37 results on '"Valentina D’Amato"'
Search Results
2. Supplementary Figure 5 from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 5320K, Validation of antibodies for analysis of SphK1 expression on FFPE colorectal cancer tissues
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 3 from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 503K, Effects of SphK1 modulation on cetuximab sensitivity in resistant colorectal cancer cell lines
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 4 from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 1047K, Effects of the combination cetuximab plus fingolimod on apoptosis of GEO-CR tumor xenografts
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 6 from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 2068K, Correlation of SphK1 expression with clinical data in colorectal cancer patients
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Methods from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 39K
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure Legend from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 32K
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 2 from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 483K, Effects of SphK1 inhibition by DMS on cetuximab sensitivity in resistant colorectal cancer cell lines
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 1 from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 357K, Sensitivity to cetuximab of a panel of human colorectal cancer cell lines
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
Purpose: Although the anti–EGF receptor (EGFR) monoclonal antibody cetuximab is an effective strategy in colorectal cancer therapy, its clinical use is limited by intrinsic or acquired resistance. Alterations in the “sphingolipid rheostat”—the balance between the proapoptotic molecule ceramide and the mitogenic factor sphingosine-1-phosphate (S1P)—due to sphingosine kinase 1 (SphK1) overactivation have been involved in resistance to anticancer-targeted agents. Moreover, cross-talks between SphK1 and EGFR-dependent signaling pathways have been described.Experimental design: We investigated SphK1 contribution to cetuximab resistance in colorectal cancer, in preclinical in vitro/in vivo models, and in tumor specimens from patients.Results: SphK1 was found overexpressed and overactivated in colorectal cancer cells with intrinsic or acquired resistance to cetuximab. SphK1 contribution to resistance was supported by the demonstration that SphK1 inhibition by N,N-dimethyl-sphingosine or silencing via siRNA in resistant cells restores sensitivity to cetuximab, whereas exogenous SphK1 overexpression in sensitive cells confers resistance to these agents. Moreover, treatment of resistant cells with fingolimod (FTY720), a S1P receptor (S1PR) antagonist, resulted in resensitization to cetuximab both in vitro and in vivo, with inhibition of tumor growth, interference with signal transduction, induction of cancer cells apoptosis, and prolongation of mice survival. Finally, a correlation between SphK1 expression and cetuximab response was found in colorectal cancer patients. Clin Cancer Res; 19(1); 138–47. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Table 2 from Sphingosine Kinase 1 Overexpression Contributes to Cetuximab Resistance in Human Colorectal Cancer Models
- Author
-
Roberto Bianco, Sabino De Placido, Giancarlo Troncone, Bianca Maria Veneziani, Chiara Carlomagno, Alfonso De Stefano, Adelaide Greco, Antonella Zannetti, Silvana Del Vecchio, Gabriella Marfè, Vincenzo Damiano, Valentina D'Amato, Claudia D'Amato, Lucia Nappi, Luigi Formisano, Umberto Malapelle, Roberta Marciano, and Roberta Rosa
- Abstract
PDF file - 908K, Multivariate analysis correlating the response to cetuximab-based regimens to different factors
- Published
- 2023
- Full Text
- View/download PDF
12. Post-intervention Status in Patients With Refractory Myasthenia Gravis Treated With Eculizumab During REGAIN and Its Open-Label Extension
- Author
-
Mantegazza, R., Wolfe, G. I., Muppidi, S., Wiendl, H., Fujita, K. P., O'Brien, F. L., Booth, H. D. E., Howard, J. F., Claudio Gabriel Mazia, Miguel, Wilken, Fabio, Barroso, Juliet, Saba, Marcelo, Rugiero, Mariela, Bettini, Marcelo, Chaves, Gonzalo, Vidal, Alejandra Dalila Garcia, Jan De Bleecker, Guy Van den Abeele, Kathy de Koning, Katrien De Mey, Rudy, Mercelis, Délphine, Mahieu, Linda, Wagemaekers, Philip Van Damme, Annelies, Depreitere, Caroline, Schotte, Charlotte, Smetcoren, Olivier, Stevens, Sien Van Daele, Nicolas, Vandenbussche, Annelies, Vanhee, Sarah, Verjans, Jan, Vynckier, Ann, D'Hont, Petra, Tilkin, Alzira Alves de Siqueira Carvalho, Igor Dias Brockhausen, David, Feder, Daniel, Ambrosio, Gabor Lovasamela César, Ana Paula Melo, Renata Martins Ribeiro, Rosana, Rocha, Bruno Bezerra Rosa, Thabata, Veiga, Luiz Augusto da Silva, Murilo Santos Engel, Jordana Gonçalves Geraldo, Maria da Penha Ananias Morita, Erica Nogueira Coelho, Gabriel, Paiva, Marina, Pozo, Natalia, Prando, Debora Dada Martineli Torres, Cristiani Fernanda Butinhao, Gustavo, Duran, Tomás Augusto Suriane Fialho, Tamires Cristina Gomes da Silva, Luiz Otavio Maia Gonçalves, Lucas Eduardo Pazetto, Luciana Renata Cubas Volpe, Luciana Souza Duca, Maurício AndréGheller Friedrich, Alexandre, Guerreiro, Henrique, Mohr, Maurer Pereira Martins, Daiane da Cruz Pacheco, Luciana, Ferreira, Ana Paula Macagnan, Graziela, Pinto, Aline de Cassia Santos, Acary Souza Bulle Oliveira, Ana Carolina Amaral de Andrade, Marcelo, Annes, Liene Duarte Silva, Valeria Cavalcante Lino, Wladimir, Pinto, Natália, Assis, Fernanda, Carrara, Carolina, Miranda, Iandra, Souza, Patrícia, Fernandes, Zaeem, Siddiqi, Cecile, Phan, Jeffrey, Narayan, Derrick, Blackmore, Ashley, Mallon, Rikki, Roderus, Elizabeth, Watt, Stanislav, Vohanka, Josef, Bednarik, Magda, Chmelikova, Marek, Cierny, Stanislava, Toncrova, Jana, Junkerova, Barbora, Kurkova, Katarina, Reguliova, Olga, Zapletalova, Jiri, Pitha, Iveta, Novakova, Michaela, Tyblova, Ivana, Jurajdova, Marcela, Wolfova, Henning, Andersen, Thomas, Harbo, Lotte, Vinge, Susanne, Krogh, Anita, Mogensen, John, Vissing, Joan, Højgaard, Nanna, Witting, Anne Mette Ostergaard Autzen, Jane, Pedersen, Juha-Pekka, Erälinna, Mikko, Laaksonen, Olli, Oksaranta, Tuula, Harrison, Jaana, Eriksson, Csilla, Rozsa, Melinda, Horvath, Gabor, Lovas, Judit, Matolcsi, Gedeonne, Jakab, Gyorgyi, Szabo, Brigitta, Szabadosne, Laszlo, Vecsei, Livia, Dezsi, Edina, Varga, Monika, Konyane, Antonini, Giovanni, Antonella Di Pasquale, Garibaldi, Matteo, Morino, Stefania, Troili, Fernanda, Fionda, Laura, Amelia, Evoli, Paolo Emilio Alboini, Valentina, D'Amato, Raffaele, Iorio, Inghilleri, Maurizio, Frasca, Vittorio, Elena, Giacomelli, Gori, MARIA CRISTINA, Diego, Lopergolo, Onesti, Emanuela, Maria, Gabriele, Francesco, Saccà, Alessandro, Filla, Teresa, Costabile, Enrico, Marano, Angiola, Fasanaro, Angela, Marsili, Giorgia, Puorro, Carlo, Antozzi, Silvia, Bonanno, Giorgia, Camera, Alberta, Locatelli, Lorenzo, Maggi, Maria, Pasanisi, Angela, Campanella, Akiyuki, Uzawa, Tetsuya, Kanai, Naoki, Kawaguchi, Masahiro, Mori, Yoko, Kaneko, Akiko, Kanzaki, Eri, Kobayashi, Hiroyuki, Murai, Katsuhisa, Masaki, Dai, Matsuse, Takuya, Matsushita, Taira, Uehara, Misa, Shimpo, Maki, Jingu, Keiko, Kikutake, Yumiko, Nakamura, Yoshiko, Sano, Kimiaki, Utsugisawa, Yuriko, Nagane, Ikuko, Kamegamori, Tomoko, Tsuda, Yuko, Fujii, Kazumi, Futono, Yukiko, Ozawa, Aya, Mizugami, Yuka, Saito, Makoto, Samukawa, Hidekazu, Suzuki, Miyuki, Morikawa, Sachiko, Kamakura, Eriko, Miyawaki, Meinoshin, Okumura, Soichiro, Funaka, Tomohiro, Kawamura, Masayuki, Nakamori, Masanori, Takahashi, Namie, Taichi, Tomoya, Hasuike, Eriko, Higuchi, Hisako, Kobayashi, Kaori, Osakada, Hirokazu, Shiraishi, Teiichiro, Miyazaki, Masakatsu, Motomura, Akihiro, Mukaino, Shunsuke, Yoshimura, Shizuka, Asada, Seiko, Yoshida, Shoko, Amamoto, Tomomi, Kobashikawa, Megumi, Koga, Maeda, Yasuko, Kazumi, Takada, Mihoko, Takada, Masako, Tsurumaru, Yumi, Yamashita, Yasushi, Suzuki, Tetsuya, Akiyama, Koichi, Narikawa, Ohito, Tano, Kenichi, Tsukita, Rikako, Kurihara, Fumie, Meguro, Yusuke, Fukuda, Miwako, Sato, Tomihiro, Imai, Emiko, Tsuda, Shun, Shimohama, Takashi, Hayashi, Shin, Hisahara, Jun, Kawamata, Takashi, Murahara, Masaki, Saitoh, Shuichiro, Suzuki, Daisuke, Yamamoto, Yoko, Ishiyama, Naoko, Ishiyama, Mayuko, Noshiro, Rumi, Takeyama, Kaori, Uwasa, Ikuko, Yasuda, Anneke van der Kooi, Marianne de Visser, Tamar, Gibson, Byung-Jo, Kim, Chang Nyoung Lee, Yong Seo Koo, Hung Youl Seok, Hoo Nam Kang, Hyejin, Ra, Byoung Joon Kim, Eun Bin Cho, Misong, Choi, Hyelim, Lee, Ju-Hong, Min, Jinmyoung, Seok, Jieun, Lee, Da Yoon Koh, Juyoung, Kwon, Sangae, Park, Eun Haw Choi, Yoon-Ho, Hong, So-Hyun, Ahn, Dae Lim Koo, Jae-Sung, Lim, Chae Won Shin, Ji Ye Hwang, Miri, Kim, Seung Min Kim, Ha-Neul, Jeong, Jinwoo, Jung, Yool-Hee, Kim, Hyung Seok Lee, Ha Young Shin, Eun Bi Hwang, Miju, Shin, Carlos, Casasnovas, Maria Antonia Alberti Aguilo, Christian, Homedes-Pedret, Natalia Julia Palacios, Laura Diez Porras, Valentina Velez Santamaria, Ana, Lazaro, Josep Gamez Carbonell, Pilar, Sune, Maria Salvado Figueras, Gisela, Gili, Gonzalo, Mazuela, Isabel, Illa, Elena Cortes Vicente, Jordi, Diaz-Manera, Luis Antonio Querol Gutiérrez, Ricardo Rojas Garcia, Nuria, Vidal, Elisabet, Arribas-Ibar, Exuperio Diez Tejedor, Pilar Gomez Salcedo, Mireya, Fernandez-Fournier, Pedro Lopez Ruiz, Francisco Javier Rodriguez de Rivera, Maria, Sastre, Fredrik, Piehl, Albert, Hietala, Lena, Bjarbo, Ihsan, Sengun, Arzu, Meherremova, Pinar, Ozcelik, Bengu, Balkan, Celal, Tuga, Muzeyyen, Ugur, Sevim, Erdem-Ozdamar, Can Ebru Bekircan-Kurt, Nazire Pinar Acar, Ezgi, Yilmaz, Yagmur, Caliskan, Gulsah, Orsel, Husnu, Efendi, Seda, Aydinlik, Hakan, Cavus, Ayse, Kutlu, Gulsah, Becerikli, Cansu, Semiz, Ozlem, Tun, Murat, Terzi, Baki, Dogan, Musa Kazim Onar, Sedat, Sen, Tugce Kirbas Cavdar, Adife, Veske, Fiona, Norwood, Aikaterini, Dimitriou, Jakit, Gollogly, Mohamed, Mahdi-Rogers, Arshira, Seddigh, Giannis, Sokratous, Gal, Maier, Faisal, Sohail, Saiju, Jacob, Girija, Sadalage, Pravin, Torane, Claire, Brown, Amna, Shah, Sivakumar, Sathasivam, Heike, Arndt, Debbie, Davies, Dave, Watling, Anthony, Amato, Thomas, Cochrane, Mohammed, Salajegheh, Kristen, Roe, Katherine, Amato, Shirli, Toska, Nicholas, Silvestri, Kara, Patrick, Karen, Zakalik, Jonathan, Katz, Robert, Miller, Marguerite, Engel, Dallas, Forshew, Elena, Bravver, Benjamin, Brooks, Mohammed, Sanjak, Sarah, Plevka, Maryanne, Burdette, Scott, Cunningham, Megan, Kramer, Joanne, Nemeth, Clara, Schommer, Tierney, Scott, Vern, Juel, Jeffrey, Guptill, Lisa, Hobson-Webb, Janice, Massey, Kate, Beck, Donna, Carnes, John, Loor, Amanda, Anderson, Robert, Pascuzzi, Cynthia, Bodkin, John, Kincaid, Riley, Snook, Sandra, Guingrich, Angela, Micheels, Vinay, Chaudhry, Andrea, Corse, Betsy, Mosmiller, Andrea, Kelley, Doreen, Ho, Jayashri, Srinivasan, Michal, Vytopil, Jordan, Jara, Nicholas, Ventura, Cynthia, Carter, Craig, Donahue, Carol, Herbert, Stephanie, Scala, Elaine, Weiner, Sharmeen, Alam, Jonathan, Mckinnon, Laura, Haar, Naya, Mckinnon, Karan, Alcon, Kaitlyn, Mckenna, Nadia, Sattar, Kevin, Daniels, Dennis, Jeffery, Miriam, Freimer, Joseph Chad Hoyle, John, Kissel, Julie, Agriesti, Sharon, Chelnick, Louisa, Mezache, Colleen, Pineda, Filiz, Muharrem, Chafic, Karam, Julie, Khoury, Tessa, Marburger, Harpreet, Kaur, Diana, Dimitrova, James, Gilchrist, Brajesh, Agrawal, Mona, Elsayed, Stephanie, Kohlrus, Angela, Ardoin, Taylor, Darnell, Laura, Golden, Barbara, Lokaitis, Jenna, Seelbach, Neelam, Goyal, Sarada, Sakamuri, Yuen, T So, Shirley, Paulose, Sabrina, Pol, Lesly, Welsh, Ratna, Bhavaraju-Sanka, Alejandro Tobon Gonzalez, Lorraine, Dishman, Floyd, Jones, Anna, Gonzalez, Patricia, Padilla, Amy, Saklad, Marcela, Silva, Sharon, Nations, Jaya, Trivedi, Steve, Hopkins, Mohamed, Kazamel, Mohammad, Alsharabati, Liang, Lu, Kenkichi, Nozaki, Sandi, Mumfrey-Thomas, Amy, Woodall, Tahseen, Mozaffar, Tiyonnoh, Cash, Namita, Goyal, Gulmohor, Roy, Veena, Mathew, Fatima, Maqsood, Brian, Minton, H James Jones, Jeffrey, Rosenfeld, Rebekah, Garcia, Laura, Echevarria, Sonia, Garcia, Michael, Pulley, Shachie, Aranke, Alan Ross Berger, Jaimin, Shah, Yasmeen, Shabbir, Lisa, Smith, Mary, Varghese, Laurie, Gutmann, Ludwig, Gutmann, Nivedita, Jerath, Christopher, Nance, Andrea, Swenson, Heena, Olalde, Nicole, Kressin, Jeri, Sieren, Richard, Barohn, Mazen, Dimachkie, Melanie, Glenn, April, Mcvey, Mamatha, Pasnoor, Jeffery, Statland, Yunxia, Wang, Tina, Liu, Kelley, Emmons, Nicole, Jenci, Jerry, Locheke, Alex, Fondaw, Kathryn, Johns, Gabrielle, Rico, Maureen, Walsh, Laura, Herbelin, Charlene, Hafer-Macko, Justin, Kwan, Lindsay, Zilliox, Karen, Callison, Valerie, Young, Beth, Disanzo, Kerry, Naunton, Michael, Benatar, Martin, Bilsker, Khema, Sharma, Anne, Cooley, Eliana, Reyes, Sara-Claude, Michon, Danielle, Sheldon, Julie, Steele, Rebecca, Traub, Manisha, Chopra, Tuan, Vu, Lara, Katzin, Terry, Mcclain, Brittany, Harvey, Adam, Hart, Kristin, Huynh, Said, Beydoun, Amaiak, Chilingaryan, Victor, Doan, Brian, Droker, Hui, Gong, Sanaz, Karimi, Frank, Lin, Krishna, Polaka, Akshay, Shah, Anh, Tran, Salma, Akhter, Ali, Malekniazi, Rup, Tandan, Michael, Hehir, Waqar, Waheed, Shannon, Lucy, Michael, Weiss, Jane, Distad, Susan, Strom, Sharon, Downing, Bryan, Kim, Tulio, Bertorini, Thomas, Arnold, Kendrick, Henderson, Rekha, Pillai, Liu, Ye, Lauren, Wheeler, Jasmine, Hewlett, Mollie, Vanderhook, Richard, Nowak, Daniel, Dicapua, Benison, Keung, Aditya, Kumar, Huned, Patwa, Kimberly, Robeson, Irene, Yang, Joan, Nye, and Hong, Vu
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Clinical Neurology ,Antibodies, Monoclonal, Humanized ,Placebo ,Article ,Antibodies ,Post-intervention ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,Refractory ,law ,Internal medicine ,Monoclonal ,Myasthenia Gravis ,Medicine and Health Sciences ,Humans ,Medicine ,Humanized ,Science & Technology ,business.industry ,Middle Aged ,Eculizumab ,Complement Inactivating Agents ,Female ,Treatment Outcome ,EFFICACY ,medicine.disease ,Myasthenia gravis ,Clinical trial ,030104 developmental biology ,SAFETY ,Neurosciences & Neurology ,Neurology (clinical) ,business ,Life Sciences & Biomedicine ,COMPLEMENT INHIBITOR ECULIZUMAB ,030217 neurology & neurosurgery ,medicine.drug - Abstract
ObjectiveTo evaluate whether eculizumab helps patients with anti–acetylcholine receptor–positive (AChR+) refractory generalized myasthenia gravis (gMG) achieve the Myasthenia Gravis Foundation of America (MGFA) post-intervention status of minimal manifestations (MM), we assessed patients' status throughout REGAIN (Safety and Efficacy of Eculizumab in AChR+ Refractory Generalized Myasthenia Gravis) and its open-label extension.MethodsPatients who completed the REGAIN randomized controlled trial and continued into the open-label extension were included in this tertiary endpoint analysis. Patients were assessed for the MGFA post-intervention status of improved, unchanged, worse, MM, and pharmacologic remission at defined time points during REGAIN and through week 130 of the open-label study.ResultsA total of 117 patients completed REGAIN and continued into the open-label study (eculizumab/eculizumab: 56; placebo/eculizumab: 61). At week 26 of REGAIN, more eculizumab-treated patients than placebo-treated patients achieved a status of improved (60.7% vs 41.7%) or MM (25.0% vs 13.3%; common OR: 2.3; 95% CI: 1.1–4.5). After 130 weeks of eculizumab treatment, 88.0% of patients achieved improved status and 57.3% of patients achieved MM status. The safety profile of eculizumab was consistent with its known profile and no new safety signals were detected.ConclusionEculizumab led to rapid and sustained achievement of MM in patients with AChR+ refractory gMG. These findings support the use of eculizumab in this previously difficult-to-treat patient population.ClinicalTrials.gov IdentifierREGAIN, NCT01997229; REGAIN open-label extension, NCT02301624.Classification of EvidenceThis study provides Class II evidence that, after 26 weeks of eculizumab treatment, 25.0% of adults with AChR+ refractory gMG achieved MM, compared with 13.3% who received placebo.
- Published
- 2020
- Full Text
- View/download PDF
13. Long-term efficacy and safety of eculizumab in Japanese patients with generalized myasthenia gravis: A subgroup analysis of the REGAIN open-label extension study
- Author
-
Hiroyuki Murai, Akiyuki Uzawa, Yasushi Suzuki, Tomihiro Imai, Hirokazu Shiraishi, Hidekazu Suzuki, Meinoshin Okumura, Fanny O’Brien, Jing-Jing Wang, Kenji P. Fujita, Kimiaki Utsugisawa, Claudio Gabriel Mazia, Miguel Wilken, Fabio Barroso, Juliet Saba, Marcelo Rugiero, Mariela Bettini, Marcelo Chaves, Gonzalo Vidal, Alejandra Dalila Garcia, Jan De Bleecker, Guy Van den Abeele, Kathy de Koning, Katrien De Mey, Rudy Mercelis, Délphine Mahieu, Linda Wagemaekers, Philip Van Damme, Annelies Depreitere, Caroline Schotte, Charlotte Smetcoren, Olivier Stevens, Sien Van Daele, Nicolas Vandenbussche, Annelies Vanhee, Sarah Verjans, Jan Vynckier, Ann D'Hondt, Petra Tilkin, Alzira Alves de Siqueira Carvalho, Igor Dias Brockhausen, David Feder, Daniel Ambrosio, Pamela César, Ana Paula Melo, Renata Martins Ribeiro, Rosana Rocha, Bruno Bezerra Rosa, Thabata Veiga, Luiz Augusto da Silva, Murilo Santos Engel, Jordana Gonçalves Geraldo, Maria da Penha Ananias Morita, Erica Nogueira Coelho, Gabriel Paiva, Marina Pozo, Natalia Prando, Debora Dada Martineli Torres, Cristiani Fernanda Butinhao, Gustavo Duran, Tamires Cristina Gomes da Silva, Luiz Otavio Maia Gonçalves, Lucas Eduardo Pazetto, Tomás Augusto Suriane Fialho, Luciana Renata Cubas Volpe, Luciana Souza Duca, Acary Souza Bulle Oliveira, Ana Carolina Amaral Andrade, Marcelo Annes, Liene Duarte Silva, Valeria Cavalcante Lino, Wladimir Pinto, Natália Assis, Fernanda Carrara, Carolina Miranda, Iandra Souza, Patricia Fernandes, Zaeem Siddiqi, Cecile Phan, Jeffrey Narayan, Derrick Blackmore, Ashley Mallon, Rikki Roderus, Elizabeth Watt, Jana Junkerova, Barbora Kurkova, Katarina Reguliova, Olga Zapletalova, Jiri Pitha, Iveta Novakova, Michaela Tyblova, Ivana Jurajdova, Marcela Wolfova, Henning Andersen, Thomas Harbo, Lotte Vinge, Susanne Krogh, Anita Mogensen, John Vissing, Joan Højgaard, Nanna Witting, Anne Mette Ostergaard Autzen, Jane Pedersen, Juha-Pekka Eralinna, Mikko Laaksonen, Olli Oksaranta, Tuula Harrison, Jaana Eriksson, Csilla Rozsa, Melinda Horvath, Gabor Lovas, Judit Matolcsi, Gyorgyi Szabo, Gedeonne Jakab, Brigitta Szabadosne, Giovanni Antonini, Antonella Di Pasquale, Matteo Garibaldi, Stefania Morino, Fernanda Troili, Laura Fionda, Allessandro Filla, Teresa Costabile, Enrico Marano, Francesco Saccà, Angiola Fasanaro, Angela Marsili, Giorgia Puorro, Renato Mantegazza, Carlo Antozzi, Silvia Bonanno, Giorgia Camera, Alberta Locatelli, Lorenzo Maggi, Maria Pasanisi, Angela Campanella, Amelia Evoli, Paolo Emilio Alboini, Valentina D'Amato, Raffaele Iorio, Tetsuya Kanai, Naoki Kawaguchi, Masahiro Mori, Yoko Kaneko, Akiko Kanzaki, Eri Kobayashi, Katsuhisa Masaki, Dai Matsuse, Takuya Matsushita, Taira Uehara, Misa Shimpo, Maki Jingu, Keiko Kikutake, Yumiko Nakamura, Yoshiko Sano, Yuriko Nagane, Ikuko Kamegamori, Tomoko Tsuda, Yuko Fujii, Kazumi Futono, Yukiko Ozawa, Aya Mizugami, Yuka Saito, Miyuki Morikawa, Makoto Samukawa, Sachiko Kamakura, Eriko Miyawaki, Teiichiro Mitazaki, Masakatsu Motomura, Akihiro Mukaino, Shunsuke Yoshimura, Shizuka Asada, Seiko Yoshida, Shoko Amamoto, Tomomi Kobashikawa, Megumi Koga, Yasuko Maeda, Kazumi Takada, Mihoko Takada, Masako Tsurumaru, Yumi Yamashita, Tetsuya Akiyama, Koichi Narikawa, Ohito Tano, Kenichi Tsukita, Rikako Kurihara, Fumie Meguro, Yusuke Fukuda, Miwako Sato, Soichiro Funaka, Tomohiro Kawamura, Masayuki Makamori, Masanori Takahashi, Namie Taichi, Tomoya Hasuike, Eriko Higuchi, Hisako Kobayashi, Kaori Osakada, Emiko Tsuda, Shun Shimohama, Takashi Hayashi, Shin Hisahara, Jun Kawamata, Takashi Murahara, Masaki Saitoh, Shuichiro Suzuki, Daisuke Yamamoto, Yoko Ishiyama, Naoko Ishiyama, Mayuko Noshiro, Rumi Takeyama, Kaori Uwasa, Ikuko Yasuda, Anneke van der Kooi, Marianne de Visser, Tamar Gibson, Carlos Casasnovas, Maria Antonia Alberti Aguilo, Christian Homedes-Pedret, Natalia Julia Palacios, Laura Diez Porras, Valentina Velez Santamaria, Ana Lazaro, Exuperio Diez Tejedor, Pilar Gomez Salcedo, Mireya Fernandez-Fournier, Pedro Lopez Ruiz, Francisco Javier Rodriguez de Rivera, Maria Sastre, Josep Gamez, Pilar Sune, Maria Salvado, Gisela Gili, Gonzalo Mazuela, Isabel Illa, Elena Cortes Vicente, Jordi Diaz-Manera, Luis Antonio Querol Gutierrez, Ricardo Rojas Garcia, Nuria Vidal, Elisabet Arribas-Ibar, Fredrik Piehl, Albert Hietala, Lena Bjarbo, Ihsan Sengun, Arzu Meherremova, Pinar Ozcelik, Bengu Balkan, Celal Tuga, Muzeyyen Ugur, Sevim Erdem-Ozdamar, Can Ebru Bekircan-Kurt, Nazire Pinar Acar, Ezgi Yilmaz, Yagmur Caliskan, Gulsah Orsel, Husnu Efendi, Seda Aydinlik, Hakan Cavus, Ayse Kutlu, Gulsah Becerikli, Cansu Semiz, Ozlem Tun, Murat Terzi, Baki Dogan, Musa Kazim Onar, Sedat Sen, Tugce Kirbas Cavdar, Adife Veske, Fiona Norwood, Aikaterini Dimitriou, Jakit Gollogly, Mohamed Mahdi-Rogers, Arshira Seddigh, Giannis Sokratous, Gal Maier, Faisal Sohail, Saiju Jacob, Girija Sadalage, Pravin Torane, Claire Brown, Amna Shah, Sivakumar Sathasivam, Heike Arndt, Debbie Davies, Dave Watling, Anthony Amato, Thomas Cochrane, Mohammed Salajegheh, Kristen Roe, Katherine Amato, Shirli Toska, Gil Wolfe, Nicholas Silvestri, Kara Patrick, Karen Zakalik, Jonathan Katz, Robert Miller, Marguerite Engel, Dallas Forshew, Elena Bravver, Benjamin Brooks, Sarah Plevka, Maryanne Burdette, Scott Cunningham, Mohammad Sanjak, Megan Kramer, Joanne Nemeth, Clara Schommer, Scott Tierney, Vern Juel, Jeffrey Guptill, Lisa Hobson-Webb, Janice Massey, Kate Beck, Donna Carnes, John Loor, Amanda Anderson, Robert Pascuzzi, Cynthia Bodkin, John Kincaid, Riley Snook, Sandra Guingrich, Angela Micheels, Vinay Chaudhry, Andrea Corse, Betsy Mosmiller, Andrea Kelley, Doreen Ho, Jayashri Srinivasan, Michal Vytopil, Jordan Jara, Nicholas Ventura, Stephanie Scala, Cynthia Carter, Craig Donahue, Carol Herbert, Elaine Weiner, Sharmeen Alam, Jonathan McKinnon, Laura Haar, Naya McKinnon, Karan Alcon, Kaitlyn McKenna, Nadia Sattar, Kevin Daniels, Dennis Jeffery, Miriam Freimer, Joseph Chad Hoyle, John Kissel, Julie Agriesti, Sharon Chelnick, Louisa Mezache, Colleen Pineda, Filiz Muharrem, Chafic Karam, Julie Khoury, Tessa Marburger, Harpreet Kaur, Diana Dimitrova, James Gilchrist, Brajesh Agrawal, Mona Elsayed, Stephanie Kohlrus, Angela Andoin, Taylor Darnell, Laura Golden, Barbara Lokaitis, Jenna Seelback, Srikanth Muppidi, Neelam Goyal, Sarada Sakamuri, Yuen T. So, Shirley Paulose, Sabrina Pol, Lesly Welsh, Ratna Bhavaraju-Sanka, Alejandro Tobon Gonzales, Lorraine Dishman, Floyd Jones, Anna Gonzalez, Patricia Padilla, Amy Saklad, Marcela Silva, Mohamed Kazamel, Mohammad Alsharabati, Liang Lu, Kenkichi Nozaki, Sandi Mumfrey-Thomas, Amy Woodall, Tahseen Mozaffar, Tiyonnoh Cash, Namita Goyal, Gulmohor Roy, Veena Mathew, Fatima Maqsood, Brian Minton, H. James Jones, Jeffrey Rosenfeld, Rebekah Garcia, Laura Echevarria, Sonia Garcia, Michael Pulley, Shachie Aranke, Alan Ross Berger, Jaimin Shah, Yasmeen Shabbir, Lisa Smith, Mary Varghese, Laurie Gutmann, Ludwig Gutmann, Nivedita Jerath, Christopher Nance, Andrea Swenson, Heena Olalde, Nicole Kressin, Jeri Sieren, Richard Barohn, Mazen Dimachkie, Melanie Glenn, April McVey, Mamatha Pasnoor, Jeffery Statland, Yunxia Wang, Tina Liu, Kelley Emmons, Nicole Jenci, Jerry Locheke, Alex Fondaw, Kathryn Johns, Gabrielle Rico, Maureen Walsh, Laura Herbelin, Charlene Hafer-Macko, Justin Kwan, Lindsay Zilliox, Karen Callison, Valerie Young, Beth DiSanzo, Kerry Naunton, Michael Benatar, Martin Bilsker, Khema Sharma, Anne Cooley, Eliana Reyes, Sara-Claude Michon, Danielle Sheldon, Julie Steele, James Howard Jr, Manisha Chopra, Rebecca Traub, Tuan Vu, Lara Katzin, Terry McClain, Brittany Harvey, Adam Hart, Kristin Huynh, Said Beydoun, Amaiak Chilingaryan, Victor Doan, Brian Droker, Hui Gong, Sanaz Karimi, Frank Lin, Krishna Pokala, Akshay Shah, Anh Tran, Salma Akhter, Ali Malekniazi, Rup Tandan, Michael Hehir, Waqar Waheed, Shannon Lucy, Michael Weiss, Jane Distad, Susan Strom, Sharon Downing, Bryan Kim, Richard Nowak, Daniel Dicapua, Benison Keung, Aditya Kumar, Huned Patwa, Kimberly Robeson, Irene Yang, Joan Nye, Hong Vu, H., Murai, A., Uzawa, Y., Suzuki, T., Imai, H., Shiraishi, H., Suzuki, M., Okumura, F., O'Brien, J. -J., Wang, K. P., Fujita, K., Utsugisawa, Gabriel Mazia 11, Claudio, Wilken 11, Miguel, Barroso 11, Fabio, Saba 11, Juliet, Rugiero 12, Marcelo, Bettini 12, Mariela, Chaves 12, Marcelo, Vidal 12, Gonzalo, Dalila Garcia 12, Alejandra, De Bleecker 13, Jan, Van den Abeele 13, Guy, de Koning 13, Kathy, De Mey 13, Katrien, Mercelis 14, Rudy, Mahieu 14, Délphine, Wagemaekers 14, Linda, Van Damme 15, Philip, Depreitere 16, Annelie, Schotte 16, Caroline, Smetcoren 16, Charlotte, Stevens 16, Olivier, Van Daele 16, Sien, Vandenbussche 16, Nicola, Vanhee 16, Annelie, Verjans 16, Sarah, Vynckier 16, Jan, D'Hondt 16, Ann, Tilkin 16, Petra, Alves de Siqueira Carvalho 17, Alzira, Dias Brockhausen 17, Igor, Feder 17, David, Ambrosio 17, Daniel, César 17, Pamela, Paula Melo 17, Ana, Martins Ribeiro 17, Renata, Rocha 17, Rosana, Bezerra Rosa 17, Bruno, Veiga 17, Thabata, Augusto da Silva 17, Luiz, Santos Engel 17, Murilo, Gonçalves Geraldo 17, Jordana, da Penha Ananias Morita 18, Maria, Nogueira Coelho 18, Erica, Paiva 18, Gabriel, Pozo 18, Marina, Prando 18, Natalia, Dada Martineli Torres 18, Debora, Fernanda Butinhao 18, Cristiani, Duran 18, Gustavo, Cristina Gomes da Silva 18, Tamire, Otavio Maia Gonçalves 18, Luiz, Eduardo Pazetto 18, Luca, Augusto Suriane Fialho 18, Tomá, Renata Cubas Volpe 18, Luciana, Souza Duca 18, Luciana, Souza Bulle Oliveira 19, Acary, Carolina Amaral Andrade 19, Ana, Annes 19, Marcelo, Duarte Silva 19, Liene, Cavalcante Lino 19, Valeria, Pinto 19, Wladimir, Assis 19, Natália, Carrara 19, Fernanda, Miranda 19, Carolina, Souza 19, Iandra, Fernandes 19, Patricia, Siddiqi 20, Zaeem, Phan 20, Cecile, Narayan 20, Jeffrey, Blackmore 20, Derrick, Mallon 20, Ashley, Roderus 20, Rikki, Watt 20, Elizabeth, Junkerova 21, Jana, Kurkova 21, Barbora, Reguliova 21, Katarina, Zapletalova 21, Olga, Pitha 22, Jiri, Novakova 22, Iveta, Tyblova 22, Michaela, Jurajdova 22, Ivana, Wolfova 22, Marcela, Andersen 23, Henning, Harbo 23, Thoma, Vinge 23, Lotte, Krogh 23, Susanne, Mogensen 23, Anita, Vissing 24, John, Højgaard 24, Joan, Witting 24, Nanna, Mette Ostergaard Autzen 24, Anne, Pedersen 24, Jane, Eralinna 25, Juha-Pekka, Laaksonen 25, Mikko, Oksaranta 25, Olli, Harrison 25, Tuula, Eriksson 25, Jaana, Rozsa 26, Csilla, Horvath 26, Melinda, Lovas 26, Gabor, Matolcsi 26, Judit, Szabo 26, Gyorgyi, Jakab 26, Gedeonne, Szabadosne 26, Brigitta, Antonini 27, Giovanni, Di Pasquale 27, Antonella, Garibaldi 27, Matteo, Morino 27, Stefania, Troili 27, Fernanda, Fionda 27, Laura, Filla, Allessandro, Costabile, Teresa, Marano, Enrico, Sacca', Francesco, Fasanaro, Angiola, Marsili, Angela, Puorro, Giorgia, Mantegazza 29, Renato, Antozzi 29, Carlo, Bonanno 29, Silvia, Camera 29, Giorgia, Locatelli 29, Alberta, Maggi 29, Lorenzo, Pasanisi 29, Maria, Campanella 29, Angela, Evoli 30, Amelia, Emilio Alboini 30, Paolo, D'Amato 30, Valentina, Iorio 30, Raffaele, Kanai 31, Tetsuya, Kawaguchi 31, Naoki, Mori 31, Masahiro, Kaneko 31, Yoko, Kanzaki 31, Akiko, Kobayashi 31, Eri, Masaki 32, Katsuhisa, Matsuse 32, Dai, Matsushita 32, Takuya, Uehara 32, Taira, Shimpo 32, Misa, Jingu 32, Maki, Kikutake 32, Keiko, Nakamura 32, Yumiko, Sano 32, Yoshiko, Nagane 33, Yuriko, Kamegamori 33, Ikuko, Tsuda 33, Tomoko, Fujii 33, Yuko, Futono 33, Kazumi, Ozawa 33, Yukiko, Mizugami 33, Aya, Saito 33, Yuka, Morikawa 34, Miyuki, Samukawa 34, Makoto, Kamakura 34, Sachiko, Miyawaki 34, Eriko, Mitazaki 35, Teiichiro, Motomura 35, Masakatsu, Mukaino 35, Akihiro, Yoshimura 35, Shunsuke, Asada 35, Shizuka, Yoshida 35, Seiko, Amamoto 35, Shoko, Kobashikawa 35, Tomomi, Koga 35, Megumi, Maeda 35, Yasuko, Takada 35, Kazumi, Takada 35, Mihoko, Tsurumaru 35, Masako, Yamashita 35, Yumi, Akiyama 36, Tetsuya, Narikawa 36, Koichi, Tano 36, Ohito, Tsukita 36, Kenichi, Kurihara 36, Rikako, Meguro 36, Fumie, Fukuda 36, Yusuke, Sato 36, Miwako, Funaka 37, Soichiro, Kawamura 37, Tomohiro, Makamori 37, Masayuki, Takahashi 37, Masanori, Taichi 37, Namie, Hasuike 37, Tomoya, Higuchi 37, Eriko, Kobayashi 37, Hisako, Osakada 37, Kaori, Tsuda 38, Emiko, Shimohama 38, Shun, Hayashi 38, Takashi, Hisahara 38, Shin, Kawamata 38, Jun, Murahara 38, Takashi, Saitoh 38, Masaki, Suzuki 38, Shuichiro, Yamamoto 38, Daisuke, Ishiyama 38, Yoko, Ishiyama 38, Naoko, Noshiro 38, Mayuko, Takeyama 38, Rumi, Uwasa 38, Kaori, Yasuda 38, Ikuko, van der Kooi 39, Anneke, de Visser 39, Marianne, Gibson 39, Tamar, Casasnovas 40, Carlo, Antonia Alberti Aguilo 40, Maria, Homedes-Pedret 40, Christian, Julia Palacios 40, Natalia, Diez Porras 40, Laura, Velez Santamaria 40, Valentina, Lazaro 40, Ana, Diez Tejedor 41, Exuperio, Gomez Salcedo 41, Pilar, Fernandez-Fournier 41, Mireya, Lopez Ruiz 41, Pedro, Javier Rodriguez de Rivera 41, Francisco, Sastre 41, Maria, Gamez 42, Josep, Sune 42, Pilar, Salvado 42, Maria, Gili 42, Gisela, Mazuela 42, Gonzalo, Illa 43, Isabel, Cortes Vicente 43, Elena, Diaz-Manera 43, Jordi, Antonio Querol Gutierrez 43, Lui, Rojas Garcia 43, Ricardo, Vidal 43, Nuria, Arribas-Ibar 43, Elisabet, Piehl 44, Fredrik, Hietala 44, Albert, Bjarbo 44, Lena, Sengun 45, Ihsan, Meherremova 45, Arzu, Ozcelik 45, Pinar, Balkan 45, Bengu, Tuga 45, Celal, Ugur 45, Muzeyyen, Erdem-Ozdamar 46, Sevim, Ebru Bekircan-Kurt 46, Can, Pinar Acar 46, Nazire, Yilmaz 46, Ezgi, Caliskan 46, Yagmur, Orsel 46, Gulsah, Efendi 47, Husnu, Aydinlik 47, Seda, Cavus 47, Hakan, Kutlu 47, Ayse, Becerikli 47, Gulsah, Semiz 47, Cansu, Tun 47, Ozlem, Terzi 48, Murat, Dogan 48, Baki, Kazim Onar 48, Musa, Sen 48, Sedat, Kirbas Cavdar 48, Tugce, Veske 48, Adife, Norwood 49, Fiona, Dimitriou 49, Aikaterini, Gollogly 49, Jakit, Mahdi-Rogers 49, Mohamed, Seddigh 49, Arshira, Sokratous 49, Gianni, Maier 49, Gal, Sohail 49, Faisal, Jacob 50, Saiju, Sadalage 50, Girija, Torane 50, Pravin, Brown 50, Claire, Shah 50, Amna, Sathasivam 51, Sivakumar, Arndt 51, Heike, Davies 51, Debbie, Watling 51, Dave, Amato 52, Anthony, Cochrane 52, Thoma, Salajegheh 52, Mohammed, Roe 52, Kristen, Amato 52, Katherine, Toska 52, Shirli, Wolfe 53, Gil, Silvestri 53, Nichola, Patrick 53, Kara, Zakalik 53, Karen, Katz 54, Jonathan, Miller 54, Robert, Engel 54, Marguerite, Forshew 54, Dalla, Bravver 55, Elena, Brooks 55, Benjamin, Plevka 55, Sarah, Burdette 55, Maryanne, Cunningham 55, Scott, Sanjak 55, Mohammad, Kramer 55, Megan, Nemeth 55, Joanne, Schommer 55, Clara, Tierney 55, Scott, Juel 56, Vern, Guptill 56, Jeffrey, Hobson-Webb 56, Lisa, Massey 56, Janice, Beck 56, Kate, Carnes 56, Donna, Loor 56, John, Anderson 56, Amanda, Pascuzzi 57, Robert, Bodkin 57, Cynthia, Kincaid 57, John, Snook 57, Riley, Guingrich 57, Sandra, Micheels 57, Angela, Chaudhry 58, Vinay, Corse 58, Andrea, Mosmiller 58, Betsy, Kelley 58, Andrea, Ho 59, Doreen, Srinivasan 59, Jayashri, Vytopil 59, Michal, Jara 59, Jordan, Ventura 59, Nichola, Scala 59, Stephanie, Carter 59, Cynthia, Donahue 59, Craig, Herbert 59, Carol, Weiner 59, Elaine, Alam 59, Sharmeen, McKinnon 60, Jonathan, Haar 60, Laura, McKinnon 60, Naya, Alcon 60, Karan, McKenna 60, Kaitlyn, Sattar 60, Nadia, Daniels 60, Kevin, Jeffery 60, Denni, Freimer 61, Miriam, Chad Hoyle 61, Joseph, Kissel 61, John, Agriesti 61, Julie, Chelnick 61, Sharon, Mezache 61, Louisa, Pineda 61, Colleen, Muharrem 61, Filiz, Karam 62, Chafic, Khoury 62, Julie, Marburger 62, Tessa, Kaur 62, Harpreet, Dimitrova 62, Diana, Gilchrist 63, Jame, Agrawal 63, Brajesh, Elsayed 63, Mona, Kohlrus 63, Stephanie, Andoin 63, Angela, Darnell 63, Taylor, Golden 63, Laura, Lokaitis 63, Barbara, Seelback 63, Jenna, Muppidi 64, Srikanth, Goyal 64, Neelam, Sakamuri 64, Sarada, T So 64, Yuen, Paulose 64, Shirley, Pol 64, Sabrina, Welsh 64, Lesly, Bhavaraju-Sanka 65, Ratna, Tobon Gonzales 65, Alejandro, Dishman 65, Lorraine, Jones 65, Floyd, Gonzalez 65, Anna, Padilla 65, Patricia, Saklad 65, Amy, Silva 65, Marcela, Kazamel 66, Mohamed, Alsharabati 66, Mohammad, Lu 66, Liang, Nozaki 66, Kenkichi, Mumfrey-Thomas 66, Sandi, Woodall 66, Amy, Mozaffar 67, Tahseen, Cash 67, Tiyonnoh, Goyal 67, Namita, Roy 67, Gulmohor, Mathew 67, Veena, Maqsood 67, Fatima, Minton 67, Brian, James Jones 68, H, Rosenfeld 68, Jeffrey, Garcia 68, Rebekah, Echevarria 68, Laura, Garcia 68, Sonia, Pulley 69, Michael, Aranke 69, Shachie, Ross Berger 69, Alan, Shah 69, Jaimin, Shabbir 69, Yasmeen, Smith 69, Lisa, Varghese 69, Mary, Gutmann 70, Laurie, Gutmann 70, Ludwig, Jerath 70, Nivedita, Nance 70, Christopher, Swenson 70, Andrea, Olalde 70, Heena, Kressin 70, Nicole, Sieren 70, Jeri, Barohn 71, Richard, Dimachkie 71, Mazen, Glenn 71, Melanie, McVey 71, April, Pasnoor 71, Mamatha, Statland 71, Jeffery, Wang 71, Yunxia, Liu 71, Tina, Emmons 71, Kelley, Jenci 71, Nicole, Locheke 71, Jerry, Fondaw 71, Alex, Johns 71, Kathryn, Rico 71, Gabrielle, Walsh 71, Maureen, Herbelin 71, Laura, Hafer-Macko 72, Charlene, Kwan 72, Justin, Zilliox 72, Lindsay, Callison 72, Karen, Young 72, Valerie, DiSanzo 72, Beth, Naunton 72, Kerry, Benatar 73, Michael, Bilsker 73, Martin, Sharma 73, Khema, Cooley 73, Anne, Reyes 73, Eliana, Michon 73, Sara-Claude, Sheldon 73, Danielle, Steele 73, Julie, Howard Jr 74, Jame, Karam 74, Chafic, Chopra 74, Manisha, Traub 74, Rebecca, Vu 75, Tuan, Katzin 75, Lara, McClain 75, Terry, Harvey 75, Brittany, Hart 75, Adam, Huynh 75, Kristin, Beydoun 76, Said, Chilingaryan 76, Amaiak, Doan 76, Victor, Droker 76, Brian, Gong 76, Hui, Karimi 76, Sanaz, Lin 76, Frank, McClain 76, Terry, Pokala 76, Krishna, Shah 76, Akshay, Tran 76, Anh, Akhter 76, Salma, Malekniazi 76, Ali, Tandan 77, Rup, Hehir 77, Michael, Waheed 77, Waqar, Lucy 77, Shannon, Weiss 78, Michael, Distad 78, Jane, Strom 78, Susan, Downing 78, Sharon, Kim 78, Bryan, Nowak 79, Richard, Dicapua 79, Daniel, Keung 79, Benison, Kumar 79, Aditya, Patwa 79, Huned, Robeson 79, Kimberly, Yang 79, Irene, Nye 79, Joan, and Hong Vu
- Subjects
Male ,Pediatrics ,Myasthenia gravi ,Open-label extension study ,Complement inhibitor ,0302 clinical medicine ,Quality of life ,Japan ,Medicine and Health Sciences ,Medicine ,030212 general & internal medicine ,Myasthenia gravis ,education.field_of_study ,MG-ADL ,Japanese patient ,Eculizumab ,Middle Aged ,HLA ,MG-QoL15 ,Treatment Outcome ,Neurology ,Female ,Life Sciences & Biomedicine ,COMPLEMENT INHIBITOR ECULIZUMAB ,medicine.drug ,Adult ,medicine.medical_specialty ,Population ,Clinical Neurology ,Subgroup analysis ,Placebo ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,Refractory ,Asian People ,Myasthenia Gravis ,Humans ,education ,Aged ,Science & Technology ,business.industry ,Neurosciences ,medicine.disease ,Complement Inactivating Agents ,Japanese patients ,ONSET ,Neurology (clinical) ,Neurosciences & Neurology ,business ,030217 neurology & neurosurgery - Abstract
The terminal complement inhibitor eculizumab was shown to improve myasthenia gravis-related symptoms in the 26-week, phase 3, randomized, double-blind, placebo-controlled REGAIN study (NCT01997229). In this 52-week sub-analysis of the open-label extension of REGAIN (NCT02301624), eculizumab's efficacy and safety were assessed in 11 Japanese and 88 Caucasian patients with anti-acetylcholine receptor antibody-positive refractory generalized myasthenia gravis. For patients who had received placebo during REGAIN, treatment with open-label eculizumab resulted in generally similar outcomes in the Japanese and Caucasian populations. Rapid improvements were maintained for 52 weeks, assessed by change in score from open-label extension baseline to week 52 (mean [standard error]) using the following scales (in Japanese and Caucasian patients, respectively): Myasthenia Gravis Activities of Daily Living (-2.4 [1.34] and - 3.3 [0.65]); Quantitative Myasthenia Gravis (-2.9 [1.98] and - 4.3 [0.79]); Myasthenia Gravis Composite (-4.5 [2.63] and - 4.9 [1.19]); and Myasthenia Gravis Quality of Life 15-item questionnaire (-8.6 [5.68] and - 6.5 [1.93]). Overall, the safety of eculizumab was consistent with its known safety profile. In this interim sub-analysis, the efficacy and safety of eculizumab in Japanese and Caucasian patients were generally similar, and consistent with the overall REGAIN population. ispartof: JOURNAL OF THE NEUROLOGICAL SCIENCES vol:407 ispartof: location:Netherlands status: published
- Published
- 2019
14. Approaches for targeting cancer stem cells drug resistance
- Author
-
Valentina D’Amato, Sabino De Placido, Roberto Bianco, Roberta De Rosa, Rosa, Roberta, D'Amato, Valentina, DE PLACIDO, Sabino, and Bianco, Roberto
- Subjects
0301 basic medicine ,Drug ,media_common.quotation_subject ,medicine.medical_treatment ,Population ,Antineoplastic Agents ,Drug resistance ,Pharmacology ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer stem cell ,Neoplasms ,Cancer stem cells, resistance, stemness, EMT, immune evasion ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,education ,media_common ,education.field_of_study ,Cancer ,medicine.disease ,Radiation therapy ,030104 developmental biology ,Drug Resistance, Neoplasm ,Research Design ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Efflux - Abstract
Introduction: Several reports have suggested that a population of undifferentiated cells known as cancer stem cells (CSCs), is responsible for cancer formation and maintenance. In the last decade, the presence of CSCs in solid cancers have been reported. Areas covered: This review summarizes the main approaches for targeting CSCs drug resistance. It is indeed known that CSCs may contribute to resistance to conventional chemotherapy, radiotherapy and targeted agents. Among the mechanisms by which CSCs escape anticancer therapies, removal of therapeutic agents by drug efflux pumps, enhanced DNA damage repair, activation of mitogenic/anti-apoptotic pathways; the main features of CSCs, stemness and EMT, are involved, as well as the capability to evade immune response. Expert opinion: Different approaches are suitable to target CSCs mediated drug resistance. Some of them are currently under clinical evaluation in different cancer types. A better understanding of CSC biology, as well as more accurate study design, may maximize the therapeutic effects of these agents. In this respect, it is important to establish: (i) which molecules should be targeted; (ii) what drug combinations may be suitable; (iii) which patient settings will CSC targeting offer the highest clinical benefit; and (iv) how to integrate therapeutic approaches targeting CSCs with standard cancer therapy.
- Published
- 2016
- Full Text
- View/download PDF
15. Mechanisms of resistance to mTOR inhibitors
- Author
-
Cataldo Bianco, Roberto Bianco, Luigi Formisano, Fabiana Napolitano, Roberta De Rosa, Alberto Servetto, Roberta Marciano, Valentina D’Amato, Pietro De Placido, Formisano, L., Napolitano, F., Rosa, R., D'Amato, V., Servetto, A., Marciano, R., De Placido, P., Bianco, C., and Bianco, R.
- Subjects
Adult ,0301 basic medicine ,mTOR inhibitor ,Population ,Antineoplastic Agents ,Breast Neoplasms ,Lymphoma, Mantle-Cell ,Neuroendocrine tumors ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Humans ,Medicine ,education ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,education.field_of_study ,business.industry ,Kinase ,Cell growth ,Mechanisms of resistance ,TOR Serine-Threonine Kinases ,Hematology ,medicine.disease ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Mantle cell lymphoma ,Phosphatidylinositol 3-Kinase ,business ,Proto-Oncogene Proteins c-akt ,Predictive factors - Abstract
In several tumors the PI3K/AKT/mTOR pathway is frequently disrupted, an event that results in uncontrolled cell proliferation and tumor growth. Through the years, several compounds have been developed to inhibit the pathway at different steps: the mammalian target of rapamycin (mTOR) seemed to be the most qualified target. However, this kinase has such a key role in cell survival that mechanisms of resistance are rapidly developed. Nevertheless, clinical results obtained with mTOR inhibitors in breast cancer, renal cell carcinoma, neuroendocrine tumors and mantle cell lymphoma push oncologists to actively further develop these drugs, maybe by better selecting the population to which they are offered, through the research of predictive factors of responsiveness. In this review, we aim to describe mechanisms of resistance to mTOR inhibitors, from preclinical and clinical perspectives.
- Published
- 2020
- Full Text
- View/download PDF
16. Evaluation of HER2-specific peptide ligand for its employment as radiolabeled imaging probe
- Author
-
Roberto Bianco, Vladimir Tolmachev, Enrica Calce, Jos Buijs, Stefania De Luca, Emma Langella, Valentina D’Amato, Hadis Honarvar, Michele Saviano, Anna Orlova, Nunzianna Doti, Honarvar, H., Calce, E., Doti, N., Langella, E., Orlova, A., Buijs, J., D'Amato, V., Bianco, R., Saviano, M., Tolmachev, V., and De Luca, S.
- Subjects
0301 basic medicine ,Receptor, ErbB-2 ,medicine.medical_treatment ,lcsh:Medicine ,Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy) ,Plasma protein binding ,Animals, Outbred Strain ,Mice ,Drug Delivery Systems ,0302 clinical medicine ,radiolabeled imaging probe ,lcsh:Science ,skin and connective tissue diseases ,Receptor ,Multidisciplinary ,peptide ligand ,Chemistry ,imaging ,Transmembrane protein ,3. Good health ,Molecular Probe ,030220 oncology & carcinogenesis ,Peptide ,Female ,Breast Neoplasm ,Human ,Protein Binding ,drug design ,Breast Neoplasms ,Article ,Immunoglobulin Fab Fragments ,03 medical and health sciences ,Text mining ,HER2 ,Cell Line, Tumor ,Animals, Outbred Strains ,medicine ,Animals ,Humans ,Medicinsk bioteknologi (med inriktning mot cellbiologi (inklusive stamcellsbiologi), molekylärbiologi, mikrobiologi, biokemi eller biofarmaci) ,neoplasms ,Peptide ligand ,Immunoglobulin Fab Fragment ,Animal ,business.industry ,lcsh:R ,Immunotherapy ,Trastuzumab ,030104 developmental biology ,Radioimmunodetection ,Cell culture ,Molecular Probes ,Cancer research ,lcsh:Q ,Molecular imaging ,Peptides ,business ,Drug Delivery System - Abstract
HER2 transmembrane receptor is an important target in immunotherapy treatment of breast and gastroesophageal cancer. Molecular imaging of HER2 expression may provide essential prognostic and predictive information concerning disseminated cancer and aid in selection of an optimal therapy. Radiolabeled low molecular weight peptide ligands are particularly attractive as probes for molecular imaging, since they reach and bind to the target and clear from non-target organs and blood stream faster than bulky antibodies. In this study, we evaluated a potential HER2-imaging probe, an A9 nonapeptide, derived from the trastuzumab-Fab portion. Its cellular uptake was investigated by mass spectrometry analysis of the cytoplasmic cellular extracts. Moreover, based on in-silico modeling, DTPA chelator was conjugated to N-terminus of A9. 111In-labeled A9 demonstrated nanomolar affinity to HER2-expressing BT474 cells and favorable biodistribution profile in NMRI mice. This study suggests that the peptide A9 represents a good lead candidate for development of molecular probe, to be used for imaging purposes and for the delivery of cytotoxic agents.
- Published
- 2018
- Full Text
- View/download PDF
17. Urokinase-type plasminogen activator receptor (uPAR) expression enhances invasion and metastasis in RAS mutated tumors
- Author
-
Paola Ciciola, Bianca Maria Veneziani, Nunzia Montuori, Gabriella Fontanini, Giancarlo Troncone, Roberta Clara Orsini, Francesca Monteleone, Giuseppe Pignataro, Lucia Grumetto, Alberto Servetto, Valentina D’Amato, Ada Pesapane, Antonino Iaccarino, Roberta De Rosa, Adele Servadio, Antonio Lavecchia, Dario Bruzzese, Concetta Di Mauro, Nicola Zambrano, Luigi Formisano, Roberta Marciano, Sabino De Placido, Roberto Bianco, Mauro, Concetta Di, Pesapane, Ada, Formisano, Luigi, Rosa, Roberta, D'Amato, Valentina, Ciciola, Paola, Servetto, Alberto, Marciano, Roberta, Orsini, Roberta Clara, Monteleone, Francesca, Zambrano, Nicola, Fontanini, Gabriella, Servadio, Adele, Pignataro, Giuseppe, Grumetto, Lucia, Lavecchia, Antonio, Bruzzese, Dario, Iaccarino, Antonino, Troncone, Giancarlo, Veneziani, BIANCA MARIA, Montuori, Nunzia, Placido, Sabino De, and Bianco, Roberto
- Subjects
0301 basic medicine ,Pathology ,Cell ,lcsh:Medicine ,Receptor tyrosine kinase ,Metastasis ,Mice ,0302 clinical medicine ,Cell Movement ,Carcinoma, Non-Small-Cell Lung ,Neoplasms ,Neoplasm Metastasis ,lcsh:Science ,skin and connective tissue diseases ,Receptor ,Multidisciplinary ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Colorectal Neoplasms ,Signal Transduction ,medicine.drug ,medicine.medical_specialty ,Antineoplastic Agents ,Biology ,Article ,Receptors, Urokinase Plasminogen Activator ,03 medical and health sciences ,Cell Adhesion ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,neoplasms ,Urokinase ,lcsh:R ,medicine.disease ,Xenograft Model Antitumor Assays ,biological factors ,Urokinase receptor ,Disease Models, Animal ,030104 developmental biology ,Mutation ,ras Proteins ,biology.protein ,Cancer research ,lcsh:Q ,Plasminogen activator - Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a GPI-anchored cell membrane receptor that focuses urokinase (uPA) proteolytic activity on the cell surface. Its expression is increased in many human cancers, including non-small cell lung cancer (NSCLC) and colorectal cancer (CRC), and correlates with a poor prognosis and early invasion and metastasis. uPAR is able to control, through a cross-talk with tyrosine kinase receptors, the shift between tumor dormancy and proliferation, that usually precedes metastasis formation. Therefore, we investigated the role of uPAR expression in RAS mutated NSCLC and CRC cells. In this study we provided evidence, for the first time, that RAS mutational condition is functionally correlated to uPAR overexpression in NSCLC and CRC cancer cell lines and patient-derived tissue samples. Moreover, oncogenic features related to uPAR overexpression in RAS mutated NSCLC and CRC, such as adhesion, migration and metastatic process may be targeted, in vitro and in vivo, by new anti-uPAR small molecules, specific inhibitors of uPAR-vitronectin interaction. Therefore, anti-uPAR drugs could represent an effective pharmacological strategy for NSCLC and CRC patients carrying RAS mutations.
- Published
- 2017
- Full Text
- View/download PDF
18. The dual PI3K/mTOR inhibitor PKI-587 enhances sensitivity to cetuximab in EGFR-resistant human head and neck cancer models
- Author
-
C. Di Mauro, Lucia Raimondo, Lucia Nappi, R. Marciano, Bianca Maria Veneziani, C Fusciello, Valentina D’Amato, C. D'Amato, Roberto Bianco, R. Rosa, S. De Placido, Alberto Servetto, Luigi Formisano, D'Amato, V, Rosa, R, D'Amato, C, Formisano, L, Marciano, R, Nappi, L, Raimondo, L, Di Mauro, C, Servetto, A, Fusciello, C, Veneziani, BIANCA MARIA, DE PLACIDO, Sabino, and Bianco, Roberto
- Subjects
Cancer Research ,Pathology ,Cetuximab ,Apoptosis ,HNSCC ,Mice ,Antineoplastic Combined Chemotherapy Protocols ,Neoplasm ,Phosphoinositide-3 Kinase Inhibitors ,Mice, Inbred BALB C ,Caspase 3 ,Triazines ,TOR Serine-Threonine Kinases ,Cetuximab resistance ,ErbB Receptors ,Oncology ,Head and Neck Neoplasms ,Monoclonal ,Carcinoma, Squamous Cell ,medicine.drug ,medicine.medical_specialty ,Morpholines ,PI3K-mTOR inhibitors ,Mice, Nude ,Antineoplastic Agents ,Biology ,Antibodies, Monoclonal, Humanized ,Cell Line, Tumor ,Autophagy ,medicine ,Carcinoma ,Animals ,Humans ,neoplasms ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,cetuximab resistance ,Squamous Cell Carcinoma of Head and Neck ,target therapy ,Cell growth ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,Drug Resistance, Neoplasm ,PI3K7mTOR inhibitors ,Cancer research ,Translational Therapeutics - Abstract
Background:Cetuximab is the only targeted agent approved for the treatment of head and neck squamous cell carcinomas (HNSCC), but low response rates and disease progression are frequently reported. As the phosphoinositide 3-kinase (PI3K) and the mammalian target of rapamycin (mTOR) pathways have an important role in the pathogenesis of HNSCC, we investigated their involvement in cetuximab resistance.Methods:Different human squamous cancer cell lines sensitive or resistant to cetuximab were tested for the dual PI3K/mTOR inhibitor PF-05212384 (PKI-587), alone and in combination, both in vitro and in vivo.Results:Treatment with PKI-587 enhances sensitivity to cetuximab in vitro, even in the condition of epidermal growth factor receptor (EGFR) resistance. The combination of the two drugs inhibits cells survival, impairs the activation of signalling pathways and induces apoptosis. Interestingly, although significant inhibition of proliferation is observed in all cell lines treated with PKI-587 in combination with cetuximab, activation of apoptosis is evident in sensitive but not in resistant cell lines, in which autophagy is pre-eminent. In nude mice xenografted with resistant Kyse30 cells, the combined treatment significantly reduces tumour growth and prolongs mice survival.Conclusions:Phosphoinositide 3-kinase/mammalian target of rapamycin inhibition has an important role in the rescue of cetuximab resistance. Different mechanisms of cell death are induced by combined treatment depending on basal anti-EGFR responsiveness. © 2014 Cancer Research UK.
- Published
- 2014
- Full Text
- View/download PDF
19. Hedgehog signalling pathway orchestrates angiogenesis in triple-negative breast cancers
- Author
-
Paola Ciciola, Angela Chambery, Roberta Clara Orsini, Sabino De Placido, Concetta Di Mauro, Roberta Marciano, Roberto Bianco, Bianca Maria Veneziani, Monica Cantile, Luigi Formisano, Valeria Cicatiello, Roberta Di Rosa, Maurizio Di Bonito, Alberto Servetto, Francesca Collina, Valentina D’Amato, Sandro De Falco, Di Mauro, Concetta, Rosa, Roberta, D'Amato, Valentina, Ciciola, Paola, Servetto, Alberto, Marciano, Roberta, Orsini, Roberta Clara, Formisano, Luigi, De Falco, Sandro, Cicatiello, Valeria, Di Bonito, Maurizio, Cantile, Monica, Collina, Francesca, Chambery, Angela, Veneziani, Bianca Maria, De Placido, Sabino, Bianco, Roberto, and Veneziani, BIANCA MARIA
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cancer Research ,Pyridines ,Angiogenesis ,GLI1 ,Triple Negative Breast Neoplasms ,Thrombospondin 1 ,Mice ,angiogenesis ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Aged, 80 and over ,Neovascularization, Pathologic ,Middle Aged ,Bevacizumab ,Angiogenesi ,Oncology ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Female ,TNBC ,Signal Transduction ,Adult ,medicine.medical_specialty ,Paclitaxel ,Mice, Nude ,NVP-LDE225 ,Biology ,Transfection ,Zinc Finger Protein GLI1 ,Young Adult ,03 medical and health sciences ,Paracrine signalling ,Internal medicine ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Gene Silencing ,RNA, Messenger ,Autocrine signalling ,Hedgehog ,Aged ,Cell Proliferation ,Oncogene ,Biphenyl Compounds ,Endothelial Cells ,Membrane Proteins ,Kinase insert domain receptor ,Vascular Endothelial Growth Factor Receptor-2 ,Coculture Techniques ,030104 developmental biology ,Endocrinology ,Tissue Array Analysis ,Cancer cell ,biology.protein ,Cancer research ,Translational Therapeutics ,Neoplasm Transplantation - Abstract
Several evidences suggest a marked angiogenic dependency in triple-negative breast cancer (TNBC) tumorigenesis and a potential sensitivity to anti-angiogenic agents. Herein, the putative role of Hedgehog (Hh) pathway in regulating TNBC-dependent angiogenesis was investigated. Background: Several evidences suggest a marked angiogenic dependency in triple-negative breast cancer (TNBC) tumorigenesis and a potential sensitivity to anti-angiogenic agents. Herein, the putative role of Hedgehog (Hh) pathway in regulating TNBC-dependent angiogenesis was investigated.Methods: Expression and regulation of the Hh pathway transcription factor glioma-associated oncogene homolog1 protein (GLI1) were studied on the endothelial compartment and on TNBC-initiated angiogenesis. To evaluate the translational relevance of our findings, the combination of paclitaxel with the Smo inhibitor NVP-LDE225 was tested in TNBC xenografted mice.Results: Tissue microarray analysis on 200 TNBC patients showed GLI1 overexpression paired with vascular endothelial growth factor receptor 2 (VEGFR2) expression. In vitro, Hh pathway promotes TNBC progression in an autocrine manner, regulating the VEGF/VEGFR2 loop on cancer cell surface, and in a paracrine manner, orchestrating tumour vascularisation. These effects were counteracted by Smo pharmacological inhibition. In TNBC xenografted mice, scheduling NVP-LDE225 rather than bevacizumab provided a better sustained inhibition of TNBC cells proliferation and endothelial cells organisation.Conclusions: This study identifies the Hh pathway as one of the main regulators of tumour angiogenesis in TNBC, thus suggesting Hh inhibition as a potential new anti-angiogenic therapeutic option to be clinically investigated in GLI1 overexpressing TNBC patients.
- Published
- 2017
- Full Text
- View/download PDF
20. Vitamin D3 improves the effects of low dose Der p 2 allergoid treatment in Der p 2 sensitized BALB/c mice
- Author
-
Valentina Amato, Mario Di Gioacchino, Roberto Paganelli, Alessia Lamolinara, Cosmo Rossi, Emanuela Clemente, Alessia Gatta, Sara Cortese, Gianni Mistrello, Claudia Petrarca, and Stefania Zanotta
- Subjects
0301 basic medicine ,Der p 2 ,Allergy ,T regulatory cells ,medicine.medical_treatment ,Immunology ,Inflammation ,Pharmacology ,Allergoid ,BALB/c ,Airborne allergen ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Oral administration ,medicine ,Immunology and Allergy ,Molecular Biology ,Allergen immunotherapy ,biology ,business.industry ,Research ,Immunotherapy ,biology.organism_classification ,Asthma ,Eosinophils ,030104 developmental biology ,IL-10 ,IgE ,medicine.symptom ,business ,Adjuvant ,Vitamin D3 ,030215 immunology - Abstract
Background Airborne allergens can induce an immunological chronic disease characterized by airway hyper responsiveness and inflammation, mediated by exaggerated Th2 immune response. Allergen-specific immunotherapy (AIT) is effective for treating this condition because it is able to modify its natural course by opposing the underlying pathogenic mechanisms and determining immune suppression, immune deviation and tolerance. The rational for the present study was to investigate the possibility of improving allergoid-based IT in terms of efficacy and safety. Recently, 1α,25-dihydroxyvitamin D3 (VD3), the active metabolite of vitamin D3, was described to be a potent inducer of T regulatory cells and to be a good adjuvant in AIT settings. Methods We investigated whether the co-administration of VD3 could potentiate the effect of AIT even when added to a low dose of chemically-modified monomeric allergoid of Der p 2 (d2-OID), in a Derp p 2 (d2)-sensitized BALB/c mice model. Control groups where treated with sham, VD3 alone or d2-OID only. Results The d2-OID alone was not fully successful, as expected for a low dose. VD3 administration was associated with some valuable, although limited, changes in the immunological parameters in the lung. On the contrary, the VD3 adjuvated allergoid vaccine induced the most prominent reduction of airway eosinophilia and Th2 cytokines and concomitant increase of T regulatory cells and IL-10 in the lung and Der p 2-specific IgG2a in the serum. Conclusions The addition of VD3 to a conventional AIT protocol would allow the reduction of allergoid dose needed and therefore, the production costs. Moreover, beneficial immunomodulatory effects have been achieved by the oral administration which might favour the management of the therapy by the patients and their adherence, possibly enhancing the efficacy of the treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12948-016-0044-1) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
21. Wnt4 inhibits cell motility induced by oncogenic Ras
- Author
-
G De Vita, Corrado Garbi, Valentina D’Amato, Angelo Ferraro, R Di Lauro, M De Menna, Alfredo Fusco, DE MENNA, Marta, V., D'Amato, A., Ferraro, Fusco, Alfredo, DI LAURO, Roberto, Garbi, Corrado, and DE VITA, Gabriella
- Subjects
Cancer Research ,animal structures ,Actin cytoskeleton reorganization ,Thyroid Gland ,Motility ,GTPase ,Cell cycle ,Biology ,Rats ,Malignant transformation ,Cell biology ,Phosphatidylinositol 3-Kinases ,Cell Transformation, Neoplastic ,Genes, ras ,Cell Movement ,Wnt4 Protein ,Anti-apoptotic Ras signalling cascade ,Cancer cell ,microRNA ,Genetics ,Animals ,Humans ,Thyroid Neoplasms ,Molecular Biology ,Cytoskeleton - Abstract
Aberrant motility and invasive ability are relevant hallmarks of malignant tumor cells. Pathways regulating the movement of cancer cells from the site of primary tumor toward adjacent and/or distant tissues are not entirely defined. By using a model of malignant transformation induced by Ras, we identified Wnt4 as an early target of Ras oncogenic signaling. Here we show that Wnt4 is repressed by Ras and that forced Wnt4 expression inhibits Ras-induced cell motility. Accordingly, we found that Wnt4 is downregulated in human anaplastic thyroid carcinomas, the most malignant and metastatic thyroid cancer histotype. Wnt4 interferes with Ras-induced actin cytoskeleton reorganization through non-canonical pathways, by altering the balance between the activation of different Rho-family small guanosine triphosphatases (GTPases). Finally, we demonstrate that Wnt4 is post-transcriptionally repressed by miR-24, a Ras-induced micro RNA (miRNA) targeting the 3'-untranslated region (UTR) of Wnt4. Taken together our data highlight a novel Ras-regulated miRNA-dependent circuitry regulating the motile phenotype of cancer cells.Oncogene advance online publication, 1 October 2012; doi:10.1038/onc.2012.419.
- Published
- 2012
- Full Text
- View/download PDF
22. Hedgehog pathway influence in the immune escape of tumor cells through PDL-1 modulation
- Author
-
Filomena Napolitano, R. Marciano, Ada Pesapane, Luigi Formisano, C. Di Mauro, Antonio Santaniello, Alberto Servetto, Valentina D’Amato, R. Rosa, and Roberto Bianco
- Subjects
Oncology ,Modulation ,business.industry ,Immune escape ,Medicine ,Tumor cells ,Hematology ,business ,Hedgehog signaling pathway ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
23. Role of p21-activated kinase (PAK) in K-RAS mutant human colorectal cancer models
- Author
-
Roberto Bianco, Paola Ciciola, Roberta Clara Orsini, Filomena Napolitano, C. Di Mauro, S. De Placido, Alberto Servetto, R. Marciano, Valentina D’Amato, and R. Rosa
- Subjects
Oncology ,Colorectal cancer ,Kinase ,business.industry ,Mutant ,Cancer research ,medicine ,Hematology ,medicine.disease ,business - Published
- 2018
- Full Text
- View/download PDF
24. Mechanisms of lapatinib resistance in HER2-driven breast cancer
- Author
-
Valentina D’Amato, Roberta De Rosa, Luigi Formisano, Lucia Raimondo, Roberto Bianco, Mario Giuliano, Sabino De Placido, D'Amato, Valentina, Raimondo, Lucia, Formisano, Luigi, Giuliano, Mario, DE PLACIDO, Sabino, Rosa, Roberta, and Bianco, Roberto
- Subjects
Oncology ,medicine.medical_specialty ,Poor prognosis ,Receptor, ErbB-2 ,Intrinsic resistance ,Resistance ,Antineoplastic Agents ,Breast Neoplasms ,Aggressive phenotype ,ErbB2/HER2 ,Lapatinib ,Breast cancer ,Acquired resistance ,Trastuzumab ,Internal medicine ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,skin and connective tissue diseases ,neoplasms ,business.industry ,Phosphotransferases ,Gene Amplification ,General Medicine ,Protein-Tyrosine Kinases ,medicine.disease ,Receptors, Estrogen ,Drug Resistance, Neoplasm ,Mutation ,Quinazolines ,Female ,Pertuzumab ,business ,medicine.drug - Abstract
Targeted therapies have been approved for various malignancies but the acquisition of resistance remains a substantial challenge in the clinical management of advanced cancers. Twenty-five per cent of breast cancers overexpress ErbB2/HER2, which confers a more aggressive phenotype and is associated with a poor prognosis. HER2-targeting therapies (trastuzumab, pertuzumab, TDM1 and lapatinib) are available, but a significant fraction of HER2-positive breast cancers eventually relapse or progress. This suggests that acquired or intrinsic resistance enables escape from HER2 inhibition. This review focuses on mechanisms of intrinsic/acquired resistance to lapatinib identified in preclinical and clinical studies. A better understanding of these mechanisms could lead to novel predictive markers of lapatinib response and to novel therapeutic strategies for breast cancer patients.
- Published
- 2015
25. Epidermal growth factor-receptor activation modulates Src-dependent resistance to lapatinib in breast cancer models
- Author
-
Valentina D’Amato, Sarah J. Parsons, Giancarlo Troncone, Sabino De Placido, Vincenzo Damiano, Lucia Raimondo, Bianca Maria Veneziani, Roberto Bianco, Lucia Nappi, Francesca Iommelli, C. D'Amato, Roberta Marciano, Luigi Formisano, Roberta De Rosa, Antonella Scorziello, Formisano, L, Nappi, L, Rosa, R, Marciano, R, D., Amato C, D., Amato V, Damiano, V, Raimondo, L, Iommelli, F, Scorziello, Antonella, Troncone, Giancarlo, Veneziani, BIANCA MARIA, Parsons, Sj, DE PLACIDO, Sabino, and Bianco, Roberto
- Subjects
Lung Neoplasms ,Receptor, ErbB-2 ,Metastasis ,Mice ,Cell Movement ,cetuximab ,Antineoplastic Combined Chemotherapy Protocols ,Epidermal growth factor receptor ,Enzyme Inhibitors ,RNA, Small Interfering ,skin and connective tissue diseases ,Medicine(all) ,Mice, Inbred BALB C ,biology ,Drug Synergism ,ErbB Receptors ,src-Family Kinases ,Female ,RNA Interference ,Signal transduction ,Tyrosine kinase ,Research Article ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug ,Cell Survival ,Transplantation, Heterologous ,Mice, Nude ,Antineoplastic Agents ,Breast Neoplasms ,Lapatinib ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Benzodioxoles ,Protein Kinase Inhibitors ,Cell Proliferation ,Epidermal Growth Factor ,Cell growth ,Cancer ,medicine.disease ,Enzyme Activation ,Drug Resistance, Neoplasm ,Quinazolines ,Cancer research ,biology.protein ,saracatinib ,Neoplasm Transplantation - Abstract
Introduction: Src tyrosine kinase overactivation has been correlated with a poor response to human epidermal growth factor receptor 2 (HER2) inhibitors in breast cancer. To identify the mechanism by which Src overexpression sustains this resistance, we tested a panel of breast cancer cell lines either sensitive or resistant to lapatinib.Methods: To determine the role of Src in lapatinib resistance, we evaluated the effects of Src inhibition/silencing in vitro on survival, migration, and invasion of lapatinib-resistant cells. In vivo experiments were performed in JIMT-1 lapatinib-resistant cells orthotopically implanted in nude mice. We used artificial metastasis assays to evaluate the effect of Src inhibition on the invasiveness of lapatinib-resistant cells. Src-dependent signal transduction was investigated with Western blot and ELISA analyses.Results: Src activation was higher in lapatinib-resistant than in lapatinib-sensitive cells. The selective small-molecule Src inhibitor saracatinib combined with lapatinib synergistically inhibited the proliferation, migration, and invasion of lapatinib-resistant cells. Saracatinib combined with lapatinib significantly prolonged survival of JIMT-1-xenografted mice compared with saracatinib alone, and impaired the formation of lung metastases. Unexpectedly, in lapatinib-resistant cells, Src preferentially interacted with epidermal growth factor receptor (EGFR) rather than with HER2. Moreover, EGFR targeting and lapatinib synergistically inhibited survival, migration, and invasion of resistant cells, thereby counteracting Src-mediated resistance. These findings demonstrate that Src activation in lapatinib-resistant cells depends on EGFR-dependent rather than on HER2-dependent signaling.Conclusions: Complete pharmacologic EGFR/HER2 inhibition is required to reverse Src-dependent resistance to lapatinib in breast cancer. © 2014 Formisano et al.; licensee BioMed Central Ltd.
- Published
- 2014
- Full Text
- View/download PDF
26. P0081 Src inhibitors act through different mechanisms to cooperate with EGFR or MEK inhibitors in NSCLC models sensitive or resistant to erlotinib
- Author
-
B.M. Veneziani, Roberta Marciano, C. Di Mauro, Luigi Formisano, Valentina D’Amato, S. De Placido, R. Rosa, Lucia Nappi, Lucia Raimondo, Vincenzo Damiano, Alberto Servetto, C. D'Amato, Roberto Bianco, Raimondo, L., Formisano, L., Rosa, R., D?amato, C., D?amato, V., Nappi, L., Marciano, R., Di Mauro, C., Servetto, A., Damiano, V., Veneziani, BIANCA MARIA, DE PLACIDO, Sabino, and Bianco, Roberto
- Subjects
Cancer Research ,NSCLC cell lines ,Chemistry ,Pharmacology ,respiratory tract diseases ,Dasatinib ,Gefitinib ,Oncology ,Cancer research ,medicine ,Selumetinib ,Src inhibitor ,Erlotinib ,neoplasms ,Bosutinib ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug ,EGFR inhibitors - Abstract
Background The EGFR tyrosine kinase inhibitors gefitinib and erlotinib are first-line therapy for NSCLCs harbouring EGFR-activating mutations. The intrinsic and acquired resistance to these agents is a relevant clinical issue. Although Src tyrosine kinase has been involved in such resistance in preclinical models, clinical development of these agents has been so far limited. Methods We used a panel of human NSCLC cell lines: PC9 and HCC827 (EGFR-activating mutation; highly sensitive to erlotinib), Calu3 (EGFR/Ras wild-type, wt; moderately sensitive), Calu3-ER (with acquired resistance), H1299 and A549 (Ras mutant; resistant), H1975 (EGFR T790M mutant; resistant). In these models, we tested three different Src inhibitors (saracatinib, dasatinib, and bosutinib), both in vitro and in vivo. Findings NSCLC cell lines showed different activation of EGFR-dependent and Src-dependent pathways and variable sensitivity to Src inhibitors. A kinase assay demonstrated that all the compounds are able to directly inhibit EGFR tyrosine kinase variants. In cell lysates only saracatinib and bosutinib efficiently reduced EGFR activation, while dasatinib was the more effective agent in inhibiting Src tyrosine kinase. Consistently, in EGFR-activating mutant, erlotinib sensitive cells, saracatinib and bosutinib showed anti-proliferative effects related to simultaneous EGFR/Src inhibition. In EGFR wt/Ras mutant cells Src inhibition by dasatinib interfered with cell proliferation and signal transduction. Since Src inhibitors had only moderate effects as single agents we tested the combination of saracatinib with EGFR inhibitors (erlotinib or cetuximab) in EGFR-addicted cells, and of dasatanib with MEK inhibitors (selumetinib) in Ras mutant, erlotinib resistant models. These combinations were effective both in vitro and in nude mice, inhibiting tumour growth, prolonging mice survival, and interfering with signal transduction. Importantly, the combination of saracatanib and cetuximab was effective also in the erlotinib resistant, EGFR T790M mutant model. Interpretation Src inhibitors may act with different mechanisms in NSCLC cell lines, depending on EGFR/Ras mutational profile. Integration of anti-Src agents with EGFR or MEK inhibitors could represent effective therapeutic options for different cohorts of NSCLC patients.
- Published
- 2014
27. EFFECTS OF HEDGEHOG SIGNALING INHIBITION ON EPITHELIAL-STROMAL INTERACTIONS IN TRIPLE NEGATIVE BREAST CANCER CELLS
- Author
-
B.M. Veneziani, R. Marciano, Roberto Bianco, R. Rosa, Valentina D’Amato, C. Di Mauro, Lucia Raimondo, S. De Placido, Alberto Servetto, Luigi Formisano, A., Servetto, L., Raimondo, L., Formisano, R., Marciano, C., Di Mauro, R., Rosa, V., D'Amato, Veneziani, BIANCA MARIA, S., De Placido, and R., Bianco
- Subjects
Oncology ,medicine.medical_specialty ,Stromal cell ,biology ,Bevacizumab ,business.industry ,Angiogenesis ,Hematology ,medicine.disease ,Hedgehog signaling pathway ,Breast cancer ,GLI1 ,Internal medicine ,Cancer cell ,biology.protein ,Cancer research ,Medicine ,business ,Triple-negative breast cancer ,medicine.drug - Abstract
Aim: Triple-negative breast cancer (TNBC) is a group of tumors that do not express HER2, estrogen and progesterone receptors. Due to reduced response to conventional antitumor therapies and poor prognosis, new targeted agents are needed for such aggressive sub-type of breast cancer. Multiple lines of evidence support the idea that deregulation of Hedgehog (Hh) signaling has a role in the pathogenesis of breast cancer, in part through the promotion of epithelial-stromal interactions. Therefore, the inhibition of the Hh pathway has been proposed as an interesting therapeutic approach Methods: The main objective of this study is to investigate the role of Hh signaling pathway in TNBC. To this aim, we used a panel of human breast cancer cell lines, including five cancer cells lines positive for ER, PR and HER2 expression (nTNBC) and five Triple Negative Breast Cancer cell lines (TNBC). The effects induced by the Smo-inhibitor NVP-LDE225 on proliferation, angiogenesis and signal transduction of breast cancer cells were investigated Results: GLI1, one of the major transcription factors induced by Hh signaling activation, is more expressed in TNBC than in nTNBC cell lines. Consistently, NVP-LDE225 treatment induced a more pronounced inhibitory effect on TNBC, in terms of tumor growth: while nTNBC cells display an IC50 of∼5mM, TNBC cell lines are more sensitive, with an average IC50 of∼2mM. In addition, Hh inhibition caused a robust impairment of TNBC cells invasion capabilities. These effects are coupled with a strong inhibition of VEGFA production by both tumor and stromal cells (human fibroblasts and HUVECs). Accordingly, NVP-LDE225 treatment interfered with HUVEC capillary tube formation, an effect even more evident than that observed with bevacizumab, the only targeted agent approved to date for TNBC patients Conclusions: Our results suggest that Hh has a specific role in breast epithelial-stromal interactions by regulation of angiogenesis. An orthotopic in vivo experiment in nude mice xenografted with TNBC cells, testing the combination of NVP-LDE225 with bevacizumab, is ongoing Disclosure: All authors have declared no conflicts of interest.
- Published
- 2014
28. Inhibition of Hedgehog signalling by NVP-LDE225 (Erismodegib) interferes with growth and invasion of human renal cell carcinoma cells
- Author
-
Lucia Raimondo, Valentina D’Amato, Luigi Formisano, Franco Fulciniti, Lucia Nappi, R. Rosa, Roberto Bianco, S. De Placido, Cesar A. Bianco, Alberto Servetto, C. D'Amato, A Cipolletta, R. Marciano, Fortunato Ciardiello, Bianca Maria Veneziani, C. Di Mauro, D'Amato, C., Rosa, R., Marciano, R., D'Amato, V., Formisano, L., Nappi, L., Raimondo, L., Di Mauro, C., Servetto, A., Fulciniti, F., Cipolletta, A., Bianco, C., Ciardiello, F., Veneziani, BIANCA MARIA, DE PLACIDO, Sabino, Bianco, Roberto, D'Amato, C, Rosa, R, Marciano, R, D'Amato, V, Formisano, L, Nappi, L, Raimondo, L, Di Mauro, C, Servetto, A, Fulciniti, F, Cipolletta, A, Bianco, C, Ciardiello, Fortunato, Veneziani, Bm, De Placido, S, and Bianco, R.
- Subjects
Cancer Research ,Indoles ,Lung Neoplasms ,Pyridines ,urologic and male genital diseases ,Receptors, G-Protein-Coupled ,Mice ,sunitinib resistance ,Cell Movement ,immune system diseases ,Antineoplastic Combined Chemotherapy Protocols ,Sunitinib ,Mice, Inbred BALB C ,Nuclear Proteins ,Ribosomal Protein S6 Kinases, 70-kDa ,virus diseases ,Drug Synergism ,RCC ,Smoothened Receptor ,female genital diseases and pregnancy complications ,Kidney Neoplasms ,Tumor Burden ,Biphenyl compound ,Actin Cytoskeleton ,Oncology ,Neoplasm Micrometastasis ,Mitogen-Activated Protein Kinases ,Signal transduction ,Signal Transduction ,medicine.medical_specialty ,Kruppel-Like Transcription Factors ,NVP-LDE225 ,Mice, Nude ,Zinc Finger Protein Gli2 ,Biology ,Zinc Finger Protein GLI1 ,Inhibitory Concentration 50 ,Cell Line, Tumor ,Internal medicine ,Carcinoma ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Pyrroles ,Everolimus ,Carcinoma, Renal Cell ,Hedgehog ,Cell Proliferation ,Sirolimus ,Cell growth ,Biphenyl Compounds ,medicine.disease ,Actin cytoskeleton ,Xenograft Model Antitumor Assays ,Actins ,Endocrinology ,Cell culture ,Cancer research ,Hedgehog, RCC, NVP-LDE225, sunitinib resistance ,Paxillin ,Translational Therapeutics ,Proto-Oncogene Proteins c-akt ,Transcription Factors - Abstract
Background: Multiple lines of evidence support that the Hedgehog (Hh) signalling has a role in the maintenance and progression of different human cancers. Therefore, inhibition of the Hh pathway represents a valid anticancer therapeutic approach for renal cell carcinoma (RCC) patients. NVP-LDE225 is a Smoothened (Smo) antagonist that induces dose-related inhibition of Hh and Smo-dependent tumour growth. Methods: We assayed the effects of NVP-LDE225 alone or in combination with everolimus or sunitinib on the growth and invasion of human RCC models both in vitro and in vivo. To this aim, we used a panel of human RCC models, comprising cells with acquired resistance to sunitinib - a multiple tyrosine kinase inhibitor approved as a first-line treatment for RCC. Results: NVP-LDE225 cooperated with either everolimus or sunitinib to inhibit proliferation, migration, and invasion of RCC cells even in sunitinib-resistant (SuR) cells. Some major transducers involved in tumour cell motility, including paxillin, were also efficiently inhibited by the combination therapy, as demonstrated by western blot and confocal microscopy assays. Moreover, these combined treatments inhibited tumour growth and increased animal survival in nude mice xenografted with SuR RCC cells. Finally, lung micrometastasis formation was reduced when mice were treated with NVP-LDE225 plus everolimus or sunitinib, as evidenced by artificial metastatic assays. Conclusions: Hedgehog inhibition by NVP-LDE225 plus sunitinib or everolimus bolsters antitumour activity by interfering with tumour growth and metastatic spread, even in SuR cells. Thus, this new evidence puts forward a new promising therapeutic approach for RCC patients.
- Published
- 2014
29. The role of p21 activated kinase (PAK) in human colorectal cancer cell lines and resistance to cetuximab
- Author
-
R. Marciano, Valentina D’Amato, S. De Placido, R. Rosa, C. Di Mauro, Paola Ciciola, Alberto Servetto, Roberta Clara Orsini, and Roberto Bianco
- Subjects
Oncology ,medicine.medical_specialty ,Cetuximab ,Kinase ,Colorectal cancer ,business.industry ,Hematology ,medicine.disease ,Cell culture ,Internal medicine ,medicine ,Cancer research ,business ,medicine.drug - Published
- 2016
- Full Text
- View/download PDF
30. Sphingosine kinase 1 overexpression contributes to cetuximab resistance in human colorectal cancer models
- Author
-
Sabino De Placido, Umberto Malapelle, Lucia Nappi, Roberta Marciano, Roberto Bianco, Chiara Carlomagno, Gabriella Marfe, Silvana Del Vecchio, Bianca Maria Veneziani, C. D'Amato, Adelaide Greco, Giancarlo Troncone, Luigi Formisano, Antonella Zannetti, Valentina D’Amato, Roberta De Rosa, Alfonso De Stefano, and Vincenzo Damiano
- Subjects
Cancer Research ,Ceramide ,Colorectal cancer ,Cetuximab ,Gene Expression ,Mice, Nude ,Antineoplastic Agents ,Pharmacology ,Antibodies, Monoclonal, Humanized ,chemistry.chemical_compound ,Mice ,Sphingosine ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Sphingosine-1-Phosphate Receptors ,biology ,business.industry ,Fingolimod Hydrochloride ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Phosphotransferases (Alcohol Group Acceptor) ,Receptors, Lysosphingolipid ,Treatment Outcome ,Oncology ,chemistry ,Sphingosine kinase 1 ,Drug Resistance, Neoplasm ,Propylene Glycols ,Cancer cell ,biology.protein ,Signal transduction ,business ,Colorectal Neoplasms ,medicine.drug - Abstract
Purpose: Although the anti–EGF receptor (EGFR) monoclonal antibody cetuximab is an effective strategy in colorectal cancer therapy, its clinical use is limited by intrinsic or acquired resistance. Alterations in the “sphingolipid rheostat”—the balance between the proapoptotic molecule ceramide and the mitogenic factor sphingosine-1-phosphate (S1P)—due to sphingosine kinase 1 (SphK1) overactivation have been involved in resistance to anticancer-targeted agents. Moreover, cross-talks between SphK1 and EGFR-dependent signaling pathways have been described. Experimental design: We investigated SphK1 contribution to cetuximab resistance in colorectal cancer, in preclinical in vitro/in vivo models, and in tumor specimens from patients. Results: SphK1 was found overexpressed and overactivated in colorectal cancer cells with intrinsic or acquired resistance to cetuximab. SphK1 contribution to resistance was supported by the demonstration that SphK1 inhibition by N,N-dimethyl-sphingosine or silencing via siRNA in resistant cells restores sensitivity to cetuximab, whereas exogenous SphK1 overexpression in sensitive cells confers resistance to these agents. Moreover, treatment of resistant cells with fingolimod (FTY720), a S1P receptor (S1PR) antagonist, resulted in resensitization to cetuximab both in vitro and in vivo, with inhibition of tumor growth, interference with signal transduction, induction of cancer cells apoptosis, and prolongation of mice survival. Finally, a correlation between SphK1 expression and cetuximab response was found in colorectal cancer patients. Clin Cancer Res; 19(1); 138–47. ©2012 AACR.
- Published
- 2012
- Full Text
- View/download PDF
31. Reactive Oxygen Species, Ki-Ras, and Mitochondrial Superoxide Dismutase Cooperate in Nerve Growth Factor-induced Differentiation of PC12 Cells*
- Author
-
Valentina D’Amato, Corrado Garbi, Stefano Amente, Mariarosaria Santillo, Antonio Porcellini, Silvana Cassano, Patrizio Castagnola, Enrico V. Avvedimento, Emmanuele De Vendittis, Immacolata Castellano, Massimo Papale, Savina Agnese, Maria Rosaria Ruocco, Cassano, S., Agnese, Savina, D'Amato, Valentina, Papale, M., Garbi, Corrado, Castagnola, P., Ruocco, MARIA ROSARIA, Castellano, Immacolata, DE VENDITTIS, Emmanuele, Santillo, Mariarosaria, Amente, Stefano, Porcellini, Antonio, and Avvedimento, VITTORIO ENRICO
- Subjects
MAPK/ERK pathway ,Small interfering RNA ,Neurite ,MAP Kinase Signaling System ,Cellular differentiation ,Mitochondrion ,Biochemistry ,PC12 Cells ,Superoxide dismutase ,Nerve Growth Factor ,KRAS ,Animals ,Humans ,Molecular Biology ,Cytoskeleton ,chemistry.chemical_classification ,Reactive oxygen species ,oxidative stre ,Mitogen-Activated Protein Kinase 3 ,biology ,Superoxide Dismutase ,NERVE GROWTH-FACTOR ,Cell Differentiation ,Cell Biology ,Molecular biology ,Mitochondria ,Rats ,Nerve growth factor ,Genes, ras ,chemistry ,nervous system ,Mutation ,biology.protein ,ras Proteins ,Reactive Oxygen Species ,Signal Transduction - Abstract
Nerve growth factor (NGF) induces terminal differentiation in PC12, a pheochromocytoma-derived cell line. NGF binds a specific receptor on the membrane and triggers the ERK1/2 cascade, which stimulates the transcription of neural genes. We report that NGF significantly affects mitochondrial metabolism by reducing mitochondrial-produced reactive oxygen species and stabilizing the electrochemical gradient. This is accomplished by stimulation of mitochondrial manganese superoxide dismutase (MnSOD) both transcriptionally and post-transcriptionally via Ki-Ras and ERK1/2. Activation of MnSOD is essential for completion of neuronal differentiation because 1) expression of MnSOD induces the transcription of a neuronal specific promoter and neurite outgrowth, 2) silencing of endogenous MnSOD by small interfering RNA significantly reduces transcription induced by NGF, and 3) a Ki-Ras mutant in the polylysine stretch at the COOH terminus, unable to stimulate MnSOD, fails to induce complete differentiation. Overexpression of MnSOD restores differentiation in cells expressing this mutant. ERK1/2 is also downstream of MnSOD, as a SOD mimetic drug stimulates ERK1/2 with the same kinetics of NGF and silencing of MnSOD reduces NGF-induced late ERK1/2. Long term activation of ERK1/2 by NGF requires SOD activation, low levels of hydrogen peroxide, and the integrity of the microtubular cytoskeleton. Confocal immunofluorescence shows that NGF stimulates the formation of a complex containing membrane-bound Ki-Ras, microtubules, and mitochondria. We propose that active NGF receptor induces association of mitochondria with plasma membrane. Local activation of ERK1/2 by Ki-Ras stimulates mitochondrial SOD, which reduces reactive oxygen species and produces H(2)O(2). Low and spatially restricted levels of H(2)O(2) induce and maintain long term ERK1/2 activity and ultimately differentiation of PC12 cells.
- Published
- 2010
32. Src Inhibitors Act Through Different Mechanisms to Cooperate with Egfr or Mek Inhibitors in Nsclc Models Sensitive or Resistant to Erlotinib
- Author
-
Lucia Raimondo, Roberto Bianco, Luigi Formisano, Valentina D’Amato, Lucia Nappi, R. Rosa, C. Di Mauro, C. D'Amato, R. Marciano, S. De Placido, and Alberto Servetto
- Subjects
Cetuximab ,business.industry ,Hematology ,respiratory tract diseases ,Dasatinib ,Gefitinib ,Oncology ,Cancer research ,medicine ,Selumetinib ,Erlotinib ,business ,neoplasms ,Bosutinib ,medicine.drug ,EGFR inhibitors ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Aim: The EGFR TKIs gefitinib and erlotinib are the first-line therapy for NSCLCs harboring EGFR-activating mutations. The intrinsic resistance to these agents and the onset of acquired resistance in the responders is a relevant clinical issue and the aim of this work is to test the efficacy of three different Src inhibitors in these models of resistance. Methods: We used a panel of human NSCLC cell lines with different EGFR/Ras mutational profile and erlotinib sensitivity: PC9 and HCC827 (EGFR-activating mutation, highly sensitive), Calu3 (EGFR/Ras wt; moderately sensitive), Calu3-ER (with acquired resistance), H1299 and A549 (Ras mutant; resistant), H1975 (EGFR T790M mutant; resistant). In these models, we tested three different Src inhibitors (saracatinib, dasatinib and bosutinib), both in vitro and in vivo Results: NSCLC cell lines showed different activation of EGFR- and Src-dependent pathways and variable sensitivity to Src inhibitors. A kinase assay demonstrated that all the compounds are able to directly inhibit not only Src, but also EGFR TK variants. However, in cell lysates only saracatinib and bosutinib efficiently reduced EGFR activation, while dasatinib was the more effective agent in inhibiting Src TK. In EGFR-activating mutant, erlotinib sensitive cells, saracatinib and bosutinib showed anti-proliferative effects related to simultaneous EGFR/Src inhibition. In EGFR wt/Ras mutant cells Src inhibition by dasatinib interfered with cell proliferation and signal transduction. Since Src inhibitors had only moderate effects as single agents, both in vitro and in vivo, we tested the combination of saracatinib with EGFR inhibitors (erlotinib or cetuximab) in EGFR-addicted cells, and of dasatanib with MEK inhibitors (selumetinib) in Ras mutant, erlotinib resistant models. These combinations were effective both in vitro and in vivo, inhibiting tumor growth, prolonging mice survival and interfering with signal transduction. Importantly, the combination of saracatanib and cetuximab was effective also in the erlotinib resistant, EGFR T790M mutant model. Conclusions: Src inhibitors may act with different mechanisms in NSCLC cell lines, depending on EGFR/Ras mutational profile. Integration of anti-Src agents with EGFR or MEK inhibitors could represent effective therapeutic options for different cohorts of NSCLC patients. Disclosure: All authors have declared no conflicts of interest.
- Published
- 2014
- Full Text
- View/download PDF
33. Abstract 950: Src tyrosine kinase contributes to lapatinib resistance in human breast cancer models
- Author
-
Giancarlo Troncone, Luigi Formisano, Roberto Bianco, Sabino De Placido, Umberto Malapelle, Ana Paula De Maio, Vincenzo Damiano, Roberta De Rosa, Valentina D’Amato, Lucia Raimondo, Roberta Marciano, C. D'Amato, and Lucia Nappi
- Subjects
Cancer Research ,medicine.drug_class ,business.industry ,Cancer ,Pharmacology ,medicine.disease ,Lapatinib ,Metastatic breast cancer ,Tyrosine-kinase inhibitor ,Oncology ,Trastuzumab ,Cancer cell ,medicine ,Cancer research ,skin and connective tissue diseases ,business ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug - Abstract
Background. The dual EGFR/HER2 tyrosine kinase inhibitor (TKI) lapatinib is approved in metastatic breast cancer patients after trastuzumab failure. Unfortunately, several patients do not respond to treatment due to constitutive resistance, and even in responders the occurrence of disease progression is often observed. Recently, the activation of Src TK has been associated with resistance to anti-HER2 drugs, both trastuzumab and lapatinib, in HER2 overexpressing breast cancer cells. However, the molecular mechanisms by which Src activation may affect response to HER2 antagonists are still partially unclear. Methods. We used a panel of HER2 expressing human breast cancer cell lines with different degree of response to lapatinib: SKBR3, BT474 and MDA-MB-361 (high sensitivity), KPL-4 (moderate sensitivity), JIMT-1 (low sensitivity). Moreover, we generated a cell line with acquired resistance to lapatinib, MDA-MB-361-LR (Lapatinib Resistant). On these models, we investigated the effect of Src silencing via small interference RNA (siRNA) or pharmacological inhibition by saracatinib on lapatinib response, both in vitro and in vivo. Results. The combination of Src inhibition with lapatinib synergistically inhibited survival of human breast cancer cells with both intrinsic or acquired resistance to lapatinib, by interfering with signal transducers involved in cell proliferation and survival. Moreover, both saracatinib and Src specific siRNAs were able to reduce migration and invasion capabilities of breast cancer cells, by affecting Focal Adhesion Kinase (FAK) and paxillin activation. In nude mice xenografted with JIMT-1 cells, the combined treatment with saracatinib and lapatinib produced a cooperative antitumor effect with inhibition of tumor growth, interference with signal transduction and prolongation of mice survival. The combination strongly reduced the incidence of lung metastasis, as revealed through experimental metastasis assays. Interestingly, a significant reduction of EGFR/HER2 co-immunoprecipitation was found in resistant cells, thus suggesting that growth signals could be transduced by other type of TKRs dimers in these cells. Moreover, in breast cancer cell lines with both intrinsic or acquired resistance to lapatinib, Src interacted with EGFR rather than with HER2, in opposition to what was observed in sensitive cells. Consistently, EGFR inhibition through cetuximab or EGFR specific siRNAs was able to partially restore lapatinib sensitivity in resistant cells, by interfering with the reciprocal activation of EGFR and Src. Conclusions. Src plays an important role in the onset of resistance to lapatinib in breast cancer cell lines, and its interaction with EGFR may be involved in this event. Therefore, the combined inhibition of Src and EGFR/HER2 axis may represent a potentially effective therapeutic strategy in order to prevent or overcome resistance to lapatinib. Citation Format: Roberta Rosa, Lucia Nappi, Luigi Formisano, Claudia D'Amato, Valentina D'Amato, Vincenzo Damiano, Roberta Marciano, Ana Paula De Maio, Lucia Raimondo, Umberto Malapelle, Giancarlo Troncone, Sabino De Placido, Roberto Bianco. Src tyrosine kinase contributes to lapatinib resistance in human breast cancer models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 950. doi:10.1158/1538-7445.AM2013-950 Note: This abstract was not presented at the AACR Annual Meeting 2013 because the presenter was unable to attend.
- Published
- 2013
- Full Text
- View/download PDF
34. Effects of SRC Inhibition on Survival, Migration and Invasion of Human Breast Cancer Cells Resistant to Lapatinib
- Author
-
Vincenzo Damiano, C. D'Amato, R. Marciano, T. Gelardi, Roberto Bianco, Lucia Nappi, Luigi Formisano, A.P. De Maio, R. Rosa, and Valentina D’Amato
- Subjects
Cetuximab ,biology ,business.industry ,Hematology ,Lapatinib ,medicine.disease ,Metastatic breast cancer ,Receptor tyrosine kinase ,Breast cancer ,Oncology ,Trastuzumab ,Cancer cell ,medicine ,Cancer research ,biology.protein ,skin and connective tissue diseases ,business ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug - Abstract
Background HER2, a member of the HER family, is a transmembrane tyrosine kinase receptor overexpressed in almost 30% of breast cancer patients. Lapatinib is a small molecule HER2 inhibitor approved in metastatic breast cancer patients after trastuzumab failure. Unfortunately, the resistance against lapatinib is observed even in responding patients. Src is an intracellular kinase involved in tumor growth, angiogenesis and migration and it is related to trastuzumab resistance in human breast cancer cell lines. Materials and methods We used different HER2 expressing human breast cancer cell lines, such as MDA-MB-361, MDA-MB-231 and BT474 (sensitive to lapatinib) JIMT-1 and KPL-4 (low sensitivity against lapatinib), and MDA-MB-361-LR (acquired resistance to lapatinib). We performed survival, migration and invasion assays and WB analysis in all cell lines treated with lapatinib, saracatinib (a Src inhibitor) or the combination. We also used nude mice xenografted with JIMT-1 cells and in vivo artificial metastatic assay. Results We observed that the combination treatment of lapatinib plus saracatinib is able to inhibit survival, migration and invasion in vitro and to regulate several proteins such as Src, Akt, MAPK, paxillin and FAK in all cancer cell lines tested. In nude mice xenografted with JIMT-1 cells, the combined treatment reduced tumor growth, prolonged survival and strongly decreased the incidence of lung metastases. Interestingly, we observed that Src preferentially binds to HER2 in lapatinib sensitive cells and with EGFR in resistant ones. EGFR inhibition, through the use of cetuximab or siRNA silencing, strongly enhanced the effect of lapatinib on the growth of resistant cancer cells. The combined treatment of lapatinib and cetuximab, moreover, significantly reduced the activation of several intracellular transducers. Conclusions We demonstrated that Src plays an important role in the development of resistance to lapatinib in breast cancer cell lines, in particular affecting invasion and migration, and that EGFR may mediate its activation. These results may suggest the clinical development of lapatinib and cetuximab combination in patients resistant to lapatinib. Disclosure All authors have declared no conflicts of interest.
- Published
- 2012
- Full Text
- View/download PDF
35. Src inhibitors act through different mechanisms in Non-Small Cell Lung Cancer models depending on EGFR and RAS mutational status
- Author
-
Roberta Marciano, Concetta Di Mauro, Antonio Randazzo, Sabino De Placido, Luigi Formisano, Sandro Cosconati, Bianca Maria Veneziani, Roberto Bianco, Nunzia Montuori, Sarah J. Parsons, Roberta De Rosa, Simona Brillante, Alberto Servetto, Lucia Raimondo, Valentina D’Amato, Roberta Clara Orsini, Formisano, L, D'Amato, V, Servetto, A, Brillante, S, Raimondo, L, Di Mauro, C, Marciano, R, Clara Orsini, R, Cosconati, Sandro, Randazzo, A, Parsons, Sj, Montuori, N, Maria Veneziani, B, De Placido, S, Rosa, R, Bianco, R., Formisano, Luigi, Valentina D'Amato, Null, Servetto, Alberto, Brillante, Simona, Raimondo, Lucia, Mauro, Concetta Di, Marciano, Roberta, Orsini, Roberta Clara, Randazzo, Antonio, Parsons, Sarah, Montuori, Nunzia, Veneziani, BIANCA MARIA, Placido, Sabino De, Rosa, Roberta, and Bianco, Roberto
- Subjects
Lung Neoplasms ,Cell Survival ,Blotting, Western ,Dasatinib ,Cetuximab ,Mice, Nude ,Pharmacology ,NSCLC ,T790M ,Erlotinib Hydrochloride ,Gefitinib ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Benzodioxoles ,Protein Kinase Inhibitors ,MEK inhibitors ,EGFR inhibitors ,Mice, Inbred BALB C ,MEK inhibitor ,business.industry ,Xenograft Model Antitumor Assays ,3. Good health ,Tumor Burden ,respiratory tract diseases ,ErbB Receptors ,EGFR inhibitor ,src-Family Kinases ,Oncology ,Drug resistance ,Mutation ,Quinazolines ,ras Proteins ,RNA Interference ,business ,Bosutinib ,medicine.drug ,Proto-oncogene tyrosine-protein kinase Src ,Research Paper ,Src - Abstract
Resistance to the EGFR tyrosine kinase inhibitors (TKIs) gefitinib and erlotinib, often related to Ras or secondary EGFR mutations, is a relevant clinical issue in Non- Small Cell Lung Cancer (NSCLC). Although Src TK has been involved in such resistance, clinical development of its inhibitors has been so far limited. To better define the molecular targets of the Src TKIs saracatinib, dasatinib and bosutinib, we used a variety of in vitro/in vivo studies. Kinase assays supported by docking analysis demonstrated that all the compounds directly inhibit EGFR TK variants. However, in live cells only saracatinib efficiently reduced EGFR activation, while dasatinib was the most effective agent in inhibiting Src TK. Consistently, a pronounced anti-proliferative effect was achieved with saracatinib, in EGFR mutant cells, or with dasatinib, in wt EGFR/Ras mutant cells, poorly dependent on EGFR and erlotinib-resistant. We then identified the most effective drug combinations to overcome resistance to EGFR inhibitors, both in vitro and in nude mice: in T790M EGFR erlotinib-resistant cells, saracatinib with the anti-EGFR mAb cetuximab; in Ras mutant erlotinib-resistant models, dasatinib with the MEK inhibitor selumetinib. Src inhibitors may act with different mechanisms in NSCLCs, depending on EGFR/Ras mutational profile, and may be integrated with EGFR or MEK inhibitors for different cohorts of NSCLCs.
36. Engineered metal based nanoparticles and innate immunity
- Author
-
Sara Cortese, Valentina Amato, Giovanni Bernardini, Roberto Paganelli, Ivo Iavicoli, Enrico Sabbioni, Takemi Otsuki, Qiao Niu, Emanuela Clemente, Claudia Petrarca, Mario Di Gioacchino, Paola Pedata, Petrarca, Claudia, Clemente, Emanuela, Amato, Valentina, Pedata, Paola, Sabbioni, Enrico, Bernardini, Giovanni, Iavicoli, Ivo, Cortese, Sara, Niu, Qiao, Otsuki, Takemi, Paganelli, Roberto, and Di Gioacchino, Mario
- Subjects
Barrier ,Cell receptor ,medicine.medical_treatment ,Immunology ,Review ,medicine.disease_cause ,Autoimmunity ,Immune system ,Immunity ,medicine ,Immunotoxicity ,Immunology and Allergy ,Cytokine ,Molecular Biology ,Adjuvant ,Cell receptors ,Innate immune system ,business.industry ,In vitro toxicology ,Barriers ,Cytokines ,Immunostimulation ,business ,Foreign substance - Abstract
Almost all people in developed countries are exposed to metal nanoparticles (MeNPs) that are used in a large number of applications including medical (for diagnostic and therapeutic purposes). Once inside the body, absorbed by inhalation, contact, ingestion and injection, MeNPs can translocate to tissues and, as any foreign substance, are likely to encounter the innate immunity system that represent a non-specific first line of defense against potential threats to the host. In this review, we will discuss the possible effects of MeNPs on various components of the innate immunity (both specific cells and barriers). Most important is that there are no reports of immune diseases induced by MeNPs exposure: we are operating in a safe area. However, in vitro assays show that MeNPs have some effects on innate immunity, the main being toxicity (both cyto- and genotoxicity) and interference with the activity of various cells through modification of membrane receptors, gene expression and cytokine production. Such effects can have both negative and positive relevant impacts on humans. On the one hand, people exposed to high levels of MeNPs, as workers of industries producing or applying MeNPs, should be monitored for possible health effects. On the other hand, understanding the modality of the effects on immune responses is essential to develop medical applications for MeNPs. Indeed, those MeNPs that are able to stimulate immune cells could be used to develop of new vaccines, promote immunity against tumors and suppress autoimmunity.
- Full Text
- View/download PDF
37. Everolimus induces Met inactivation by disrupting the FKBP12/Met complex
- Author
-
Roberta De Rosa, Nunzia Montuori, Concetta Di Mauro, Roberta Marciano, Agostino Bruno, Sandro Cosconati, Valentina D’Amato, Filomena Napolitano, Antonio Randazzo, Ana Paula De Maio, Sabino De Placido, Alberto Servetto, Priscilla Cascetta, Roberto Bianco, Luigi Formisano, Maria Flavia Di Renzo, Paola Ciciola, Lucia Raimondo, Bianca Maria Veneziani, Roberta Clara Orsini, Raimondo, Lucia, D'Amato, Valentina, Servetto, Alberto, Rosa, Roberta, Marciano, Roberta, Formisano, Luigi, DI MAURO, Concetta, Orsini, Roberta Clara, Cascetta, Priscilla, Ciciola, Paola, DE MAIO, ANA PAULA, di Renzo, Maria Flavia, Cosconati, Sandro, Bruno, Agostino, Randazzo, Antonio, Napolitano, Filomena, Montuori, Nunzia, Veneziani, BIANCA MARIA, DE PLACIDO, Sabino, Bianco, Roberto, Di Mauro, Concetta, Clara Orsini, Roberta, De Maio, Ana Paula, Di Renzo, Maria Flavia, Veneziani, Bianca Maria, and De Placido, Sabino
- Subjects
0301 basic medicine ,Mice, Nude ,Pharmacology ,Everolimus resistance ,Mice ,03 medical and health sciences ,0302 clinical medicine ,FKBP12 ,Met ,everolimus ,everolimus resistance ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Phosphorylation ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Everolimus ,biology ,business.industry ,TOR Serine-Threonine Kinases ,Receptor Protein-Tyrosine Kinases ,Cancer ,HCT116 Cells ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Everolimu ,030104 developmental biology ,FKBP ,Mechanism of action ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Female ,RNA Interference ,medicine.symptom ,business ,Allosteric Site ,Neoplasm Transplantation ,Research Paper ,medicine.drug - Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) is a promising treatment strategy for several cancer types. Rapamycin derivatives such as everolimus are allosteric mTOR inhibitors acting through interaction with the intracellular immunophilin FKBP12, a prolyl isomerase with different cellular functions. Although mTOR inhibitors have significantly improved survival of different cancer patients, resistance and lack of predictive factors of response remain unsolved issues. To elucidate the mechanisms of resistance to everolimus, we evaluated Met activation in everolimus-sensitive/resistant human cancer cells, in vitro and in vivo. Biochemical and computational analyses were performed. Everolimus-resistant cells were xenografted into mice (10/group) and studied for their response to everolimus and Met inhibitors. The statistical significance of the in vitro results was evaluated by Student's t test. Everolimus reduced Met phosphorylation in everolimus-sensitive cells. This event was mediated by the formation of a Met-FKBP12 complex, which in turn is disrupted by everolimus. Aberrant Met activation in everolimus-resistant cells and overexpression of wild-type/mutant Met caused everolimus resistance. Pharmacological inhibition and RNA silencing of Met are effective in condition of everolimus resistance (P
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.