopy ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,PI3K/mTOR inhibitors ,Chemistry ,Cell growth ,TOR Serine-Threonine Kinases ,Mesothelioma, Malignant ,Organic Chemistry ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,General Medicine ,Mitochondria ,Computer Science Applications ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer cell ,Cancer research ,CDK4/6 inhibition ,CDK4/6 Inhibition ,Energy Metabolism ,Glycolysis ,Proto-Oncogene Proteins c-akt ,metabolism ,Signal Transduction - Abstract
Background: Malignant pleural mesothelioma (MPM) is an aggressive malignancy associated to asbestos exposure. One of the most frequent genetic alteration in MPM patients is CDKN2A/ARF loss, leading to aberrant activation of the Rb pathway. In MPM cells, we previously demonstrated the therapeutic efficacy of targeting this signaling with the CDK4/6 inhibitor palbociclib in combination with PI3K/mTOR inhibitors. Here, we investigated whether such combination may have an impact on cell energy metabolism. Methods: The study was performed in MPM cells of different histotypes, metabolic analyses were conducted by measuring GLUT-1 expression and glucose uptake/consumption, and by SeaHorse technologies. Results: MPM cell models differed for their ability to adapt to metabolic stress conditions, such as glucose starvation and hypoxia. Independently of these differences, combined treatments with palbociclib and PI3K/mTOR inhibitors inhibited cell proliferation more efficaciously than single agents. The drugs alone reduced glucose uptake/consumption as well as glycolysis, and their combination further enhanced these effects under both normoxic and hypoxic conditions. Moreover, the drug combinations significantly impaired mitochondrial respiration as compared with individual treatments. These metabolic effects were mediated by the concomitant inhibition of Rb/E2F/c-myc and PI3K/AKT/mTOR signaling. Conclusions: Dual blockade of glycolysis and respiration contributes to the anti-tumor efficacy of palbociclib-PI3K/mTOR inhibitors combination.
- Published
- 2020
- Full Text
- View/download PDF
21. Balancing reactivity and antitumor activity: heteroarylthioacetamide derivatives as potent and time-dependent inhibitors of EGFR
- Author
-
Graziana Digiacomo, Marco Mor, Riccardo Castelli, Nicole Bozza, Andrea Cavazzoni, Francesca Ferlenghi, Mara Bonelli, Federica Vacondio, Claudia Fumarola, Roberta Alfieri, Donatella Callegari, Alessio Lodola, Silvia Rivara, Claudia Silva, and Pier Giorgio Petronini
- Subjects
Lung Neoplasms ,Thio ,Antineoplastic Agents ,Thioacetamide ,01 natural sciences ,03 medical and health sciences ,chemistry.chemical_compound ,T790M ,Cell Line, Tumor ,Acetamides ,Drug Discovery ,Nucleophilic substitution ,Humans ,Cysteine ,Phosphorylation ,Cell Proliferation ,030304 developmental biology ,EGFR inhibitors ,Pharmacology ,0303 health sciences ,010405 organic chemistry ,Organic Chemistry ,Autophosphorylation ,Leaving group ,Gefitinib ,General Medicine ,respiratory tract diseases ,0104 chemical sciences ,ErbB Receptors ,chemistry ,Biochemistry ,A549 Cells ,Acetamide - Abstract
Second- and third-generation inhibitors of EGFR possess an acrylamide group which alkylates Cys797, allowing to overcome resistance due to insurgence of T790M mutation. Less reactive warheads, yet capable to bind the target cysteine, may be useful to design newer and safer inhibitors. In the present work, we synthesized a 2-chloro-N-(4-(phenylamino)quinazolin-6-yl)acetamide (8) derivative as a prototype of EGFR inhibitor potentially able to react with Cys797 by nucleophilic substitution. We then tuned the reactivity of the acetamide fragment by replacing the chlorine leaving group with (hetero)-aromatic thiols or carboxylate esters. Among the synthesized derivatives, the 2-((1H-imidazol-2-yl)thio)acetamide 16, while showing negligible reactivity with cysteine in solution, caused long-lasting inhibition of wild-type EGFR autophosphorylation in A549 cells, resulted able to bind recombinant EGFR L858R/T790M in a time-dependent manner, and inhibited both EGFR autophosphorylation and proliferation in gefitinib-resistant H1975 lung cancer cells (expressing EGFR L858R/T790M mutant) at low micromolar concentration.
- Published
- 2019
- Full Text
- View/download PDF
22. Pemetrexed Enhances Membrane PD-L1 Expression and Potentiates T Cell-Mediated Cytotoxicity by Anti-PD-L1 Antibody Therapy in Non-Small-Cell Lung Cancer
- Author
-
Mara Bonelli, Maricla Galetti, Andrea Ardizzoni, Pier Giorgio Petronini, Silvia La Monica, Valeria Barili, Michelangelo Fiorentino, Elisa Giovannetti, Graziana Digiacomo, Alessandra Zecca, Marcello Tiseo, Andrea Cavazzoni, Roberta Alfieri, Daniele Cretella, Claudia Fumarola, Medical oncology laboratory, CCA - Cancer biology and immunology, Amsterdam Gastroenterology Endocrinology Metabolism, Cavazzoni A., Digiacomo G., Alfieri R., La Monica S., Fumarola C., Galetti M., Bonelli M., Cretella D., Barili V., Zecca A., Giovannetti E., Fiorentino M., Tiseo M., Petronini P.G., and Ardizzoni A.
- Subjects
0301 basic medicine ,PD-L1 ,Cancer Research ,medicine.medical_treatment ,Pembrolizumab ,NSCLC ,chemotherapy ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Lung cancer ,Cytotoxicity ,pemetrexed ,IFN-γ ,Chemotherapy ,biology ,business.industry ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Pemetrexed ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,T cell mediated cytotoxicity ,business ,medicine.drug - Abstract
Immunotherapy has significantly changed the treatment landscape for advanced non-small-cell lung cancer (NSCLC) with the introduction of drugs targeting programmed cell death protein-1 (PD-1) and programmed cell death ligand-1 (PD-L1). In particular, the addition of the anti-PD-1 antibody pembrolizumab to platinum-pemetrexed chemotherapy resulted in a significantly improved overall survival in patients with non-squamous NSCLC, regardless of PD-L1 expression. In this preclinical study, we investigated whether chemotherapy can modulate PD-L1 expression in non-squamous NSCLC cell lines, thus potentially affecting immunotherapy efficacy. Among different chemotherapeutic agents tested, only pemetrexed increased PD-L1 levels by activating both mTOR/P70S6K and STAT3 pathways. Moreover, it also induced the secretion of cytokines, such as IFN-&gamma, and IL-2, by activated peripheral blood mononuclear cells PBMCs that further stimulated the expression of PD-L1 on tumor cells, as demonstrated in a co-culture system. The anti-PD-1/PD-L1 therapy enhanced T cell-mediated cytotoxicity of NSCLC cells treated with pemetrexed and expressing high levels of PD-L1 in comparison with untreated cells. These data may explain the positive results obtained with pemetrexed-based chemotherapy combined with pembrolizumab in PD-L1-negative NSCLC and can support pemetrexed as one of the preferable chemotherapy partners for immunochemotherapy combination regimens.
- Published
- 2020
- Full Text
- View/download PDF
23. Impairing energy metabolism in solid tumors through agents targeting oncogenic signaling pathways
- Author
-
Roberta Alfieri, Pier Giorgio Petronini, and Claudia Fumarola
- Subjects
0301 basic medicine ,Glutamine ,Cell ,Antineoplastic Agents ,Biology ,Biochemistry ,03 medical and health sciences ,Immune system ,Immunity ,Neoplasms ,medicine ,Humans ,Pharmacology ,Cancer ,Metabolism ,Lipid Metabolism ,medicine.disease ,Metabolic pathway ,030104 developmental biology ,medicine.anatomical_structure ,Cell metabolism ,Cancer cell ,Carcinogens ,Cancer research ,Energy Metabolism ,Glycolysis ,Signal Transduction - Abstract
Cell metabolic reprogramming is one of the main hallmarks of cancer and many oncogenic pathways that drive the cancer-promoting signals also drive the altered metabolism. This review focuses on recent data on the use of oncogene-targeting agents as potential modulators of deregulated metabolism in different solid cancers. Many drugs, originally designed to inhibit a specific target, then have turned out to have different effects involving also cell metabolism, which may contribute to the mechanisms underlying the growth inhibitory activity of these drugs. Metabolic reprogramming may also represent a way by which cancer cells escape from the selective pressure of targeted drugs and become resistant. Here we discuss how targeting metabolism could emerge as a new effective strategy to overcome such resistance. Finally, accumulating evidence indicates that cancer metabolic rewiring may have profound effects on tumor-infiltrating immune cells. Modulating cancer metabolic pathways through oncogene-targeting agents may not only restore more favorable conditions for proper lymphocytes activation, but also increase the persistence of memory T cells, thereby improving the efficacy of immune-surveillance.
- Published
- 2018
- Full Text
- View/download PDF
24. Anti-proliferative effects of copper(II) complexes with hydroxyquinoline-thiosemicarbazone ligands
- Author
-
Claudia Fumarola, Pier Giorgio Petronini, Giorgio Pelosi, Andrea Cavazzoni, Mauro Carcelli, Daniele Cretella, Anna Gatti, Matteo Tegoni, Dominga Rogolino, and Vincenzo Verdolino
- Subjects
Models, Molecular ,Thiosemicarbazones ,DNA damage ,Stereochemistry ,Blotting, Western ,chemistry.chemical_element ,Antineoplastic Agents ,Crystallography, X-Ray ,Ligands ,010402 general chemistry ,01 natural sciences ,Metal ,chemistry.chemical_compound ,Coordination Complexes ,Neoplasms ,Drug Discovery ,Humans ,Molecule ,Semicarbazone ,Cell Proliferation ,Pharmacology ,chemistry.chemical_classification ,Reactive oxygen species ,Molecular Structure ,010405 organic chemistry ,Cell growth ,Cell Cycle ,Organic Chemistry ,8-Hydroxyquinoline ,General Medicine ,Copper ,0104 chemical sciences ,chemistry ,visual_art ,Hydroxyquinolines ,visual_art.visual_art_medium ,Reactive Oxygen Species - Abstract
The possibility to influence the physiological concentration of copper ions through the careful choice of ligands is emerging as a novel intriguing strategy in the treatment of pathologies such as cancer and Alzheimer. Thiosemicarbazones play an important role in this field, because they offer a wide variety of potential functionalizations and different kinds of coordination modes. Here we report the synthesis of some 8-hydroxyquinoline thiosemicarbazone ligands containing an ONN'S donor set and their Zn(II) and Cu(II) complexes. The metal complexes were characterized in solution and in the solid state and the X-ray structure of one of the copper(II) complex is reported. The Cu(II) complexes were characterized also by means of quantum mechanical calculations. The Cu(II) complexes displayed cytostatic activity in different cancer cell models. In particular, the most active Cu(II) complex significantly inhibited cell proliferation with an IC50 value lower than 1 μM; this effect was associated with a block of the cell cycle in the G2/M phase. This Cu(II) complex induced neither the production of reactive oxygen species (ROS) nor the accumulation of p53 protein, suggesting the lack of DNA damage.
- Published
- 2017
- Full Text
- View/download PDF
25. Multiple effects of CDK4/6 inhibition in cancer: From cell cycle arrest to immunomodulation
- Author
-
Silvia La Monica, Mara Bonelli, Claudia Fumarola, and Roberta Alfieri
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Pyridines ,Aminopyridines ,Palbociclib ,Biochemistry ,Piperazines ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Neoplasms ,medicine ,Animals ,Humans ,Pharmacology ,Tumor microenvironment ,business.industry ,Cancer ,Cyclin-Dependent Kinase 4 ,Cell Cycle Checkpoints ,Cyclin-Dependent Kinase 6 ,Cell cycle ,medicine.disease ,Metastatic breast cancer ,030104 developmental biology ,Purines ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Benzimidazoles ,CDK4/6 Inhibition ,business - Abstract
Dysregulation of the cell cycle is a hallmark of cancer that leads to aberrant cellular proliferation. CDK4/6 are cyclin-dependent kinases activated in response to proliferative signaling, which induce RB hyper-phosphorylation and hence activation of E2F transcription factors, thus promoting cell cycle progression through the S phase. Pharmacologic inhibition of CDK4/6 by palbociclib, ribociclib, or abemaciclib has been showing promising activity in multiple cancers with the best results achieved in combination with other agents. Indeed, CDK4/6 inhibitors are currently approved in combination with endocrine therapy for the treatment of estrogen receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer. Moreover, a number of clinical trials are currently underway to test the efficacy of combining CDK4/6 inhibitors with different drugs not only in breast but also in other types of cancer. Beyond the inhibition of cell proliferation, CDK4/6 inhibitors have recently revealed new effects on cancer cells and on tumor microenvironment. In particular, it has been reported that these agents induce a senescent-like phenotype, impact on cell metabolism and exert both immunomodulatory and immunogenic effects. Here we describe recent data on the anti-tumor effects of CDK4/6 inhibitors as single agents or in combined therapies, focusing in particular on their metabolic and immunomodulatory activities.
- Published
- 2019
26. Pre-treatment with the CDK4/6 inhibitor palbociclib improves the efficacy of paclitaxel in TNBC cells
- Author
-
Maricla Galetti, Graziana Digiacomo, Silvia La Monica, Daniele Generali, Claudia Fumarola, Mara Bonelli, Pier Giorgio Petronini, Daniele Cretella, Andrea Cavazzoni, and Roberta Alfieri
- Subjects
Paclitaxel ,Pyridines ,medicine.medical_treatment ,lcsh:Medicine ,Apoptosis ,Triple Negative Breast Neoplasms ,Palbociclib ,Article ,Piperazines ,chemistry.chemical_compound ,Breast cancer ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Cells, Cultured ,Humans ,lcsh:Science ,Protein kinase B ,Triple-negative breast cancer ,Cell Proliferation ,Chemotherapy ,Multidisciplinary ,Cell growth ,business.industry ,lcsh:R ,Cancer ,Cyclin-Dependent Kinase 4 ,Drug Synergism ,Cyclin-Dependent Kinase 6 ,Cell cycle ,medicine.disease ,chemistry ,Cancer research ,lcsh:Q ,Female ,business - Abstract
Triple Negative Breast Cancer (TNBC) is a challenging disease due to the lack of druggable targets; therefore, chemotherapy remains the standard of care and the identification of new targets is a high clinical priority. Alterations in the components of the cell cycle machinery have been frequently reported in cancer; given the success obtained with the CDK4/6 inhibitor palbocicib in ER-positive BC, we explored the potential of combining this drug with chemotherapy in Rb-positive TNBC cell models. The simultaneous combination of palbociclib with paclitaxel exerted an antagonistic effect; by contrast, the sequential treatment inhibited cell proliferation and increased cell death more efficaciously than single treatments. By down-regulating the E2F target c-myc, palbociclib reduced HIF-1α and GLUT-1 expression, and hence glucose uptake and consumption both under normoxic and hypoxic conditions. Importantly, these inhibitory effects on glucose metabolism were enhanced by palbociclib/paclitaxel sequential combination; the superior efficacy of such combination was ascribed to the ability of paclitaxel to inhibit palbociclib-mediated induction of AKT and to further down-regulate the Rb/E2F/c-myc signaling. Our results suggest that the efficacy of standard chemotherapy can be significantly improved by a pre-treatment with palbociclib, thus offering a better therapeutic option for Rb-proficient TNBC.
- Published
- 2019
27. Abstract 3011: Cancer of Unknown Primary: novel therapeutic opportunities from patient-derived cell cultures and in vivo models
- Author
-
Andrea Cavazzoni, Maria Naddeo, Stefano Cairo, Noemi Laprovitera, Claudia Fumarola, Manuela Ferracin, Andrea Ardizzoni, Elisa Porcellini, Giulia Gallerani, Francesco Gelsomino, and Irene Salamon
- Subjects
Cancer Research ,Oncology ,Cancer of unknown primary ,In vivo ,business.industry ,Cell culture ,Cancer research ,Medicine ,business - Abstract
Patients with occult primary cancer or cancer of unknown primary (CUP) carry the double burden of an aggressive disease and reduced access to therapies, which are typically tumor-type oriented. Moreover, experimental in-vitro and in-vivo models summing up CUP features and heterogeneity are currently unavailable. We derived and expanded two CUP cell lines, and corresponding patient-derived xenografts (PDXs), from ascites tumor cells. CUP cell lines, one spontaneously growing as organoid, and PDXs were subjected to histological, immune-phenotypical, molecular and genomic (CNV and target sequencing) characterization to show their ability to recapitulate the original tumor. MicroRNA profile and genomic information were used to identify a possible primary site and patient-specific therapeutic approaches. Data on CUP most frequently altered genes were used to design a custom CUP-specific, 92-gene panel for target NGS analysis with Sure Select XS technology (Agilent Technology). Drug-screening assays were performed in vitro and in vivo on two models targeting the most promising actionable pathways. We obtained unexpected and promising results on drug combinations efficacy. This study brings personalized therapy closer to CUP patients and paves the way to future applications of personalized medicine for metastatic patients with adverse prognosis. This work was supported by grants from the Italian Association for Cancer Research (AIRC) to MF (IG 18464). Citation Format: Noemi Laprovitera, Claudia Fumarola, Elisa Porcellini, Stefano Cairo, Giulia Gallerani, Francesco Gelsomino, Irene Salamon, Maria Naddeo, Andrea Ardizzoni, Andrea Cavazzoni, Manuela Ferracin. Cancer of Unknown Primary: novel therapeutic opportunities from patient-derived cell cultures and in vivo models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 3011.
- Published
- 2021
- Full Text
- View/download PDF
28. Immunotherapeutic Approaches in Malignant Pleural Mesothelioma
- Author
-
Mara Bonelli, Claudia Fumarola, Silvia Zoppi, Roberta Alfieri, and Rita Terenziani
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,VISTA ,Review ,Disease ,PD-1/PD-L1 ,immune checkpoint inhibitors ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,malignant pleural mesothelioma ,RC254-282 ,Cisplatin ,Chemotherapy ,business.industry ,Clinical study design ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,Chimeric antigen receptor ,030104 developmental biology ,Pemetrexed ,CTLA-4 ,030220 oncology & carcinogenesis ,immunotherapy ,business ,medicine.drug - Abstract
Simple Summary Immune checkpoint inhibitors (ICIs) have emerged as a very promising therapeutic option for the treatment of many difficult-to-treat cancers and a number of clinical trials have explored their efficacy in malignant pleural mesothelioma patients. ICIs were initially evaluated in the salvage setting, resulting in a modest activity, not superior to chemotherapy. However, in the last year the combination of nivolumab and ipilimumab as first line treatment has proved a superior efficacy compared to chemotherapy especially in the non-epithelioid subtype, obtaining the FDA approval in October 2020. Encouraging results are also emerging from other immunological approaches that take advantage of tumor-specific antigens, such as advanced cell-based therapies with the CAR-T cells and tumor vaccines. Abstract Malignant pleural mesothelioma (MPM) is a rare and aggressive malignant disease affecting the mesothelium, commonly associated to asbestos exposure. The current therapeutic actions, based on cisplatin/pemetrexed treatment, are limited due to the late stage at which most patients are diagnosed and to the intrinsic chemo-resistance of the tumor. Another relevant point is the absence of approved therapies in the second line setting following progression of MPM after chemotherapy. Considering the poor prognosis of the disease and the fact that the incidence of this tumor is expected to increase in the next decade, novel therapeutic approaches are urgently needed. In the last few years, several studies have investigated the efficacy and safety of immune-checkpoint inhibitors (ICIs) in the treatment of unresectable advanced MPM, and a number of trials with immunotherapeutic agents are ongoing in both first line and second line settings. In this review, we describe the most promising emerging immunotherapy treatments for MPM (ICIs, engineered T cells to express chimeric antigen receptors (CARs), dendritic cells (DCs) vaccines), focusing on the biological and immunological features of this tumor as well as on the issues surrounding clinical trial design.
- Published
- 2021
- Full Text
- View/download PDF
29. Efficacy of the CDK4/6 Dual Inhibitor Abemaciclib in EGFR-Mutated NSCLC Cell Lines with Different Resistance Mechanisms to Osimertinib
- Author
-
Graziana Digiacomo, Maricla Galetti, Daniele Cretella, Marcello Tiseo, Silvia La Monica, Andrea Cavazzoni, Roberta Minari, Pier Giorgio Petronini, Maicol Mancini, Mara Bonelli, Roberta Alfieri, Claudia Fumarola, and Francesco Volta
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,abemaciclib ,lcsh:RC254-282 ,Article ,Tyrosine-kinase inhibitor ,resistance ,CDK4/6 inhibitors ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cyclin-dependent kinase ,medicine ,Osimertinib ,Epidermal growth factor receptor ,Abemaciclib ,non-small cell lung cancer ,biology ,Kinase ,Cell growth ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,chemistry ,Cell culture ,osimertinib ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,epidermal growth factor receptor - Abstract
Simple Summary Osimertinib, a third-generation irreversible epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), has shown marked clinical benefit for non-small cell lung cancer (NSCLC) patients with EGFR activating mutations. However, resistance to osimertinib inevitably develops and heterogeneous mechanisms of acquired resistance have been documented. Therefore, new strategies to bypass resistance are urgently needed. In this study, we investigated the potential activity of abemaciclib as second-line therapeutic approach after osimertinib progression and the effect of combining abemaciclib with osimertinib on the appearance of resistance in osimertinib-sensitive models. Abstract Abemaciclib is an inhibitor of cyclin-dependent kinases (CDK) 4 and 6 that inhibits the transition from the G1 to the S phase of the cell cycle by blocking downstream CDK4/6-mediated phosphorylation of Rb. The effects of abemaciclib alone or combined with the third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) osimertinib were examined in a panel of PC9 and HCC827 osimertinib-resistant non-small cell lung cancer (NSCLC) cell lines carrying EGFR-dependent or -independent mechanisms of intrinsic or acquired resistance. Differently from sensitive cells, all the resistant cell lines analyzed maintained p-Rb, which may be considered as a biomarker of osimertinib resistance and a potential target for therapeutic intervention. In these models, abemaciclib inhibited cell growth, spheroid formation, colony formation, and induced senescence, and its efficacy was not enhanced in the presence of osimertinib. Interestingly, in osimertinib sensitive PC9, PC9T790M, and H1975 cells the combination of abemaciclib with osimertinib significantly inhibited the onset of resistance in long-term experiments. Our findings provide a preclinical support for using abemaciclib to treat resistance in EGFR mutated NSCLC patients progressed to osimertinib either as single treatment or combined with osimertinib, and suggest the combination of osimertinib with abemaciclib as a potential approach to prevent or delay osimertinib resistance in first-line treatment.
- Published
- 2020
- Full Text
- View/download PDF
30. Overcoming palbociclib resistance by combined treatment with PI3K/AKT/mTOR inhibitors in mesothelioma cells
- Author
-
Andrea Cavazzoni, Graziana Digiacomo, Claudia Fumarola, Daniele Cretella, Pier Giorgio Petronini, Mara Bonelli, Silvia La Monica, and Roberta Alfieri
- Subjects
Combined treatment ,business.industry ,Cancer research ,Medicine ,Mesothelioma ,Palbociclib ,business ,medicine.disease ,Discovery and development of mTOR inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway - Published
- 2018
- Full Text
- View/download PDF
31. The anti-tumor efficacy of CDK4/6 inhibition is enhanced by the combination with PI3K/AKT/mTOR inhibitors through impairment of glucose metabolism in TNBC cells
- Author
-
Alessandra Biondi, Daniele Cretella, Pier Giorgio Petronini, Mara Bonelli, Claudia Fumarola, Roberta Alfieri, Silvia La Monica, Andrea Cavazzoni, Daniele Generali, Andrea Ravelli, Graziana Digiacomo, Cretella, Daniele, Ravelli, Andrea, Fumarola, Claudia, La Monica, Silvia, Digiacomo, Graziana, Cavazzoni, Andrea, Alfieri, Roberta, Biondi, Alessandra, Generali, Daniele, Bonelli, Mara, and Petronini, Pier Giorgio
- Subjects
0301 basic medicine ,Cancer Research ,Pyridines ,Triple Negative Breast Neoplasms ,Piperazines ,0302 clinical medicine ,Phosphoinositide-3 Kinase Inhibitors ,Glucose metabolism ,biology ,Cell Death ,Chemistry ,TOR Serine-Threonine Kinases ,Cell Cycle ,Drug Synergism ,PI3K/mTOR inhibitor ,Cell cycle ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncology ,030220 oncology & carcinogenesis ,Female ,CDK4/6 Inhibition ,medicine.drug ,Palbociclib ,lcsh:RC254-282 ,03 medical and health sciences ,Triple-negative breast cancer ,Cell Line, Tumor ,medicine ,Humans ,Protein kinase B ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,PI3K/mTOR inhibitors ,Fulvestrant ,Dose-Response Relationship, Drug ,Cell growth ,Research ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,030104 developmental biology ,Glucose ,CDK4/6 inhibition ,Cancer research ,biology.protein ,Cyclin-dependent kinase 6 ,Energy Metabolism ,Proto-Oncogene Proteins c-akt - Abstract
Background Cell cycle regulators have gain attention as potential targets for anticancer therapy. Palbociclib is a selective inhibitor of the cyclin-dependent kinases 4 and 6 (CDK4/6), which coordinate the G1-S transition. Palbociclib is currently approved for the treatment of hormone receptor positive, HER2-negative advanced breast cancer (BC) in association with letrozole or fulvestrant. In contrast, its efficacy in triple negative BC (TNBC), either alone or in combined therapies, has not been fully investigated to date. Methods Here we evaluated the potential of combining palbociclib with PI3K/mTOR inhibitors in Rb-proficient TNBC cells comparing different schedules of treatment: simultaneous, sequential, or sequential combined treatment (pre-incubation with palbociclib followed by exposure to both palbociclib and PI3K/mTOR inhibitors). We assessed the effects on cell proliferation, cell death, and cell cycle distribution, and looked at the impact of such treatments on glucose metabolism. Results Palbociclib exerted cytostatic effects in Rb-positive TNBC cells, inducing a reversible blockade in G0/G1 cell cycle phase associated with down-regulation of CDK6, Rb, and c-myc expression and/or activity. Palbociclib treatment induced AKT signaling, providing a rationale for its combination with PI3K/mTOR inhibitors. The simultaneous or sequential treatment resulted in an additive inhibition of cell proliferation. On the other hand, the sequential combined treatment in which palbociclib was maintained also during exposure to PI3K/mTOR inhibitors gave rise to synergistic anti-proliferative and pro-apoptotic effects, by inhibiting both CDK4/6/Rb/myc and PI3K/mTOR signaling. Interestingly, the inhibition of the Rb/E2F/myc axis mediated by palbociclib resulted in a significant down-regulation of glucose metabolism; most importantly, these inhibitory effects were enhanced by the combination of palbociclib with BYL719 (specific inhibitor of the p110α PI3K-subunit), which promoted a stronger inhibition of GLUT-1 glucose transporter expression, glucose uptake and consumption in comparison with individual treatments, under both normoxic and hypoxic conditions. Conclusions Combination of palbociclib with PI3K/mTOR inhibitors may represent a promising therapeutic option for the treatment of Rb-proficient TNBC, with the sequential combined schedule showing a superior efficacy over the other schedules. In addition our results demonstrate that the impairment of glucose metabolism may contribute to the anti-tumor activity of such drug combinations.
- Published
- 2018
32. Enhanced efficacy of AKT and FAK kinase combined inhibition in squamous cell lung carcinomas with stable reduction in PTEN
- Author
-
Roberta Alfieri, Daniele Cretella, Claudia Fumarola, Mara Bonelli, Costanza Lagrasta, Nele Van Der Steen, Godefridus J. Peters, Elisa Giovannetti, Andrea Cavazzoni, Pier Giorgio Petronini, Marcello Tiseo, Silvia La Monica, Graziana Digiacomo, Denise Madeddu, Federico Quaini, Andrea Ravelli, Andrea Ardizzoni, Rocco Sciarrillo, Cavazzoni, Andrea, La Monica, Silvia, Alfieri, Roberta, Ravelli, Andrea, Van Der Steen, Nele, Sciarrillo, Rocco, Madeddu, Denise, Lagrasta, Costanza Anna Maria, Quaini, Federico, Bonelli, Mara, Fumarola, Claudia, Cretella, Daniele, Digiacomo, Graziana, Tiseo, Marcello, Peters, Godefridus J, Ardizzoni, Andrea, Petronini, Pier Giorgio, Giovannetti, Elisa, Medical oncology laboratory, CCA - Cancer Treatment and quality of life, Hematology laboratory, AGEM - Digestive immunity, and AGEM - Re-generation and cancer of the digestive system
- Subjects
0301 basic medicine ,PTEN ,Pathology ,medicine.medical_specialty ,Cell ,Buparlisib ,PI3K ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Protein kinase B ,PI3K/AKT/mTOR pathway ,FAK ,biology ,target therapy ,business.industry ,Cell growth ,Kinase ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,squamous lung carcinoma ,Phosphorylation ,business ,Research Paper - Abstract
Squamous cell lung carcinoma (SCC) accounts for 30% of patients with NSCLC and to date, no molecular targeted agents are approved for this type of tumor. However, recent studies have revealed several oncogenic mutations in SCC patients, including an alteration of the PI3K/AKT pathway, i.e. PI3K point mutations and amplification, AKT mutations and loss or reduced PTEN expression. Prompted by our observation of a correlation between PTEN loss and FAK phosphorylation in a cohort of patients with stage IV SCC, we evaluated the relevance of PTEN loss in cancer progression as well as the efficacy of a new combined treatment with the pan PI3K inhibitor buparlisip and the FAK inhibitor defactinib. An increase in AKT and FAK phosphorylation, associated with increased proliferation and invasiveness, paralleled by the acquisition of mesenchymal markers, and overexpression of the oncomir miR-21 were observed in SKMES-1-derived cell clones with a stable reduction of PTEN. Notably, the combined treatment induced a synergistic inhibition of cell proliferation, and a significant reduction in cell migration and invasion only in cells with reduced PTEN. The molecular mechanisms underlying these findings were unraveled using a specific RTK array that showed a reduction in phosphorylation of key kinases such as JNK, GSK-3 α/β, and AMPK-α2, due to the concomitant decrease in AKT and FAK activation. In conclusion, the combination of buparlisib and defactinib was effective against cells with reduced PTEN and warrants further studies as a novel therapeutic strategy for stage IV SCC patients with loss of PTEN expression.
- Published
- 2017
- Full Text
- View/download PDF
33. Pure anti-tumor effect of zoledronic acid in naïve bone-only metastatic and locally advanced breast cancer: proof from the 'biological window therapy'
- Author
-
Elena A Takano, Giulia Brugnoli, Manuela Milan, Laura B Pritzker, Daniele Santini, Sergio Venturini, Alberto Bottini, Carla Strina, Mara Bonelli, Daniele Generali, Claudia Fumarola, Chiara Foroni, Pier Giorgio Petronini, Vanessa Zanoni, Ramona Bertoni, Mariarosa Cappelletti, Giuseppina Ferrero, Mara Maldotti, Amadeo M. Parissenti, Stephen B. Fox, Daniele Andreis, Kenneth P.H. Pritzker, Foroni, Chiara, Milan, Manuela, Strina, Carla, Cappelletti, Mariarosa, Fumarola, Claudia, Bonelli, Mara, Bertoni, Ramona, Ferrero, Giuseppina, Maldotti, Mara, Takano, Elena, Andreis, Daniele, Venturini, Sergio, Brugnoli, Giulia, Petronini, Pier Giorgio, Zanoni, Vanessa, Pritzker, Laura, Pritzker, Kenneth, Parissenti, Amadeo, Santini, Daniele, Fox, Stephen B., Bottini, Alberto, and Generali, Daniele
- Subjects
Oncology ,BREAST CANCER ,CIRCULATING TUMOR CELLS ,KI67 ,WINDOW THERAPY ,ZOLEDRONIC ACID ,ADULT ,AGED ,80 AND OVER ,BONE DENSITY CONSERVATION AGENTS ,BONE NEOPLASMS ,BREAST NEOPLASMS ,DIPHOSPHONATES ,FEMALE ,HUMANS ,IMIDAZOLES ,IMMUNOHISTOCHEMISTRY ,MIDDLE AGED ,NEOPLASTIC CELLS ,CIRCULATING ,ONCOLOGY ,CANCER RESEARCH ,Cancer Research ,Pathology ,CD34 ,Neoplastic Cells ,Basal (phylogenetics) ,Breast cancer ,Circulating tumor cell ,Window therapy ,80 and over ,Circulating ,Aged, 80 and over ,Bone Density Conservation Agents ,Diphosphonates ,BREAST CANCER, CIRCULATING TUMOR CELLS, KI67, WINDOW THERAPY, ZOLEDRONIC ACID, ADULT, AGED, AGED, 80 AND OVER, BONE DENSITY CONSERVATION AGENTS, BONE NEOPLASMS, BREAST NEOPLASMS, DIPHOSPHONATES, FEMALE, HUMANS, IMIDAZOLES, IMMUNOHISTOCHEMISTRY, MIDDLE AGED, NEOPLASTIC CELLS, CIRCULATING, ONCOLOGY, CANCER RESEARCH ,Imidazoles ,Bone metastasis ,Middle Aged ,Neoplastic Cells, Circulating ,Immunohistochemistry ,Diphosphonate ,Settore SECS-S/01 - STATISTICA ,Female ,Ki67 ,Breast Neoplasm ,Human ,medicine.drug ,Adult ,medicine.medical_specialty ,Bone Density Conservation Agent ,Bone Neoplasms ,Breast Neoplasms ,Bone Neoplasm ,Circulating tumor cells ,Zoledronic acid ,Aged ,Humans ,In vivo ,Internal medicine ,medicine ,Imidazole ,business.industry ,medicine.disease ,Apoptosis ,business - Abstract
The study investigated the anti-tumour effect of zoledronic acid (ZA) administered alone in a biological window therapy in naïve bone-only metastatic and locally advanced breast cancer (LABC) patients. 33 patients with LABC (Group 1) and 20 patients with a first diagnosis of bone metastasis only (Group 2) received 4 mg single dose of ZA, 14 days (biological window) before starting any treatment. In Group 1, Ki67, CD34, p53/bcl-2 and caspase 3 expression along with the adenosine triphosphate (ATP) levels and RNA disruption index were evaluated as markers of tumor growth in tumour specimens obtained before and after ZA administration (basal, day 14). In Group 2, the total enumeration of circulating tumour cells (CTCs), and of M30+ve CTCs along with the soluble marker of cell death (M30/M65) were carried-out as markers of tumor dissemination at baseline, at 48 h and day 14th. In Group 1, there was a significant reduction in Ki67, CD34, bcl-2 expression after 14 days ZA based-treatment (p = 0.0032; p = 0.0001, p < 0.00001 respectively). ZA showed a significant increase of RNA disruption (p < 0.0076). In Group 2, we observed a significant reduction of CTCs number after 48 h (p = 0.0012), followed by a significant rebound at 14 days (p = 0.012). The apoptotic CTCs/M30+ve and M65 levels significantly increased under treatment (p = 0.018 and p = 0.039 respectively) after drug administration when compared to the baseline. These results are the first prospective in vivo data showing the direct pure anti-tumour effect (either on the tumour cell or on CTCs) of ZA.
- Published
- 2014
- Full Text
- View/download PDF
34. Enhancement of the anti-tumor activity of FGFR1 inhibition in squamous cell lung cancer by targeting downstream signaling involved in glucose metabolism
- Author
-
Marcello Tiseo, Elisabetta Barocelli, Federico Quaini, Giulia Mazzaschi, Roberta Alfieri, Andrea Cavazzoni, Mara A. Bonelli, Daniele Cretella, Claudia Fumarola, Pier Giorgio Petronini, Angela Falco, Silvia La Monica, Graziana Digiacomo, Cristina Caffarra, Andrea Ardizzoni, Denise Madeddu, Valentina Vivo, Fumarola, Claudia, Cretella, Daniele, La Monica, Silvia, Bonelli, Mara A., Alfieri, Roberta, Caffarra, Cristina, Quaini, Federico, Madeddu, Denise, Falco, Angela, Cavazzoni, Andrea, Digiacomo, Graziana, Mazzaschi, Giulia, Vivo, Valentina, Barocelli, Elisabetta, Tiseo, Marcello, Petronini, Pier Giorgio, and Ardizzoni, Andrea
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,medicine.medical_specialty ,Glucose uptake ,Dovitinib ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,SQCLC ,medicine ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Glucose metabolism ,NVP-BGJ398 ,business.industry ,Cell growth ,Fibroblast growth factor receptor 1 ,Glucose transporter ,stomatognathic diseases ,030104 developmental biology ,Endocrinology ,FGFR1 ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,business ,Research Paper - Abstract
// Claudia Fumarola 1 , Daniele Cretella 1 , Silvia La Monica 1 , Mara A. Bonelli 1 , Roberta Alfieri 1 , Cristina Caffarra 1 , Federico Quaini 1 , Denise Madeddu 1 , Angela Falco 1 , Andrea Cavazzoni 1 , Graziana Digiacomo 1 , Giulia Mazzaschi 1 , Valentina Vivo 2 , Elisabetta Barocelli 2 , Marcello Tiseo 3 , Pier Giorgio Petronini 1, * and Andrea Ardizzoni 4, * 1 Department of Medicine and Surgery, University of Parma, Parma, Italy 2 Food and Drug Department, University of Parma, Parma, Italy 3 Medical Oncology Unit, University Hospital of Parma, Parma, Italy 4 Division of Medical Oncology, Sant’Orsola-Malpighi University Hospital, Bologna, Italy * Joint last authors Correspondence to: Claudia Fumarola, email: claudia.fumarola@unipr.it Mara A. Bonelli, email: mara.bonelli@unipr.it Keywords: FGFR1, glucose metabolism, dovitinib, NVP-BGJ398, SQCLC Received: March 03, 2017 Accepted: June 18, 2017 Published: July 17, 2017 ABSTRACT Fibroblast Growth Factor Receptor (FGFR) signaling is a complex pathway which controls several processes, including cell proliferation, survival, migration, and metabolism. FGFR1 signaling is frequently deregulated via amplification/over-expression in NSCLC of squamous histotype (SQCLC), however its inhibition has not been successfully translated in clinical setting. We determined whether targeting downstream signaling implicated in FGFR1 effects on glucose metabolism potentiates the anti-tumor activity of FGFR1 inhibition in SQCLC. In FGFR1 amplified/over-expressing SQCLC cell lines, FGF2-mediated stimulation of FGFR1 under serum-deprivation activated both MAPK and AKT/mTOR pathways and increased glucose uptake, glycolysis, and lactate production, through AKT/mTOR-dependent HIF-1α accumulation and up-regulation of GLUT-1 glucose transporter. These effects were hindered by PD173074 and NVP-BGJ398, selective FGFR inhibitors, as well as by dovitinib, a multi-kinase inhibitor. Glucose metabolism was hampered by the FGFR inhibitors also under hypoxic conditions, with consequent inhibition of cell proliferation and viability. In presence of serum, glucose metabolism was impaired only in cell models in which FGFR1 inhibition was associated with AKT/mTOR down-regulation. When the activation of the AKT/mTOR pathway persisted despite FGFR1 down-regulation, the efficacy of NVP-BGJ398 could be significantly improved by the combination with NVP-BEZ235 or other inhibitors of this signaling cascade, both in vitro and in xenotransplanted nude mice. Collectively our results indicate that inhibition of FGFR1 signaling impacts on cancer cell growth also by affecting glucose energy metabolism. In addition, this study strongly suggests that the therapeutic efficacy of FGFR1 targeting molecules in SQCLC may be implemented by combined treatments tackling on glucose metabolism.
- Published
- 2017
35. New therapeutic strategies for malignant pleural mesothelioma
- Author
-
Roberta Alfieri, Claudia Fumarola, Mara A. Bonelli, and Silvia La Monica
- Subjects
0301 basic medicine ,Oncology ,Mesothelioma ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Pleural Neoplasms ,Antineoplastic Agents ,Biochemistry ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,medicine ,Humans ,Progression-free survival ,Pleural Neoplasm ,Lung cancer ,Pharmacology ,business.industry ,Mesothelioma, Malignant ,medicine.disease ,Regimen ,030104 developmental biology ,Pemetrexed ,030220 oncology & carcinogenesis ,Immunology ,Immunotherapy ,business ,medicine.drug - Abstract
Malignant pleural mesothelioma (MPM) is a rare and aggressive malignant disease affecting the mesothelium, commonly associated to asbestos exposure. Therapeutic actions are limited due to the late stage at which most patients are diagnosed and the intrinsic chemo-resistance of the tumor. The recommended systemic therapy for MPM is cisplatin/pemetrexed regimen with a mean overall survival of about 12months and a median progression free survival of less than 6months. Considering that the incidence of this tumor is expected to increase in the next decade and that its prognosis is poor, novel therapeutic approaches are urgently needed. For some tumors, such as lung cancer and breast cancer, druggable oncogenic alterations have been identified and targeted therapy is an important option for these patients. For MPM, clinical guidelines do not recommend biological targeted therapy, mainly because of poor target definition or inappropriate trial design. Further studies are required for a full comprehension of the molecular pathogenesis of MPM and for the development of new target agents. This review updates pre-clinical and clinical data on the efficacy of targeted therapy and immune checkpoint inhibition in the treatment of mesothelioma. Finally, future perspectives in this deadly disease are also discussed.
- Published
- 2016
36. Physico-chemical characterization and biological evaluation of two fibroin materials
- Author
-
Luca Ampollini, Paola Segnana, Anna Maria Cantoni, Silvia La Monica, Francesca Gussago, Lucia Verin, Claudia Fumarola, Claudio Migliaresi, Antonella Motta, and Giovanna Bucci
- Subjects
Morphology (linguistics) ,biology ,Biocompatibility ,Chemistry ,Formic acid ,fungi ,Biomedical Engineering ,Medicine (miscellaneous) ,Fibroin ,Matrix (biology) ,biology.organism_classification ,Biomaterials ,chemistry.chemical_compound ,Chemical engineering ,Tissue engineering ,Bombyx mori ,Layer (electronics) - Abstract
Silk fibroin fibres from two different sources, Bombyx mori pure-breed silkworms and polyhybrid cross-bred silkworm cocoons, were treated with formic acid under planar stirring conditions to prepare non-woven nets. The treatment partially dissolved the fibres, which bound together and formed a non-woven micrometric net with fibres coated by a thin layer of low molecular weight fibroin matrix. The starting fibres, net materials and fibroin coating layer were characterized in terms of amino acid composition, molecular weight and calorimetric properties. In vitro cell culture tests with rat fibroblasts were performed to investigate cell proliferation, morphology and spreading. Moreover, host-rat fibroblasts were preseeded on the afore-mentioned nets and implanted in the thorax of rats for histological analysis. In spite of the chemical differences between the two starting fibroins, the response of the said materials in vitro and in vivo were very similar. These results suggest that the outcome is likely correlated with the modification of the processing technique; that during the formation of the net, a thin gel layer of similar amino acid composition was formed on the fibroin fibres. Copyright © 2012 John Wiley & Sons, Ltd.
- Published
- 2012
- Full Text
- View/download PDF
37. Functional characterization of gefitinib uptake in non-small cell lung cancer cell lines
- Author
-
Silvia La Monica, Pier Giorgio Petronini, Roberta Alfieri, Mara Bonelli, Maricla Galetti, Marco Mor, Antonio Mutti, Claudia Fumarola, Marcello Tiseo, Roberta Andreoli, Andrea Cavazzoni, Giuseppe De Palma, Andrea Ardizzoni, Paola Mozzoni, GALETTI M, ALFIERI R.R, CAVAZZONI A, LA MONICA S, BONELLI M, FUMAROLA C, MOZZONI P, DE PALMA G, ANDREOLI R, MUTTI A, MOR M, TISEO M, ARDIZZONI A, and PETRONINI P.G
- Subjects
medicine.medical_specialty ,Lung Neoplasms ,Organic Cation Transport Proteins ,medicine.drug_class ,Cell ,Antineoplastic Agents ,Biology ,Kidney ,Biochemistry ,Tyrosine-kinase inhibitor ,Gefitinib ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Internal medicine ,Extracellular ,medicine ,Humans ,heterocyclic compounds ,RNA, Messenger ,Epidermal growth factor receptor ,Phosphorylation ,Antibodies, Blocking ,skin and connective tissue diseases ,neoplasms ,Pharmacology ,Organic Cation Transporter 2 ,Kidney metabolism ,Hydrogen-Ion Concentration ,respiratory tract diseases ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Endocrinology ,Drug Resistance, Neoplasm ,Cell culture ,Quinazolines ,Cancer research ,biology.protein ,Drug Screening Assays, Antitumor ,lung cancer, EGFR, gefitinib, uptake ,Intracellular ,Octamer Transcription Factor-1 ,medicine.drug - Abstract
Gefitinib, an inhibitor of epidermal growth factor receptor tyrosine kinase, has been developed and approved for treatment of advanced non-small cell lung cancer (NSCLC). In this study, we investigated the uptake of gefitinib in gefitinib-sensitive and -resistant NSCLC cell lines. The transport system was temperature-dependent, indicative of an active process and sodium- and potential-independent. Moreover, high cell densities and low extracellular pH significantly reduced the uptake of gefitinib. Inhibitors of the human organic cation transporter 1 (hOCT1) significantly decreased gefitinib uptake; however, gefitinib was not a substrate for hOCT1 or hOCT2 in overexpressing HEK293 cells. Interestingly, gefitinib significantly reduced uptake of the hOCT prototypical substrate MPP suggesting that gefitinib may exert an inhibitory effect on the intracellular accumulation of drugs transported by hOCT1 and hOCT2. After 15min of treatment at 1microM (the maximum plasma concentration of gefitinib obtained at the clinically relevant dose) gefitinib accumulated within the cell in resistant-cell lines at concentrations similar or even higher than in gefitinib-sensitive cells tending to rule out an alteration in drug uptake as a mechanism of resistance to gefitinib treatment. Moreover, our results suggest that the extrusion of lactate by crowded cells may contribute in decreasing the pH, which in turn can influence the uptake of gefinitib and as a result the inhibition of EGFR autophosphorylation.
- Published
- 2010
- Full Text
- View/download PDF
38. Everolimus restores gefitinib sensitivity in resistant non-small cell lung cancer cell lines
- Author
-
Pier Giorgio Petronini, Matteo Goldoni, Sara Tagliaferri, Maricla Galetti, Marzia Capelletti, Andrea Ardizzoni, Marcello Tiseo, Mara Bonelli, Daniele Generali, Silvia La Monica, Roberta Alfieri, Antonio Mutti, Andrea Cavazzoni, Claudia Fumarola, La Monica S, Galetti M, Alfieri RR, Cavazzoni A, Ardizzoni A, Tiseo M, Capelletti M, Goldoni M, Tagliaferri S, Mutti A, Fumarola C, Bonelli M, Generali D, Petronini PG, La Monica, Silvia, Galetti, Maricla, Alfieri, Roberta R, Cavazzoni, Andrea, Ardizzoni, Andrea, Tiseo, Marcello, Capelletti, Marzia, Goldoni, Matteo, Tagliaferri, Sara, Mutti, Antonio, Fumarola, Claudia, Bonelli, Mara, Generali, Daniele, and Petronini, Pier Giorgio
- Subjects
Lung Neoplasms ,Biochemistry ,Tyrosine-kinase inhibitor ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,heterocyclic compounds ,Epidermal growth factor receptor ,Phosphorylation ,Everolimus, gefitinib, non-small cell lung cancer, cell lines ,0303 health sciences ,Drug Synergism ,Gefitinib ,3. Good health ,ErbB Receptors ,Everolimu ,030220 oncology & carcinogenesis ,Erlotinib ,Lung cancer ,Immunosuppressive Agents ,Signal Transduction ,medicine.drug ,medicine.medical_specialty ,medicine.drug_class ,EGFR ,Antineoplastic Agents ,P70-S6 Kinase 1 ,Biology ,03 medical and health sciences ,Cell Line, Tumor ,Internal medicine ,medicine ,Humans ,Everolimus ,neoplasms ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Sirolimus ,Pharmacology ,Ribosomal Protein S6 Kinases ,medicine.disease ,respiratory tract diseases ,Endocrinology ,Drug Resistance, Neoplasm ,Quinazolines ,Cancer research ,biology.protein - Abstract
The epidermal growth factor receptor(EGFR) is a validated target for therapy in non-small cell lung cancer (NSCLC). Most patients, however, either do not benefit or develop resistance to specific inhibitors of the EGFR tyrosine kinase activity, such as gefitinib or erlotinib. The mammalian target of rapamycin (mTOR) is a key intracellular kinase integrating proliferation and survival pathways and has been associated with resistance to EGFR tyrosine kinase inhibitors. In this study, we assessed the effects of combining the mTOR inhibitor everolimus (RAD001) with gefitinib on a panel of NSCLC cell lines characterized by gefitinib resistance and able to maintain S6K phosphorylation after gefitinib treatment. Everolimus plus gefitinib induced a significant decrease in the activation of MAPK and mTOR signaling pathways downstream of EGFR and resulted in a growth-inhibitory effect rather than in an enhancement of cell death. A synergistic effect was observed in those cell lines characterized by high proliferative index and low doubling time. These data suggest that treatment with everolimus and gefitinib might be of value in the treatment of selected NSCLC patients that exhibit high tumor proliferative activity. (C) 2009 Elsevier Inc. All rights reserved. RI goldoni, matteo/E-9153-2011; Mutti, Antonio/C-1095-2011
- Published
- 2009
- Full Text
- View/download PDF
39. Dual mechanisms of action of the 5-benzylidene-hydantoin UPR1024 on lung cancer cell lines
- Author
-
Maricla Galetti, Alessio Lodola, Fabrizio Bordi, Marco Mor, Caterina Carmi, Claudia Fumarola, Pier Giorgio Petronini, Valentina Zuliani, Raffaele Frazzi, Roberta Alfieri, Andrea Cavazzoni, and Mara Bonelli
- Subjects
Cancer Research ,Lung Neoplasms ,medicine.drug_class ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,Apoptosis ,Benzylidene Compounds ,Models, Biological ,Tyrosine-kinase inhibitor ,Gefitinib ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Null cell ,Humans ,Epidermal growth factor receptor ,Phosphorylation ,S phase ,Cell Proliferation ,biology ,Hydantoins ,Drug Synergism ,Cell biology ,ErbB Receptors ,Oncology ,Mechanism of action ,Drug Resistance, Neoplasm ,Quinazolines ,biology.protein ,Tumor Suppressor Protein p53 ,medicine.symptom ,Tyrosine kinase ,Platelet-derived growth factor receptor ,DNA Damage ,medicine.drug - Abstract
In this study, we examined the mechanism of action of the novel epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor 5-benzylidene-hydantoin UPR1024, whose structure was designed to interact at the ATP-binding site of EGFR. The compound had antiproliferative and proapoptotic effects when tested on the non–small cell lung cancer cell line A549. The growth inhibitory effect was associated with an accumulation of the cells in the S phase of the cell cycle. Moreover, UPR1024 induced significant level of DNA strand breaks associated with increased expression of p53 and p21WAF1 proteins, suggesting an additive mechanism of action. The presence of wild-type p53 improved the drug efficacy, although the effect was also detectable in p53 null cells. We also noted apoptotic cell death after treatment with UPR1024 at concentrations above 10 μmol/L for >24 h, with involvement of both the extrinsic and intrinsic pathways. The present data show that UPR1024 may be considered a combi-molecule capable of both blocking EGFR tyrosine kinase activity and inducing genomic DNA damage. UPR1024 or its derivatives might serve as a basis for development of drugs for the treatment of lung cancer in patients resistant to classic tyrosine kinase inhibitors. [Mol Cancer Ther 2008;7(2):361–70]
- Published
- 2008
- Full Text
- View/download PDF
40. Effect of inducible FHIT and p53 expression in the Calu-1 lung cancer cell line
- Author
-
Luca Roz, Michele Rusca, Maricla Galetti, Andrea Cavazzoni, Paolo Carbognani, Roberta Alfieri, Claudia Fumarola, P. G. Petronini, Francesca Andriani, and Gabriella Sozzi
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,Time Factors ,Blotting, Western ,Genetic Vectors ,Cell ,Apoptosis ,Biology ,Transfection ,medicine.disease_cause ,FHIT ,Cell Line, Tumor ,medicine ,Humans ,neoplasms ,Cell Proliferation ,Regulation of gene expression ,Cell growth ,Proto-Oncogene Proteins c-mdm2 ,Blotting, Northern ,Acid Anhydride Hydrolases ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Cell culture ,Cancer cell ,Cancer research ,Tumor Suppressor Protein p53 ,Carcinogenesis - Abstract
Loss of FHIT expression and p53 mutations are critical events in the early stages of lung carcinogenesis. The restoration of Fhit function in FHIT-negative cancer cells has been reported to cause tumour suppression by inhibition of cell proliferation and/or activation of apoptotic pathways. However, the studies designed to elucidate the biological role of Fhit and its potential interaction with p53 have produced conflicting results. We investigated here the effects of the simultaneous restoration of FHIT and p53 in Calu-1 cells by using a hormone-inducible gene expression system. We demonstrate that the restoration of FHIT expression reinforces the anti-proliferative effect associated with the simultaneous replacement of p53. Indeed, a more pronounced inhibition of cell proliferation associated with an earlier and higher induction of p21(waf1) mRNA and protein expression was observed in Fhit/p53-expressing cells compared with cells expressing p53 alone. This effect was not due to Fhit-mediated up-regulation of p53 expression; in fact p53 protein was expressed at the same level in both FHIT-positive and FHIT-negative cell clones. Consistent with this result, Fhit did not affect the expression of MDM2, a protein known to interact directly with p53 and target p53 for proteolytic degradation, thus down-regulating its activity.
- Published
- 2007
- Full Text
- View/download PDF
41. Combination of Gefitinib and Pemetrexed Prevents the Acquisition of TKI Resistance in NSCLC Cell Lines Carrying EGFR-Activating Mutation
- Author
-
Roberta Alfieri, Costanza Lagrasta, Maricla Galetti, Nadia Naldi, Silvia La Monica, Valentina Vivo, Angela Falco, Mara Bonelli, Pier Giorgio Petronini, Andrea Ardizzoni, Denise Madeddu, Claudia Fumarola, Daniele Cretella, Andrea Cavazzoni, Elisabetta Barocelli, Federico Quaini, Marcello Tiseo, Andrea Gervasi, La Monica, Silvia, Madeddu, Denise, Tiseo, Marcello, Vivo, Valentina, Galetti, Maricla, Cretella, Daniele, Bonelli, Mara, Fumarola, Claudia, Cavazzoni, Andrea, Falco, Angela, Gervasi, Andrea, Lagrasta, Costanza Annamaria, Naldi, Nadia, Barocelli, Elisabetta, Ardizzoni, Andrea, Quaini, Federico, Petronini, Pier Giorgio, and Alfieri, Roberta
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,EGFR ,Antineoplastic Agents ,Pemetrexed ,NSCLC ,03 medical and health sciences ,T790M ,0302 clinical medicine ,Gefitinib ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,heterocyclic compounds ,Epidermal growth factor receptor ,skin and connective tissue diseases ,neoplasms ,Protein Kinase Inhibitors ,TKI resistance ,biology ,Cell growth ,business.industry ,Cell cycle ,respiratory tract diseases ,ErbB Receptors ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Quinazolines ,EGFR Activating Mutation ,business ,Tyrosine kinase ,medicine.drug - Abstract
Introduction Development of resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors is a clinical issue in patients with epidermal growth factor receptor gene ( EGFR ) - mutated non–small cell lung cancer (NSCLC). The aim of this study was to investigate the potential of combining gefitinib and pemetrexed in preventing the acquisition of resistance to EGFR tyrosine kinase inhibitors in NSCLC cell lines harboring EGFR exon 19 deletion. Methods The effect of different combinatorial schedules of gefitinib and pemetrexed on cell proliferation, cell cycle, apoptosis, and acquisition of gefitinib resistance in PC9 and HCC827 NSCLC cell lines and in PC9 xenograft models was investigated. Results Simultaneous treatment with gefitinib and pemetrexed enhanced cell growth inhibition and cell death and prevented the appearance of gefitinib resistance mediated by T790M mutation or epithelial-to-mesenchymal transition (EMT) in PC9 and HCC827 cells, respectively. In PC9 cells and in PC9 xenografts the combination of gefitinib and pemetrexed, with different schedules, prevented gefitinib resistance only when pemetrexed was the first treatment, given alone or together with gefitinib. Conversely, when gefitinib alone was administered first and pemetrexed sequentially alternated, a negative interaction was observed and no prevention of gefitinib resistance was documented. The mechanisms of resistance that developed in vivo included T790M mutation and EMT. The induction of EMT was a feature of tumors treated with gefitinib when given before pemetrexed, whereas T790M was recorded only in tumors treated with gefitinib alone. Conclusions The combination of gefitinib and pemetrexed is effective in preventing gefitinib resistance; the application of intermittent treatments requires that gefitinib not be administered before pemetrexed.
- Published
- 2015
42. Creatine as a compatible osmolyte in muscle cells exposed to hypertonic stress
- Author
-
Angelo F. Borghetti, Roberta Alfieri, Claudia Fumarola, Domenica Carnicelli, Kenneth P. Wheeler, Giuseppe De Palma, Federica Vacondio, Pier Giorgio Petronini, Mara Bonelli, Maurizio Brigotti, Andrea Cavazzoni, Claudia Silva, Paola Mozzoni, Piero Sestili, and Antonio Mutti
- Subjects
Taurine ,chemistry.chemical_compound ,chemistry ,Physiology ,Hypertonic Stress ,Osmolyte ,Myocyte ,Tonicity ,Creatine transport ,Biology ,Cycloheximide ,Creatine ,Molecular biology - Abstract
Exposure of C2C12 muscle cells to hypertonic stress induced an increase in cell content of creatine transporter mRNA and of creatine transport activity, which peaked after about 24 h incubation at 0.45 osmol (kg H2O)−1. This induction of transport activity was prevented by addition of either cycloheximide, to inhibit protein synthesis, or of actinomycin D, to inhibit RNA synthesis. Creatine uptake by these cells is largely Na+ dependent and kinetic analysis revealed that its increase under hypertonic conditions resulted from an increase in Vmax of the Na+-dependent component, with no significant change in the Km value of about 75 μmol l−1. Quantitative real-time PCR revealed a more than threefold increase in the expression of creatine transporter mRNA in cells exposed to hypertonicity. Creatine supplementation significantly enhanced survival of C2C12 cells incubated under hypertonic conditions and its effect was similar to that obtained with the well known compatible osmolytes, betaine, taurine and myo-inositol. This effect seemed not to be linked to the energy status of the C2C12 cells because hypertonic incubation caused a decrease in their ATP content, with or without the addition of creatine at 20 mmol l−1 to the medium. This induction of creatine transport activity by hypertonicity is not confined to muscle cells: a similar induction was shown in porcine endothelial cells.
- Published
- 2006
- Full Text
- View/download PDF
43. Cell size reduction induced by inhibition of the mTOR/S6K-signaling pathway protects Jurkat cells from apoptosis
- Author
-
Guido Guidotti, Elena Borra, Claudia Fumarola, Roberta Alfieri, and S La Monica
- Subjects
Morpholines ,Cell ,Apoptosis ,Cytochrome c Group ,P70-S6 Kinase 1 ,Cell Growth Processes ,Biology ,Jurkat cells ,Mitochondrial Proteins ,Jurkat Cells ,Leucine ,medicine ,Humans ,Enzyme Inhibitors ,Phosphorylation ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Cell Size ,Sirolimus ,Cell growth ,TOR Serine-Threonine Kinases ,RPTOR ,Ribosomal Protein S6 Kinases, 70-kDa ,Cell Biology ,Mitochondria ,Cell biology ,medicine.anatomical_structure ,Chromones ,Signal transduction ,Energy Metabolism ,Protein Kinases - Abstract
In Jurkat cells, the decreased cell growth rate associated with a long-lasting deactivation of the mammalian target of rapamycin (mTOR)/p70 ribosomal S6 kinase (S6K)-signaling pathway generates a cell population of progressively reduced cellular mass and size. When promoted by rapamycin as prototype inhibitor, the mTOR deactivation-dependent cell size reduction was associated with slowed, but not suppressed, proliferation. Small-size cells were significantly protected from apoptosis induced by Fas/Apo-1 death-receptor activation (as shown by reduced procaspase cleavage and decreased catalytic activity of relevant caspases) or by stress signals-dependent mitochondrial perturbation (as shown by reduced cleavage of caspase-2, lower dissipation of mitochondrial membrane potential and decreased release of cytochorome c and apoptosis-inducing factor from mitochondria). Protection faded when reactivation of the mTOR/S6K pathway promoted the cell recovery to normal size. These results suggest that cells induced to reduce their mass by the mTOR deactivation-dependent inhibition of cell growth become more resilient to lethal assaults by curbing the cell's suicidal response.
- Published
- 2005
- Full Text
- View/download PDF
44. Amino acid signaling through the mammalian target of rapamycin (mTOR) pathway: Role of glutamine and of cell shrinkage
- Author
-
Guido Guidotti, Claudia Fumarola, and Silvia La Monica
- Subjects
Physiology ,Glutamine ,Clinical Biochemistry ,P70-S6 Kinase 1 ,Biology ,Dephosphorylation ,Jurkat Cells ,Adenosine Triphosphate ,Leucine ,Osmotic Pressure ,Humans ,PI3K/AKT/mTOR pathway ,Cell Size ,Sirolimus ,chemistry.chemical_classification ,Antibiotics, Antineoplastic ,Kinase ,TOR Serine-Threonine Kinases ,Ribosomal Protein S6 Kinases, 70-kDa ,Water ,Cell Biology ,Water-Electrolyte Balance ,Amino acid ,Cell biology ,chemistry ,Biochemistry ,Protein Kinases ,Intracellular ,Signal Transduction - Abstract
Mammalian target of rapamycin (mTOR) mediates a signaling pathway that couples amino acid availability to S6 kinase (S6K) activation, translational initiation and cell growth rate, participating to a versatile checkpoint that inspects the energy status of the cell. The pathway is activated by branched-chain amino acids (BCAA), leucine being the most effective, whereas amino acid dearth and ATP shortage lead to its deactivation. Glutamine- or amino acid-deprivation and hyperosmotic stress induce a fast cell shrinkage (with marked decrease of the intracellular water volume) associated to mTOR-dependent S6K1 dephosphorylation. Using cultured Jurkat cells, we have measured the changes of cell content and intracellular concentration of ATP, of relevant amino acids (BCAA) and of ninhydrin-positive substances (NPS, as measure of NH(2)-bearing organic osmolytes) under conditions that deactivate (leucine-deprivation, glutamine-deprivation, amino acid withdrawal, sorbitol-induced hyperosmotic stress) or reactivate a previously deactivated, mTOR-S6K1 pathway. We have also assessed the mitochondrial function by measurements of mitochondrial transmembrane potential in cells subjected to hypertonic stress. Our results indicate that diverse control signals converge on the mTOR-S6K1 signaling pathway. In the presence of adequate energy resources, the pathway senses the amino acid availability as inward transport of effective amino acids (as BCAA and especially leucine), but its activation occurs only in the presence of an extracellular amino acid complement, with glutamine as obligatory component, and does not tolerate decrements of cell water volume incapable of maintaining adequate intracellular physicochemical conditions.
- Published
- 2005
- Full Text
- View/download PDF
45. Dose-dependent effect of FHIT-inducible expression in Calu-1 lung cancer cell line
- Author
-
Maricla Galetti, Luca Roz, Michele Rusca, Roberta Alfieri, Andrea Cavazzoni, Francesca Andriani, Claudia Fumarola, Gabriella Sozzi, Paolo Carbognani, and Pier Giorgio Petronini
- Subjects
Cancer Research ,Lung Neoplasms ,Genetic Vectors ,Apoptosis ,Biology ,Transfection ,medicine.disease_cause ,Adenoviridae ,Downregulation and upregulation ,Transduction, Genetic ,FHIT ,Cell Line, Tumor ,Gene expression ,Genetics ,medicine ,Humans ,neoplasms ,Molecular Biology ,Cell growth ,Acid Anhydride Hydrolases ,Neoplasm Proteins ,Kinetics ,Cell culture ,Cancer cell ,Cancer research ,Carcinogenesis ,Cell Division - Abstract
Abnormalities in the expression of the tumour suppressor fragile histidine triad (FHIT) gene have been reported in a variety of human tumours, including lung cancer and restoration of its expression in cancer cell lines resulted in the inhibition of proliferation and apoptosis induction. Most of the studies that have assigned a proapoptotic role to the FHIT gene were performed in adenoviral-FHIT-transduced cancer cells expressing high levels of the Fhit protein. The present work was the first study designed to investigate the effects of FHIT gene replacement in a human FHIT-negative non-small-cell lung cancer (NSCLC) cell line (Calu-1) by using a hormone-inducible expression system that allows tight modulation of the transgene expression. Through this approach, we demonstrated that a prolonged induction was required to accumulate the Fhit protein at levels adequate to promote a significant decrease of cell proliferation. Analysis of cell-cycle phase distribution showed an accumulation of cells in the G0/G1 phase and a concomitant decrease in the S phase. Moreover, an upregulation of p21waf1 transcript was found, which could account for the alteration of the cycling properties of the cells. The growth-inhibitory effects observed were not associated with apoptosis appearance, and although in these conditions the Fhit protein content was higher than in normal bronchial human epithelial cells (NHBE), it was still significantly lower than the level capable of inducing apoptosis in Calu-1 cells after adenoviral-mediated FHIT gene transfer. These results indicate that the tumour suppressor properties of Fhit are strictly related to its expression level and show that the Fhit protein has a dose-dependent antiproliferative effect on the Fhit-negative Calu-1 lung cancer cell line.
- Published
- 2004
- Full Text
- View/download PDF
46. Inhibition of PI3K pathway reduces invasiveness and epithelial-to-mesenchymal transition in squamous lung cancer cell lines harboring PIK3CA gene alterations
- Author
-
Maricla Galetti, Costanza Lagrasta, Roberta Alfieri, Denise Madeddu, Cristina Caffarra, Francesca Saccani, Daniele Cretella, Andrea Cavazzoni, Claudia Fumarola, Pier Giorgio Petronini, Andrea Ardizzoni, Pietro Rossetti, Federico Quaini, Angela Falco, Marcello Tiseo, Mara A. Bonelli, Caterina Frati, Silvia La Monica, Bonelli, Mara A, Cavazzoni, Andrea, Saccani, Francesca, Alfieri, Roberta R., Quaini, Federico, La Monica, Silvia, Galetti, Maricla, Cretella, Daniele, Caffarra, Cristina, Madeddu, Denise, Frati, Caterina, Lagrasta, Costanza Annamaria, Falco, Angela, Rossetti, Pietro, Fumarola, Claudia, Tiseo, Marcello, Petronini, Pier Giorgio, and Ardizzoni, Andrea
- Subjects
Morpholine ,Cancer Research ,Lung Neoplasms ,RHOA ,Xenograft Model Antitumor Assay ,Epithelial-Mesenchymal Transition ,Class I Phosphatidylinositol 3-Kinases ,Morpholines ,Quinoline ,Aminopyridines ,Biology ,P110α ,medicine.disease_cause ,Phosphatidylinositol 3-Kinases ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,neoplasms ,Imidazole ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Cell Proliferation ,Mutation ,Cell growth ,Animal ,Medicine (all) ,Mesenchymal stem cell ,Imidazoles ,Xenograft Model Antitumor Assays ,Molecular biology ,Lung Neoplasm ,Thiazoles ,Disease Models, Animal ,Aminopyridine ,Oncology ,Cell culture ,Quinolines ,Cancer research ,biology.protein ,Carcinoma, Squamous Cell ,Phosphatidylinositol 3-Kinase ,Thiazole ,Human ,Signal Transduction - Abstract
A prominent role in the pathogenesis of squamous cell carcinoma of the lung (SQCLC) has been attributed to the aberrant activation of the PI3K signaling pathway, due to amplification or mutations of the p110α subunit of class I phosphatidylinositol 3-kinase (PIK3CA) gene. The aim of our study was to determine whether different genetic alterations of PIK3CA affect the biologic properties of SQCLC and to evaluate the response to specific targeting agents in vitro and in vivo. The effects of NVP-BEZ235, NVP-BKM120, and NVP-BYL719 on two-dimensional/three-dimensional (2D/3D) cellular growth, epithelial-to-mesenchymal transition, and invasiveness were evaluated in E545K or H1047R PIK3CA–mutated SQCLC cells and in newly generated clones carrying PIK3CA alterations, as well as in a xenograft model. PIK3CA mutated/amplified cells showed increased growth rate and enhanced migration and invasiveness, associated with an increased activity of RhoA family proteins and the acquisition of a mesenchymal phenotype. PI3K inhibitors reverted this aggressive phenotype by reducing metalloproteinase production, RhoA activity, and the expression of mesenchymal markers, with the specific PI3K inhibitors NVP-BKM120 and NVP-BYL719 being more effective than the dual PI3K/mTOR inhibitor NVP-BEZ235. A xenograft model of SQCLC confirmed that PIK3CA mutation promotes the acquisition of a mesenchymal phenotype in vivo and proved the efficacy of its specific targeting drug NVP-BYL719 in reducing the growth and the expression of mesenchymal markers in xenotransplanted tumors. These data indicate that PIK3CA mutation/amplification may represent a good predictive feature for the clinical application of specific PI3K inhibitors in SQCLC patients. Mol Cancer Ther; 14(8); 1916–27. ©2015 AACR.
- Published
- 2015
47. Effect of ABCG2/BCRP Expression on Efflux and Uptake of Gefitinib in NSCLC Cell Lines
- Author
-
Cristina Caffarra, Daniele Cretella, Francesca Saccani, Maricla Galetti, Pier Giorgio Petronini, Andrea Cavazzoni, Roberta Alfieri, Claudia Fumarola, Mara A. Bonelli, Antonio Mutti, Silvia La Monica, Roberta Andreoli, Andrea Ardizzoni, Marcello Tiseo, Galetti, Maricla, Petronini, Pier Giorgio, Fumarola, Claudia, Cretella, Daniele, La Monica, Silvia, Bonelli, Mara, Cavazzoni, Andrea, Saccani, Francesca, Caffarra, Cristina, Andreoli, Roberta, Mutti, Antonio, Tiseo, Marcello, Ardizzoni, Andrea, and Alfieri, Roberta R.
- Subjects
Small interfering RNA ,Indoles ,Lung Neoplasms ,Abcg2 ,lcsh:Medicine ,HEK293 Cell ,Tandem Mass Spectrometry ,Carcinoma, Non-Small-Cell Lung ,ATP Binding Cassette Transporter, Subfamily G, Member 2 ,RNA, Small Interfering ,lcsh:Science ,Chromatography, High Pressure Liquid ,Multidisciplinary ,biology ,Medicine (all) ,Gefitinib ,Transfection ,Cell biology ,Neoplasm Proteins ,ErbB Receptors ,embryonic structures ,RNA Interference ,Efflux ,Tyrosine kinase ,Intracellular ,Human ,medicine.drug ,Research Article ,ATP-Binding Cassette Transporter ,Protein Kinase Inhibitor ,Neoplasm Protein ,ATP Binding Cassette Transporter, Sub-Family G, Member 2 ,Cell Line, Tumor ,medicine ,Gene silencing ,Humans ,Protein Kinase Inhibitors ,Biochemistry, Genetics and Molecular Biology (all) ,lcsh:R ,Quinazoline ,Biological Transport ,respiratory tract diseases ,Lung Neoplasm ,HEK293 Cells ,Agricultural and Biological Sciences (all) ,Gene Expression Regulation ,Indole ,Cancer research ,biology.protein ,Quinazolines ,lcsh:Q ,ATP-Binding Cassette Transporters ,Receptor, Epidermal Growth Factor ,sense organs - Abstract
Background BCRP/ABCG2 emerged as an important multidrug resistance protein, because it confers resistance to several classes of cancer chemotherapeutic agents and to a number of novel molecularly-targeted therapeutics such as tyrosine kinase inhibitors. Gefitinib is an orally active, selective EGFR tyrosine kinase inhibitor used in the treatment of patients with advanced non small cell lung cancer (NSCLC) carrying activating EGFR mutations. Membrane transporters may affect the distribution and accumulation of gefitinib in tumour cells; in particular a reduced intracellular level of the drug may result from poor uptake, enhanced efflux or increased metabolism. Aim The present study, performed in a panel of NSCLC cell lines expressing different ABCG2 plasma membrane levels, was designed to investigate the effect of the efflux transporter ABCG2 on intracellular gefitinib accumulation, by dissecting the contribution of uptake and efflux processes. Methods and Results Our findings indicate that gefitinib, in lung cancer cells, inhibits ABCG2 activity, as previously reported. In addition, we suggest that ABCG2 silencing or overexpression affects intracellular gefitinib content by modulating the uptake rather than the efflux. Similarly, overexpression of ABCG2 affected the expression of a number of drug transporters, altering the functional activities of nutrient and drug transport systems, in particular inhibiting MPP, glucose and glutamine uptake. Conclusions Therefore, we conclude that gefitinib is an inhibitor but not a substrate for ABCG2 and that ABCG2 overexpression may modulate the expression and activity of other transporters involved in the uptake of different substrates into the cells.
- Published
- 2014
48. Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer
- Author
-
Claudia Fumarola, Pier Giorgio Petronini, Roberta Alfieri, and Mara Bonelli
- Subjects
Lung Neoplasms ,medicine.medical_treatment ,Antineoplastic Agents ,Pharmacology ,Biochemistry ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Adenocarcinoma of the lung ,Animals ,Humans ,Molecular Targeted Therapy ,Enzyme Inhibitors ,Lung cancer ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Chemotherapy ,business.industry ,TOR Serine-Threonine Kinases ,RPTOR ,medicine.disease ,Neoplasm Proteins ,Drug Resistance, Neoplasm ,Cancer research ,Adenocarcinoma ,Non small cell ,Phosphatidylinositol 3-Kinase ,business ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
While PI3K/AKT/mTOR pathway is altered in a variety of cancers including non small cell lung cancer, abnormalities in this pathway are more common in squamous cell lung carcinoma than in adenocarcinoma of the lung. Moreover, aberrant activation of PI3K/AKT/mTOR pathway is one of the mechanisms of acquired resistance to EGFR-TK inhibitors in patients with adenocarcinoma carrying EGFR activating mutations. Several inhibitors of the PI3K pathway are undergoing evaluation in preclinical and clinical studies. These include pan and selective inhibitors of PI3K, AKT inhibitors, rapamycin and rapalogs for mTOR inhibition, dual mTORC1-mTORC2 inhibitors and dual PI3K-mTOR inhibitors. This review focuses on recent preclinical and clinical data on the efficacy of PI3K pathway inhibitors in NSCLC either as monotherapy approach or in combination with chemotherapy or with drugs that target other signaling transduction pathways.
- Published
- 2014
49. Glutamine deprivation-mediated cell shrinkage induces ligand-independent CD95 receptor signaling and apoptosis
- Author
-
A Zerbini, Claudia Fumarola, and Guido Guidotti
- Subjects
Fas Ligand Protein ,Leukemia, T-Cell ,Fas-Associated Death Domain Protein ,Glutamine ,Cell ,Antineoplastic Agents ,Apoptosis ,HL-60 Cells ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,medicine ,Asparaginase ,Humans ,fas Receptor ,Viability assay ,Receptor ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Cell Size ,Membrane Glycoproteins ,Chemistry ,Cell Membrane ,Cell Biology ,Transfection ,Fas receptor ,Molecular biology ,Clone Cells ,Culture Media ,Cell biology ,Kinetics ,medicine.anatomical_structure ,Mutation ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Carrier Proteins ,Signal Transduction - Abstract
Cell shrinkage and loss of cell viability by apoptosis have been examined in cultured CD95(Fas/Apo-1)-expressing leukemia-derived CEM and HL-60 cells subjected to acute deprivation of glutamine, a major compatible osmolyte engaged in cell volume control. Glutamine deprivation-mediated cell shrinkage promoted a ligand-independent activation of the CD95-mediated apoptotic pathway. Cell transfection with plasmids expressing FADD-DN or v-Flip viral proteins pointed to a functional clustering of CD95 receptors at the cell surface with activation of the 'extrinsic pathway' caspase cascade. Accordingly, cell shrinkage did not induce apoptosis in CD95 receptor-negative lymphoma L1210 cells. Replacement of glutamine with surrogate compatible osmolytes counteracted cell volume decrement and protected the CD95-expressing cells from apoptosis. A glutamine deprivation-dependent cell shrinkage with activation of the CD95-mediated pathway was also observed when asparaginase was added to the medium. Asparagine depletion had no role in this process. The cell-size shrinkage-dependent apoptosis induced by glutamine restriction in CD95-expressing leukemic cells may therefore be of clinical relevance in amidohydrolase enzyme therapies.
- Published
- 2001
- Full Text
- View/download PDF
50. Endocrine Therapy in Breast Cancer
- Author
-
Claudia Fumarola
- Subjects
Oncology ,medicine.medical_specialty ,Breast cancer ,business.industry ,Internal medicine ,medicine ,Endocrine therapy ,business ,medicine.disease - Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.