35 results on '"Alfieri, Roberta R."'
Search Results
2. Epidermal Growth Factor Receptor Intron-1 Polymorphism Predicts Gefitinib Outcome in Advanced Non-small Cell Lung Cancer
- Author
-
Tiseo, Marcello, Capelletti, Marzia, De Palma, Giuseppe, Franciosi, Vittorio, Cavazzoni, Andrea, Mozzoni, Paola, Alfieri, Roberta R., Goldoni, Matteo, Galetti, Maricla, Bortesi, Beatrice, Bozzetti, Cecilia, Loprevite, Maura, Boni, Luca, Camisa, Roberta, Rindi, Guido, Petronini, Pier Giorgio, and Ardizzoni, Andrea
- Published
- 2008
- Full Text
- View/download PDF
3. Creatine as a compatible osmolyte in muscle cells exposed to hypertonic stress
- Author
-
Alfieri, Roberta R., Bonelli, Mara A., Cavazzoni, Andrea, Brigotti, Maurizio, Fumarola, Claudia, Sestili, Piero, Mozzoni, Paola, De Palma, Giuseppe, Mutti, Antonio, Carnicelli, Domenica, Vacondio, Federica, Silva, Claudia, Borghetti, Angelo F., Wheeler, Kenneth P., and Petronini, Pier Giorgio
- Published
- 2006
4. Roles of compatible osmolytes and heat shock protein 70 in the induction of tolerance to stresses in porcine endothelial cells
- Author
-
Alfieri, Roberta R., Petronini, Pier Giorgio, Bonelli, Mara A., Desenzani, Silvia, Cavazzoni, Andrea, Borghetti, Angelo F., and Wheeler, Kenneth P.
- Published
- 2004
5. Combined use of anti-ErbB monoclonal antibodies and erlotinib enhances antibody-dependent cellular cytotoxicity of wild-type erlotinib-sensitive NSCLC cell lines
- Author
-
Cavazzoni Andrea, Alfieri Roberta R, Cretella Daniele, Saccani Francesca, Ampollini Luca, Galetti Maricla, Quaini Federico, Graiani Gallia, Madeddu Denise, Mozzoni Paola, Galvani Elena, La Monica Silvia, Bonelli Mara, Fumarola Claudia, Mutti Antonio, Carbognani Paolo, Tiseo Marcello, Barocelli Elisabetta, Petronini Pier Giorgio, and Ardizzoni Andrea
- Subjects
Lung cancer ,EGFR ,Erlotinib ,Cetuximab ,ADCC ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The epidermal growth factor receptor (EGFR) is an established target for anti-cancer treatment in different tumour types. Two different strategies have been explored to inhibit this pivotal molecule in epithelial cancer development: small molecules TKIs and monoclonal antibodies. ErbB/HER-targeting by monoclonal antibodies such as cetuximab and trastuzumab or tyrosine-kinase inhibitors as gefitinib or erlotinib has been proven effective in the treatment of advanced NSCLC. Results In this study we explored the potential of combining either erlotinib with cetuximab or trastuzumab to improve the efficacy of EGFR targeted therapy in EGFR wild-type NSCLC cell lines. Erlotinib treatment was observed to increase EGFR and/or HER2 expression at the plasma membrane level only in NSCLC cell lines sensitive to the drug inducing protein stabilization. The combined treatment had marginal effect on cell proliferation but markedly increased antibody-dependent, NK mediated, cytotoxicity in vitro. Moreover, in the Calu-3 xenograft model, the combination significantly inhibited tumour growth when compared with erlotinib and cetuximab alone. Conclusion Our results indicate that erlotinib increases surface expression of EGFR and/or HER2 only in EGFR-TKI sensitive NSCLC cell lines and, in turns, leads to increased susceptibility to ADCC both in vitro and in a xenograft models. The combination of erlotinib with monoclonal antibodies represents a potential strategy to improve the treatment of wild-type EGFR NSCLC patients sensitive to erlotinib.
- Published
- 2012
- Full Text
- View/download PDF
6. Metabolism of the EGFR tyrosin kinase inhibitor gefitinib by cytochrome P450 1A1 enzyme in EGFR-wild type non small cell lung cancer cell lines
- Author
-
Alfieri Roberta R, Galetti Maricla, Tramonti Stefano, Andreoli Roberta, Mozzoni Paola, Cavazzoni Andrea, Bonelli Mara, Fumarola Claudia, La Monica Silvia, Galvani Elena, De Palma Giuseppe, Mutti Antonio, Mor Marco, Tiseo Marcello, Mari Ettore, Ardizzoni Andrea, and Petronini Pier
- Subjects
Lung cancer ,EGFR ,gefitinib ,metabolism ,CYP1A1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Gefitinib is a tyrosine kinase inhibitor (TKI) of the epidermal growth factor receptor (EGFR) especially effective in tumors with activating EGFR gene mutations while EGFR wild-type non small cell lung cancer (NSCLC) patients at present do not benefit from this treatment. The primary site of gefitinib metabolism is the liver, nevertheless tumor cell metabolism can significantly affect treatment effectiveness. Results In this study, we investigated the intracellular metabolism of gefitinib in a panel of EGFR wild-type gefitinib-sensitive and -resistant NSCLC cell lines, assessing the role of cytochrome P450 1A1 (CYP1A1) inhibition on gefitinib efficacy. Our results indicate that there is a significant difference in drug metabolism between gefitinib-sensitive and -resistant cell lines. Unexpectedly, only sensitive cells metabolized gefitinib, producing metabolites which were detected both inside and outside the cells. As a consequence of gefitinib metabolism, the intracellular level of gefitinib was markedly reduced after 12-24 h of treatment. Consistent with this observation, RT-PCR analysis and EROD assay showed that mRNA and activity of CYP1A1 were present at significant levels and were induced by gefitinib only in sensitive cells. Gefitinib metabolism was elevated in crowded cells, stimulated by exposure to cigarette smoke extract and prevented by hypoxic condition. It is worth noting that the metabolism of gefitinib in the sensitive cells is a consequence and not the cause of drug responsiveness, indeed treatment with a CYP1A1 inhibitor increased the efficacy of the drug because it prevented the fall in intracellular gefitinib level and significantly enhanced the inhibition of EGFR autophosphorylation, MAPK and PI3K/AKT/mTOR signalling pathways and cell proliferation. Conclusion Our findings suggest that gefitinib metabolism in lung cancer cells, elicited by CYP1A1 activity, might represent an early assessment of gefitinib responsiveness in NSCLC cells lacking activating mutations. On the other hand, in metabolizing cells, the inhibition of CYP1A1 might lead to increased local exposure to the active drug and thus increase gefitinib potency.
- Published
- 2011
- Full Text
- View/download PDF
7. Stabilization of hsp70 mRNA on prolonged cell exposure to hypertonicity
- Author
-
Alfieri, Roberta R, Bonelli, Mara A, Petronini, Pier Giorgio, and Borghetti, Angelo F
- Published
- 2002
- Full Text
- View/download PDF
8. NF-κB drives acquired resistance to a novel mutant-selective EGFR inhibitor
- Author
-
Galvani, Elena, Sun, Jing, Leon, Leticia G, Sciarrillo, Rocco, Narayan, Ravi S, Tjin Tham Sjin, Robert, Lee, Kwangho, Ohashi, Kadoaki, Heideman, Daniëlle A M, Alfieri, Roberta R, Heynen, Guus J, Bernards, René, Smit, Egbert F, Pao, William, Peters, Godefridus J, Giovannetti, Elisa, Galvani, Elena, Sun, Jing, Leon, Leticia G, Sciarrillo, Rocco, Narayan, Ravi S, Tjin Tham Sjin, Robert, Lee, Kwangho, Ohashi, Kadoaki, Heideman, Daniëlle A M, Alfieri, Roberta R, Heynen, Guus J, Bernards, René, Smit, Egbert F, Pao, William, Peters, Godefridus J, and Giovannetti, Elisa
- Published
- 2015
9. Effect of ABCG2/BCRP Expression on Efflux and Uptake of Gefitinib in NSCLC Cell Lines
- Author
-
Galetti, Maricla, primary, Petronini, Pier Giorgio, additional, Fumarola, Claudia, additional, Cretella, Daniele, additional, La Monica, Silvia, additional, Bonelli, Mara, additional, Cavazzoni, Andrea, additional, Saccani, Francesca, additional, Caffarra, Cristina, additional, Andreoli, Roberta, additional, Mutti, Antonio, additional, Tiseo, Marcello, additional, Ardizzoni, Andrea, additional, and Alfieri, Roberta R., additional
- Published
- 2015
- Full Text
- View/download PDF
10. Inhibition of PI3K Pathway Reduces Invasiveness and Epithelial-to-Mesenchymal Transition in Squamous Lung Cancer Cell Lines Harboring PIK3CA Gene Alterations
- Author
-
Bonelli, Mara A., primary, Cavazzoni, Andrea, additional, Saccani, Francesca, additional, Alfieri, Roberta R., additional, Quaini, Federico, additional, La Monica, Silvia, additional, Galetti, Maricla, additional, Cretella, Daniele, additional, Caffarra, Cristina, additional, Madeddu, Denise, additional, Frati, Caterina, additional, Lagrasta, Costanza Annamaria, additional, Falco, Angela, additional, Rossetti, Pietro, additional, Fumarola, Claudia, additional, Tiseo, Marcello, additional, Petronini, Pier Giorgio, additional, and Ardizzoni, Andrea, additional
- Published
- 2015
- Full Text
- View/download PDF
11. NF-κB drives acquired resistance to a novel mutant-selective EGFR inhibitor
- Author
-
Galvani, Elena, primary, Sun, Jing, additional, Leon, Leticia G., additional, Sciarrillo, Rocco, additional, Narayan, Ravi S., additional, Tjin Tham Sjin, Robert, additional, Lee, Kwangho, additional, Ohashi, Kadoaki, additional, Heideman, Daniëlle A.M., additional, Alfieri, Roberta R., additional, Heynen, Guus J., additional, Bernards, René, additional, Smit, Egbert F., additional, Pao, William, additional, Peters, Godefridus J., additional, and Giovannetti, Elisa, additional
- Published
- 2015
- Full Text
- View/download PDF
12. Trastuzumab emtansine is active on HER-2 overexpressing NSCLC cell lines and overcomes gefitinib resistance
- Author
-
Cretella, Daniele, primary, Saccani, Francesca, additional, Quaini, Federico, additional, Frati, Caterina, additional, Lagrasta, Costanza, additional, Bonelli, Mara, additional, Caffarra, Cristina, additional, Cavazzoni, Andrea, additional, Fumarola, Claudia, additional, Galetti, Maricla, additional, La Monica, Silvia, additional, Ampollini, Luca, additional, Tiseo, Marcello, additional, Ardizzoni, Andrea, additional, Petronini, Pier Giorgio, additional, and Alfieri, Roberta R, additional
- Published
- 2014
- Full Text
- View/download PDF
13. Gefitinib Inhibits Invasive Phenotype and Epithelial-Mesenchymal Transition in Drug-Resistant NSCLC Cells with MET Amplification
- Author
-
La Monica, Silvia, primary, Caffarra, Cristina, additional, Saccani, Francesca, additional, Galvani, Elena, additional, Galetti, Maricla, additional, Fumarola, Claudia, additional, Bonelli, Mara, additional, Cavazzoni, Andrea, additional, Cretella, Daniele, additional, Sirangelo, Rita, additional, Gatti, Rita, additional, Tiseo, Marcello, additional, Ardizzoni, Andrea, additional, Giovannetti, Elisa, additional, Petronini, Pier Giorgio, additional, and Alfieri, Roberta R., additional
- Published
- 2013
- Full Text
- View/download PDF
14. Endothelial damage induced by Shiga toxins delivered by neutrophils during transmigration
- Author
-
Brigotti, Maurizio, primary, Tazzari, Pier Luigi, additional, Ravanelli, Elisa, additional, Carnicelli, Domenica, additional, Barbieri, Stefania, additional, Rocchi, Laura, additional, Arfilli, Valentina, additional, Scavia, Gaia, additional, Ricci, Francesca, additional, Bontadini, Andrea, additional, Alfieri, Roberta R, additional, Petronini, Pier Giorgio, additional, Pecoraro, Carmine, additional, Tozzi, Alberto E, additional, and Caprioli, Alfredo, additional
- Published
- 2010
- Full Text
- View/download PDF
15. Dual mechanisms of action of the 5-benzylidene-hydantoin UPR1024 on lung cancer cell lines
- Author
-
Cavazzoni, Andrea, primary, Alfieri, Roberta R., additional, Carmi, Caterina, additional, Zuliani, Valentina, additional, Galetti, Maricla, additional, Fumarola, Claudia, additional, Frazzi, Raffaele, additional, Bonelli, Mara, additional, Bordi, Fabrizio, additional, Lodola, Alessio, additional, Mor, Marco, additional, and Petronini, Pier Giorgio, additional
- Published
- 2008
- Full Text
- View/download PDF
16. Molecular Damage and Induction of Proinflammatory Cytokines in Human Endothelial Cells Exposed to Shiga Toxin 1, Shiga Toxin 2, and α-Sarcin
- Author
-
Brigotti, Maurizio, primary, Carnicelli, Domenica, additional, Ravanelli, Elisa, additional, Vara, Antonio González, additional, Martinelli, Chiara, additional, Alfieri, Roberta R., additional, Petronini, Pier Giorgio, additional, and Sestili, Piero, additional
- Published
- 2007
- Full Text
- View/download PDF
17. Hypertonic Stress and Amino Acid Deprivation Both Increase Expression of mRNA for Amino Acid Transport System A
- Author
-
Alfieri, Roberta R., primary, Bonelli, Mara A., additional, Petronini, Pier Giorgio, additional, Desenzani, Silvia, additional, Cavazzoni, Andrea, additional, Borghetti, Angelo F., additional, and Wheeler, Kenneth P., additional
- Published
- 2004
- Full Text
- View/download PDF
18. Effects of osmolarity, ions and compatible osmolytes on cell-free protein synthesis
- Author
-
BRIGOTTI, Maurizio, primary, PETRONINI, Pier Giorgio, additional, CARNICELLI, Domenica, additional, ALFIERI, Roberta R., additional, BONELLI, Mara A., additional, BORGHETTI, Angelo F., additional, and WHEELER, Kenneth P., additional
- Published
- 2003
- Full Text
- View/download PDF
19. Compatible osmolytes modulate the response of porcine endothelial cells to hypertonicity and protect them from apoptosis
- Author
-
Alfieri, Roberta R., primary, Cavazzoni, Andrea, additional, Petronini, Pier Giorgio, additional, Bonelli, Mara A., additional, Caccamo, Alessandro E., additional, Borghetti, Angelo F., additional, and Wheeler, Kenneth P., additional
- Published
- 2002
- Full Text
- View/download PDF
20. Induction of BGT-1 and amino acid System A transport activities in endothelial cells exposed to hyperosmolarity
- Author
-
Petronini, Pier-Giorgio, primary, Alfieri, Roberta R., additional, Losio, M. Nadia, additional, Caccamo, Alessandro E., additional, Cavazzoni, Andrea, additional, Bonelli, Mara A., additional, Borghetti, Angelo F., additional, and Wheeler, Kenneth P., additional
- Published
- 2000
- Full Text
- View/download PDF
21. Shiga Toxin 1, as DNA Repair Inhibitor, Synergistically Potentiates the Activity of the Anticancer Drug, Mafosfamide, on Raji Cells.
- Author
-
Brigotti, Maurizio, Arfilli, Valentina, Carnicelli, Domenica, Rocchi, Laura, Calcabrini, Cinzia, Ricci, Francesca, Pagliaro, Pasqualepaolo, Tazzari, Pier Luigi, Alfieri, Roberta R., Petronini, Pier Giorgio, and Sestili, Piero
- Subjects
DNA repair ,HEMOLYTIC-uremic syndrome ,DNA alkylation ,CYTOTOXINS ,ESCHERICHIA coli ,BURKITT'S lymphoma ,ANTINEOPLASTIC agents - Abstract
Shiga toxin 1 (Stx1), produced by pathogenic Escherichia coli, targets a restricted subset of human cells, which possess the receptor globotriaosylceramide (Gb3Cer/CD77), causing hemolytic uremic syndrome. In spite of the high toxicity, Stx1 has been proposed in the treatment of Gb3Cer/CD77-expressing lymphoma. Here, we demonstrate in a Burkitt lymphoma cell model expressing this receptor, namely Raji cells, that Stx1, at quasi-non-toxic concentrations (0.05-0.1 pM), inhibits the repair of mafosfamide-induced DNA alkylating lesions, synergistically potentiating the cytotoxic activity of the anticancer drug. Conversely, human promyelocytic leukemia cells HL-60, which do not express Gb3Cer/CD77, were spared by the toxin as previously demonstrated for CD34+ human progenitor cells, and hence, in this cancer model, no additive nor synergistic effects were observed with the combined Stx1/mafosfamide treatment. Our findings suggest that Stx1 could be used to improve the mafosfamide-mediated purging of Gb3Cer/CD77+ tumor cells before autologous bone marrow transplantation. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
22. Molecular Damage and Induction of Proinflammatory Cytokines in Human Endothelial Cells Exposed to Shiga Toxin 1, Shiga Toxin 2, and -Sarcin
- Author
-
Brigotti, Maurizio, Carnicelli, Domenica, Ravanelli, Elisa, Vara, Antonio González, Martinelli, Chiara, Alfieri, Roberta R., Petronini, Pier Giorgio, and Sestili, Piero
- Abstract
Treatment of human endothelial cells with Shiga toxin 1 and 2 leads to the upregulation of genes encoding proinflammatory molecules involved in the pathogenesis of hemolytic-uremic syndrome. The paradoxical effect of inhibitors of mRNA translation, such as Shiga toxins, that at the same time induce protein expression was investigated by studying the relationship between their enzymatic activity (abstraction of adenine from nucleic acids) and the induction of interleukin-8 and granulocyte-macrophage colony-stimulating factor in human endothelial cells. As a positive control, the fungal toxin -sarcin, acting on the same rRNA sequence targeted by Shiga toxins with a different mechanism (RNase activity), was used. The three toxins caused ribosomal lesions that, in turn, induced the activation of p38 stress kinase with kinetics that paralleled the inhibition of translation. -Sarcin was devoid of activity on DNA. Shiga toxin 2 targeted nuclear DNA with more rapid kinetics than did Shiga toxin 1. Since the fungal ribotoxin was fully effective in the induction of proinflammatory proteins, we conclude that damage to ribosomes is indispensable and sufficient to activate protein expression via induction of the stress-kinase cascade. However, gene upregulation events induced by Shiga toxin 2 were much more efficient than those triggered by Shiga toxin 1, although the two toxins impaired translation to the same extent and had overlapping time courses of stress kinase activation. Regulations independent of the ribotoxic stress were assumed to operate in intoxicated cells. We hypothesized that the two bacterial toxins recognize different DNA sequences inducing different regulating effects on gene expression.
- Published
- 2007
23. Hypertonic Stress and Amino Acid Deprivation Both Increase Expression of mRNA for Amino Acid Transport System A.
- Author
-
Alfieri, Roberta R., Bonelli, Mara A., Petronini, Pier Giorgio, Desenzani, Silvia, Cavazzoni, Andrea, Borghetti, Angelo F., and Wheeler, Kenneth P.
- Subjects
- *
PHYSIOLOGY , *LETTERS to the editor - Abstract
Presents a letter to the editor about the efficiency of both hypertonic stress and amino acid deprivation in increasing the expression of mRNA for amino acid transport system A, published in the January 2005 issue of "The Journal of General Physiology."
- Published
- 2005
- Full Text
- View/download PDF
24. Gefitinib Inhibits Invasive Phenotype and Epithelial-Mesenchymal Transition in Drug-Resistant NSCLC Cells with MET Amplification.
- Author
-
La Monica, Silvia, Caffarra, Cristina, Saccani, Francesca, Galvani, Elena, Galetti, Maricla, Fumarola, Claudia, Bonelli, Mara, Cavazzoni, Andrea, Cretella, Daniele, Sirangelo, Rita, Gatti, Rita, Tiseo, Marcello, Ardizzoni, Andrea, Giovannetti, Elisa, Petronini, Pier Giorgio, and Alfieri, Roberta R.
- Subjects
GEFITINIB ,PHENOTYPES ,EPITHELIAL cells ,MESENCHYMAL stem cells ,AMPLIFICATION reactions ,EPIDERMAL growth factor receptors ,IMMUNE response ,THERAPEUTICS - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
25. Irreversible Inhibition of Epidermal Growth Factor Receptor Activity by 3-Aminopropanamides
- Author
-
Caterina Carmi, Silvia Rivara, Simonetta Russo, Fabrizio Bordi, Federica Vacondio, Gabriele Costantino, Andrea Cavazzoni, Andrea Ardizzoni, Roberta Alfieri, Elena Galvani, Marco Mor, Pier Giorgio Petronini, Stefania Aiello, Alessio Lodola, Carmi, C, Galvani, E, Vacondio, F, Rivara, S, Lodola, A, Russo, S, Aiello, S, Bordi, F, Costantino, G, Cavazzoni, A, Alfieri, RR, Ardizzoni, A, Petronini, PG, Mor, M, Carmi, Caterina, Galvani, Elena, Vacondio, Federica, Rivara, Silvia, Lodola, Alessio, Russo, Simonetta, Aiello, Stefania, Bordi, Fabrizio, Costantino, Gabriele, Cavazzoni, Andrea, Alfieri, Roberta R., Ardizzoni, Andrea, Petronini, Pier Giorgio, and Mor, Marco
- Subjects
Amide ,Cell Survival ,EGFR inhibitors ,Quinoline ,Antineoplastic Agents ,Antineoplastic Agent ,Structure-Activity Relationship ,T790M ,Gefitinib ,Cell Line, Tumor ,Drug Discovery ,Propionate ,medicine ,Humans ,Structure–activity relationship ,Epidermal growth factor receptor ,Phosphorylation ,Aniline Compounds ,biology ,Chemistry ,Drug Discovery3003 Pharmaceutical Science ,Autophosphorylation ,Quinazoline ,Aniline Compound ,Amides ,Settore CHIM/08 - Chimica Farmaceutica ,ErbB Receptors ,Biochemistry ,Protein kinase domain ,Drug Resistance, Neoplasm ,Quinazolines ,Quinolines ,biology.protein ,Molecular Medicine ,Receptor, Epidermal Growth Factor ,Propionates ,Drug Screening Assays, Antitumor ,Tyrosine kinase ,Human ,medicine.drug - Abstract
Irreversible epidermal growth factor receptor (EGFR) inhibitors contain a reactive warhead which covalently interacts with a conserved cysteine residue in the kinase domain. The acrylamide fragment, a commonly employed warhead, effectively alkylates Cys797 of EGFR, but its reactivity can cause rapid metabolic deactivation or nonspecific reactions with off-targets. We describe here a new series of irreversible inhibitors containing a 3-aminopropanamide linked in position 6 to 4-anilinoquinazoline or 4-anilinoquinoline-3- carbonitrile driving portions. Some of these compounds proved to be as efficient as their acrylamide analogues in inhibiting EGFR-TK (TK = tyrosine kinase) autophosphorylation in A549 lung cancer cells. Moreover, several 3-aminopropanamides suppressed proliferation of gefitinib-resistant H1975 cells, harboring the T790M mutation in EGFR, at significantly lower concentrations than did gefitinib. A prototypical compound, N-(4-(3-bromoanilino)quinazolin-6- yl)-3-(dimethylamino)propanamide (5), did not show covalent binding to cell-free EGFR-TK in a fluorescence assay, while it underwent selective activation in the intracellular environment, releasing an acrylamide derivative which can react with thiol groups. © 2012 American Chemical Society.
- Published
- 2012
- Full Text
- View/download PDF
26. 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
27. 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
28. 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
29. Gefitinib Inhibits Invasive Phenotype and Epithelial-Mesenchymal Transition in Drug-Resistant NSCLC Cells with MET Amplification
- Author
-
Silvia La Monica, Elena Galvani, Cristina Caffarra, Rita Sirangelo, Maricla Galetti, Daniele Cretella, Roberta Alfieri, Francesca Saccani, Marcello Tiseo, Pier Giorgio Petronini, Elisa Giovannetti, Andrea Cavazzoni, Claudia Fumarola, Rita Gatti, Mara A. Bonelli, Andrea Ardizzoni, La Monica, Silvia, Caffarra, Cristina, Saccani, Francesca, Galvani, Elena, Galetti, Maricla, Fumarola, Claudia, Bonelli, Mara, Cavazzoni, Andrea, Cretella, Daniele, Sirangelo, Rita, Gatti, Rita, Tiseo, Marcello, Ardizzoni, Andrea, Giovannetti, Elisa, Petronini, Pier Giorgio, Alfieri, Roberta R., Medical oncology laboratory, and CCA - Innovative therapy
- Subjects
Lung Neoplasms ,Epithelial-Mesenchymal Transition ,lcsh:Medicine ,Vimentin ,Antineoplastic Agents ,Drug resistance ,Antineoplastic Agent ,Gefitinib ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Epidermal growth factor receptor ,lcsh:Science ,Neoplasm Invasivene ,Multidisciplinary ,Biochemistry, Genetics and Molecular Biology (all) ,biology ,Cell growth ,Medicine (all) ,lcsh:R ,Gene Amplification ,Quinazoline ,Cell migration ,Proto-Oncogene Proteins c-met ,Molecular biology ,respiratory tract diseases ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Lung Neoplasm ,Agricultural and Biological Sciences (all) ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Quinazolines ,lcsh:Q ,Receptor, Epidermal Growth Factor ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug ,Research Article ,Human - 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. © 2013 La Monica et al.
- Published
- 2013
- Full Text
- View/download PDF
30. Effects of sorafenib on energy metabolism in breast cancer cells: Role of AMPK-mTORC1 signaling
- Author
-
Daniele Generali, Andrea Cavazzoni, Maricla Galetti, Pier Giorgio Petronini, Claudia Fumarola, Roberta Alfieri, Cristina Caffarra, Elena Galvani, Silvia La Monica, Mara A. Bonelli, Fumarola, Claudia, Caffarra, Cristina, La Monica, Silvia, Galetti, Maricla, Alfieri, Roberta R., Cavazzoni, Andrea, Galvani, Elena, Generali, Daniele, Petronini, Pier Giorgio, and Bonelli, Mara A.
- Subjects
Niacinamide ,MAPK/ERK pathway ,Sorafenib ,AMPK ,medicine.medical_specialty ,Cancer Research ,Glucose uptake ,Down-Regulation ,Antineoplastic Agents ,Breast Neoplasms ,mTORC1 ,AMP-Activated Protein Kinases ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Oxidative Phosphorylation ,Inhibitory Concentration 50 ,Adenosine Triphosphate ,Breast cancer ,Cell Line, Tumor ,Internal medicine ,medicine ,Humans ,Anilides ,RNA, Small Interfering ,Protein Kinase Inhibitors ,Protein kinase B ,Glucose Transporter Type 1 ,Phenylurea Compounds ,TOR Serine-Threonine Kinases ,Glucose transporter ,Energy metabolism ,Mitochondria ,Neoplasm Proteins ,Glucose ,Endocrinology ,Oncology ,Anaerobic glycolysis ,Multiprotein Complexes ,Cancer research ,Female ,RNA Interference ,Glycolysis ,Cell Division ,Signal Transduction ,medicine.drug - Abstract
In this study, we investigated the effects and the underlying molecular mechanisms of the multi-kinase inhibitor sorafenib in a panel of breast cancer cell lines. Sorafenib inhibited cell proliferation and induced apoptosis through the mitochondrial pathway. These effects were neither correlated with modulation of MAPK and AKT pathways nor dependent on the ERα status. Sorafenib promoted an early perturbation of mitochondrial function, inducing a deep depolarization of mitochondrial membrane, associated with drop of intracellular ATP levels and increase of ROS generation. As a response to this stress condition, the energy sensor AMPK was rapidly activated in all the cell lines analyzed. In MCF-7 and SKBR3 cells, AMPK enhanced glucose uptake by up-regulating the expression of GLUT-1 glucose transporter, as also demonstrated by AMPKα1 RNA interference, and stimulated aerobic glycolysis thus increasing lactate production. Moreover, the GLUT-1 inhibitor fasentin blocked sorafenib-induced glucose uptake and potentiated its cytotoxic activity in SKBR3 cells. Persistent activation of AMPK by sorafenib finally led to the impairment of glucose metabolism both in MCF-7 and SKBR3 cells as well as in the highly glycolytic MDA-MB-231 cells, resulting in cell death. This previously unrecognized long-term effect of sorafenib was mediated by AMPK-dependent inhibition of the mTORC1 pathway. Suppression of mTORC1 activity was sufficient for sorafenib to hinder glucose utilization in breast cancer cells, as demonstrated by the observation that the mTORC1 inhibitor rapamycin induced a comparable down-regulation of GLUT-1 expression and glucose uptake. The key role of AMPK-dependent inhibition of mTORC1 in sorafenib mechanisms of action was confirmed by AMPKα1 silencing, which restored mTORC1 activity conferring a significant protection from cell death. This study provides insights into the molecular mechanisms driving sorafenib anti-tumoral activity in breast cancer, and supports the need for going on with clinical trials aimed at proving the efficacy of sorafenib for breast cancer treatment.
- Published
- 2013
31. Overcoming acquired resistance to letrozole by targeting the PI3K/AKT/mTOR pathway in breast cancer cell clones
- Author
-
Pier Giorgio Petronini, Daniele Andreis, Maricla Galetti, Adrian L. Harris, Daniele Generali, Stefano Tramonti, Elena Galvani, Andrea Cavazzoni, Lesley-Ann Martin, Claudia Fumarola, Alberto Bottini, Silvia La Monica, Roberta Alfieri, Kinda Airoud, Mara A. Bonelli, Ramona Bertoni, Cavazzoni, Andrea, Bonelli, Mara A., Fumarola, Claudia, La Monica, Silvia, Airoud, Kinda, Bertoni, Ramona, Alfieri, Roberta R., Galetti, Maricla, Tramonti, Stefano, Galvani, Elena, Harris, Adrian L., Martin, Lesley Ann, Andreis, Daniele, Bottini, Alberto, Generali, Daniele, and Petronini, Pier Giorgio
- Subjects
Cancer Research ,Resistance ,Drug Resistance ,Quinoline ,Estrogen receptor ,Antineoplastic Agent ,Phosphatidylinositol 3-Kinases ,Immunosuppressive Agent ,Sirolimu ,Breast ,Tumor ,TOR Serine-Threonine Kinase ,Blotting ,Letrozole ,TOR Serine-Threonine Kinases ,MTOR ,Imidazoles ,Neoadjuvant Therapy ,Everolimu ,Oncology ,Quinolines ,Female ,Western ,Nitrile ,Immunosuppressive Agents ,Breast Neoplasm ,medicine.drug ,Human ,Signal Transduction ,medicine.medical_specialty ,Blotting, Western ,AKT ,Aged ,Antineoplastic Agents ,Breast Neoplasms ,Cell Line, Tumor ,Drug Resistance, Neoplasm ,Everolimus ,Humans ,Nitriles ,Proto-Oncogene Proteins c-akt ,Sirolimus ,Triazoles ,Cell Line ,Breast cancer ,Internal medicine ,medicine ,Protein kinase B ,Imidazole ,PI3K/AKT/mTOR pathway ,business.industry ,RPTOR ,medicine.disease ,Endocrinology ,Cancer research ,Neoplasm ,Triazole ,Phosphatidylinositol 3-Kinase ,business - Abstract
Development of resistance to endocrine therapy is a clinical issue in estrogen receptor (ER)-positive breast cancer. Here we show that persistent activation of AKT/mTOR signaling is crucial to the acquisition of letrozole resistance in cell clones generated from MCF-7/AROM-1 aromatase-expressing breast cancer cells after prolonged letrozole exposure. ERα plays a marginal role in this context. As a proof of concept, the association between PI3K/AKT/mTOR signaling and insensitivity to endocrine therapies was confirmed in breast cancer patients who developed early letrozole resistance in neoadjuvant setting. In addition our results suggest that, regardless of the mechanism mediating the activation of AKT/mTOR pathway, either RAD001 or NVP-BEZ235 treatment may represent a promising strategy to overcome acquired resistance to letrozole in breast cancers dependent on AKT/mTOR signaling. © 2012 Elsevier Ireland Ltd.
- Published
- 2012
32. Combined use of anti-ErbB monoclonal antibodies and erlotinib enhances antibody-dependent cellular cytotoxicity of wild-type erlotinib-sensitive NSCLC cell lines
- Author
-
Maricla Galetti, Claudia Fumarola, Mara Bonelli, Elena Galvani, Andrea Cavazzoni, Francesca Saccani, Antonio Mutti, Elisabetta Barocelli, Federico Quaini, Denise Madeddu, Gallia Graiani, Andrea Ardizzoni, Paolo Carbognani, Marcello Tiseo, Pier Giorgio Petronini, Daniele Cretella, Roberta Alfieri, Paola Mozzoni, Luca Ampollini, Silvia La Monica, Cavazzoni, Andrea, Alfieri, Roberta R, Cretella, Daniele, Saccani, Francesca, Ampollini, Luca, Galetti, Maricla, Quaini, Federico, Graiani, Gallia, Madeddu, Denise, Mozzoni, Paola, Galvani, Elena, La Monica, Silvia, Bonelli, Mara, Fumarola, Claudia, Mutti, Antonio, Carbognani, Paolo, Tiseo, Marcello, Barocelli, Elisabetta, Petronini, Pier Giorgio, and Ardizzoni, Andrea
- Subjects
Cancer Research ,Lung Neoplasms ,Receptor, ErbB-2 ,Cetuximab ,Mice ,Trastuzumab ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,Epidermal growth factor receptor ,Antibody-dependent cell-mediated cytotoxicity ,Mice, Inbred BALB C ,biology ,Protein Stability ,Drug Synergism ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Erlotinib ,Oncology ,Molecular Medicine ,Female ,Protein stabilization ,Lung cancer ,ADCC ,medicine.drug ,Human ,Xenograft Model Antitumor Assay ,Cell Survival ,EGFR ,Mice, Nude ,Antibodies, Monoclonal, Humanized ,lcsh:RC254-282 ,Erlotinib Hydrochloride ,Gefitinib ,ErbB ,Cell Line, Tumor ,medicine ,Animals ,Humans ,neoplasms ,Analysis of Variance ,Binding Sites ,Antineoplastic Combined Chemotherapy Protocol ,Animal ,Research ,Cell Membrane ,Antibody-Dependent Cell Cytotoxicity ,Binding Site ,Quinazoline ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,Lung Neoplasm ,Cancer research ,biology.protein ,Quinazolines ,Receptor, Epidermal Growth Factor - Abstract
Background The epidermal growth factor receptor (EGFR) is an established target for anti-cancer treatment in different tumour types. Two different strategies have been explored to inhibit this pivotal molecule in epithelial cancer development: small molecules TKIs and monoclonal antibodies. ErbB/HER-targeting by monoclonal antibodies such as cetuximab and trastuzumab or tyrosine-kinase inhibitors as gefitinib or erlotinib has been proven effective in the treatment of advanced NSCLC. Results In this study we explored the potential of combining either erlotinib with cetuximab or trastuzumab to improve the efficacy of EGFR targeted therapy in EGFR wild-type NSCLC cell lines. Erlotinib treatment was observed to increase EGFR and/or HER2 expression at the plasma membrane level only in NSCLC cell lines sensitive to the drug inducing protein stabilization. The combined treatment had marginal effect on cell proliferation but markedly increased antibody-dependent, NK mediated, cytotoxicity in vitro. Moreover, in the Calu-3 xenograft model, the combination significantly inhibited tumour growth when compared with erlotinib and cetuximab alone. Conclusion Our results indicate that erlotinib increases surface expression of EGFR and/or HER2 only in EGFR-TKI sensitive NSCLC cell lines and, in turns, leads to increased susceptibility to ADCC both in vitro and in a xenograft models. The combination of erlotinib with monoclonal antibodies represents a potential strategy to improve the treatment of wild-type EGFR NSCLC patients sensitive to erlotinib.
- Published
- 2012
33. Synergistic activity of letrozole and sorafenib on breast cancer cells
- Author
-
Mitch Dowsett, Claudia Fumarola, Silvana Belletti, Daniele Generali, Maricla Galetti, Rita Gatti, Lesley-Ann Martin, Mara Bonelli, Pier Giorgio Petronini, Roberta Alfieri, Adrian L. Harris, Silvia La Monica, Andrea Cavazzoni, Dean B. Evans, Alberto Bottini, Stephen B. Fox, Department of Experimental Medicine, University of Parma = Università degli studi di Parma [Parme, Italie], Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital [Oxford University Hospital], Peter MacCallum Cancer Centre, Peter MacCallum Cancer Center, Novartis Institutes of BioMedical Research, Oncology Research, Academic Department of Biochemistry, Royal Marsden Hospital, Breakthrough Breast Cancer Centre, London Institute of Cancer, Unità di Patologia Mammaria-Breast Cancer Unit, Centro di Medicina Molecolare, Istituti Ospitalieri di Cremona, Bonelli, Mara A., Fumarola, Claudia, Alfieri, Roberta R., La Monica, Silvia, Cavazzoni, Andrea, Galetti, Maricla, Gatti, Rita, Belletti, Silvana, Harris, Adrian L., Fox, Stephen B., Evans, Dean B., Dowsett, Mitch, Martin, Lesley Ann, Bottini, Alberto, Generali, Daniele, and Petronini, Pier Giorgio
- Subjects
Breast cancer ,Letrozole ,mTORC1 ,Sorafenib ,Oncology ,Cancer Research ,MAPK/ERK pathway ,Time Factors ,Pyridines ,Poly (ADP-Ribose) Polymerase-1 ,Apoptosis ,Cell Cycle Proteins ,Retinoblastoma Protein ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Cyclin D1 ,Testosterone ,Phosphorylation ,Caspase 7 ,0303 health sciences ,Estradiol ,Aromatase Inhibitors ,TOR Serine-Threonine Kinases ,Benzenesulfonates ,Cell Cycle ,Apoptosis Inducing Factor ,Cytochromes c ,Ribosomal Protein S6 Kinases, 70-kDa ,Drug Synergism ,Cell cycle ,Caspase 9 ,3. Good health ,030220 oncology & carcinogenesis ,Female ,Poly(ADP-ribose) Polymerases ,medicine.drug ,Niacinamide ,medicine.drug_class ,Breast Neoplasms ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Transfection ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Aromatase ,Cell Line, Tumor ,Nitriles ,medicine ,Humans ,Protein kinase B ,Protein Kinase Inhibitors ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Aromatase inhibitor ,Dose-Response Relationship, Drug ,Cell growth ,Phenylurea Compounds ,Proteins ,Triazoles ,Antiestrogen ,Phosphoproteins ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,Cancer research - Abstract
International audience; Estrogens induce breast tumor cell proliferation by directly regulating gene expression via the estrogen receptor (ER) transcriptional activity and by affecting growth factor signaling pathways such as mitogen-activated protein kinase (MAPK) and AKT/mammalian target of rapamycin Complex1 (mTORC1) cascades. In this study we demonstrated the preclinical therapeutic efficacy of combining the aromatase inhibitor letrozole with the multi-kinase inhibitor sorafenib in aromatase-expressing breast cancer cell lines. Treatment with letrozole reduced testosterone-driven cell proliferation, by inhibiting the synthesis of estrogens. Sorafenib inhibited cell proliferation in a concentration-dependent manner; this effect was not dependent on sorafenib-mediated inhibition of Raf1, but involved the down-regulation of mTORC1 and its targets p70S6K and 4E-binding protein 1 (4E-BP1). At concentrations of 5–10 μM the growth-inhibitory effect of sorafenib was associated with the induction of apoptosis, as indicated by release of cytochrome and Apoptosis-Inducing Factor into the cytosol, activation of caspase-9 and caspase-7, and PARP-1 cleavage. Combination of letrozole and sorafenib produced a synergistic inhibition of cell proliferation associated with an enhanced accumulation of cells in the G/G phase of the cell cycle and with a down-regulation of the cell cycle regulatory proteins c-myc, cyclin D1, and phospho-Rb. In addition, longer experiments (12 weeks) demonstrated that sorafenib may be effective in preventing the acquisition of resistance towards letrozole. Together, these results indicate that combination of letrozole and sorafenib might constitute a promising approach to the treatment of hormone-dependent breast cancer.
- Published
- 2009
- Full Text
- View/download PDF
34. Trastuzumab emtansine is active on HER-2 overexpressing NSCLC cell lines and overcomes gefitinib resistance
- Author
-
Maricla Galetti, Silvia La Monica, Roberta Alfieri, Costanza Lagrasta, Caterina Frati, Luca Ampollini, Daniele Cretella, Francesca Saccani, Pier Giorgio Petronini, Marcello Tiseo, Federico Quaini, Andrea Ardizzoni, Andrea Cavazzoni, Claudia Fumarola, Cristina Caffarra, Mara A. Bonelli, Cretella, Daniele, Saccani, Francesca, Quaini, Federico, Frati, Caterina, Lagrasta, Costanza, Bonelli, Mara, Caffarra, Cristina, Cavazzoni, Andrea, Fumarola, Claudia, Galetti, Maricla, La Monica, Silvia, Ampollini, Luca, Tiseo, Marcello, Ardizzoni, Andrea, Petronini, Pier G., and Alfieri, Roberta R
- Subjects
Oncology ,Cancer Research ,Immunoconjugates ,Lung Neoplasms ,Receptor, ErbB-2 ,T-DM1 ,Gene Expression ,Microtubule polymerization ,Antineoplastic Agent ,chemistry.chemical_compound ,Mice ,Trastuzumab ,Carcinoma, Non-Small-Cell Lung ,Epidermal growth factor receptor ,biology ,Medicine (all) ,Gefitinib ,Cell cycle ,Tumor Burden ,Molecular Medicine ,Female ,Lung cancer ,Tyrosine kinase ,medicine.drug ,Human ,musculoskeletal diseases ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Xenograft Model Antitumor Assay ,Mice, Nude ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Maytansine ,Animal ,Research ,Quinazoline ,medicine.disease ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,Lung Neoplasm ,chemistry ,Immunoconjugate ,Trastuzumab emtansine ,HER-2 ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Quinazolines - Abstract
Background: HER-2 represents a relatively new therapeutic target for non small cell lung cancer (NSCLC) patients. The incidence for reported HER-2 overexpression/amplification/mutations ranges from 2 to 20% in NSCLC. Moreover, HER-2 amplification is a potential mechanism of resistance to tyrosine kinase inhibitors of the epidermal growth factor receptor (EGFR-TKI) (about 10% of cases). T-DM1, trastuzumab emtansine is an antibody-drug conjugate composed by the monoclonal antibody trastuzumab and the microtubule polymerization inhibitor DM1. The activity of T-DM1 has been studied in breast cancer but the role of T-DM1 in lung cancer remains unexplored.Methods: Antiproliferative and proapoptotic effects of T-DM1 have been investigated in different NSCLC cell lines by MTT, crystal violet staining, morphological study and Western blotting. HER-2 expression and cell cycle were evaluated by flow cytometry and Western blotting. Antibody dependent cell cytotoxicity (ADCC) was measured with a CytoTox assay. Xenografted mice model has been generated using a NSCLC cell line to evaluate the effect of T-DM1 on tumor growth. Moreover, a morphometric and immunohistochemical analysis of tumor xenografts was conducted.Results: In this study we investigated the effect of T-DM1 in a panel of NSCLC cell lines with different HER-2 expression levels, in H1781 cell line carrying HER-2 mutation and in gefitinib resistant HER-2 overexpressing PC9/HER2cl1 cell clone. T-DM1 efficiently inhibited proliferation with arrest in G2-M phase and induced cell death by apoptosis in cells with a significant level of surface expression of HER-2. Antibody-dependent cytotoxicity assay documented that T-DM1 maintained the same activity of trastuzumab. Our data also suggest that targeting HER-2 with T-DM1 potentially overcomes gefitinib resistance. In addition a correlation between cell density/tumor size with both HER-2 expression and T-DM1 activity was established in vitro and in an in vivo xenograft model.Conclusions: Our results indicate that targeting HER-2 with T-DM1 may offer a new therapeutic approach in HER-2 over-expressing lung cancers including those resistant to EGFR TKIs. © 2014 Cretella et al.; licensee BioMed Central Ltd.
- Published
- 2014
35. Molecular damage and induction of proinflammatory cytokines in human endothelial cells exposed to Shiga toxin 1, Shiga toxin 2, and alpha-sarcin.
- Author
-
Brigotti M, Carnicelli D, Ravanelli E, Vara AG, Martinelli C, Alfieri RR, Petronini PG, and Sestili P
- Subjects
- Caspase 3 genetics, Caspase 3 metabolism, Endothelium, Vascular cytology, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Humans, Interleukin-8 genetics, Interleukin-8 metabolism, Umbilical Veins, Up-Regulation, p38 Mitogen-Activated Protein Kinases genetics, p38 Mitogen-Activated Protein Kinases metabolism, DNA Damage drug effects, Endoribonucleases pharmacology, Endothelial Cells drug effects, Fungal Proteins pharmacology, Protein Biosynthesis drug effects, Shiga Toxin 1 pharmacology, Shiga Toxin 2 pharmacology
- Abstract
Treatment of human endothelial cells with Shiga toxin 1 and 2 leads to the upregulation of genes encoding proinflammatory molecules involved in the pathogenesis of hemolytic-uremic syndrome. The paradoxical effect of inhibitors of mRNA translation, such as Shiga toxins, that at the same time induce protein expression was investigated by studying the relationship between their enzymatic activity (abstraction of adenine from nucleic acids) and the induction of interleukin-8 and granulocyte-macrophage colony-stimulating factor in human endothelial cells. As a positive control, the fungal toxin alpha-sarcin, acting on the same rRNA sequence targeted by Shiga toxins with a different mechanism (RNase activity), was used. The three toxins caused ribosomal lesions that, in turn, induced the activation of p38 stress kinase with kinetics that paralleled the inhibition of translation. Alpha-sarcin was devoid of activity on DNA. Shiga toxin 2 targeted nuclear DNA with more rapid kinetics than did Shiga toxin 1. Since the fungal ribotoxin was fully effective in the induction of proinflammatory proteins, we conclude that damage to ribosomes is indispensable and sufficient to activate protein expression via induction of the stress-kinase cascade. However, gene upregulation events induced by Shiga toxin 2 were much more efficient than those triggered by Shiga toxin 1, although the two toxins impaired translation to the same extent and had overlapping time courses of stress kinase activation. Regulations independent of the ribotoxic stress were assumed to operate in intoxicated cells. We hypothesized that the two bacterial toxins recognize different DNA sequences inducing different regulating effects on gene expression.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.