scispace - formally typeset
Search or ask a question

Showing papers by "Luis M. Montuenga published in 2020"


Journal ArticleDOI
John G. Edwards1, Kari Chansky2, Paul Van Schil, Andrew G. Nicholson3, Souheil Boubia, Elisabeth Brambilla4, Jessica S. Donington5, Françoise Galateau-Sallé, Hans Hoffmann6, Maurizio Infante, Mirella Marino, Edith M. Marom7, Jun Nakajima8, Marcin Ostrowski9, William D. Travis10, Ming-Sound Tsao11, Yasushi Yatabe, Dorothy J. Giroux2, Lynn Shemanski2, John Crowley2, Marc Krasnik, Hisao Asamura, Ramón Rami-Porta12, Valerie W. Rusch, Luiz H. Araujo, David G. Beer, Pietro Bertoglio, Ricardo Beyruti, Andrea Billè, Vanessa Bolejack, James D. Brierley, Ayten Kayi Cangir, David P. Carbone, Gail Darling, Frank C. Detterbeck, Xavier Benoit D’Journo, Jessica Donnington5, Wilfried Eberhardt, John Edwards, Jeremy J. Erasmus, Conrad Falkson, Wentao Fang, Dean A. Fennell, Kwun M. Fong, Oliver Gautschi, Ritu R. Gill, Dorothy Giroux2, Meredith Giuliani, Jin Mo Goo, Seiki Hasegawa, Fred R. Hirsch, Hans Hoffman6, Wayne L. Hofstetter, James Huang, Philippe Joubert, Kemp H. Kernstine, Keith M. Kerr, Young Tae Kim, Hong Kwan Kim, Hedy L. Kindler, Yolande Lievens, Hui Liu, Donald E. Low, Gustavo Lyons, Heber MacMahon, Edith Marom7, José-María Matilla, Jan P. van Meerbeeck, Luis M. Montuenga, Andrew G. Nicholson3, Katie Nishimura, Anna K. Nowak, Isabelle Opitz, Meinoshin Okumura, Raymond U. Osarogiagbon, Harvey I. Pass, Marc de Perrot, Helmut Prosch, David C. Rice, Andreas Rimner, Enrico Ruffini, Shuji Sakai, Navneet Singh, Amy Stoll-D’Astice, Francisco Su´rez, Ricardo Mingarini Terra, Ming S. Tsao11, Paula A. Ugalde, David A. Waller, Shun-ichi Watanabe, Jacinta Wiens, Ignacio I. Wistuba, Liyan Jiang, Kaoru Kubota, Akif Turna, Benny Weksler, Maria Teresa Tzukazan, Martin C. Tammemägi, Charles A. Powell, David P. Naidich, Hongxu Liu, Samuel G. Armato, Alex Brunelli, Giuseppe Cardillo, Elizabeth A. David, Brigitte Fournier, Mark Krasnik, Kauro Kubota, Catherine Labbé, Eric Lim, Paul Martin Putora, Gaetano Rocco, Pier Luigi Filosso, Kazuya Kondo, Dong Kwan Kim, Giuseppe Giaccone, Marco Lucchi, Thomas W. Rice, Mark K. Ferguson, Prasad Adsusmilli, William D. Travis10, Francisco Suárez, Kaura Kubota, Watanabe Hisao Asamura Shun-ichi, Rami-Porta Edith Marom Ramón, M. Tsao11, Watanabe Ming Tsao Shun-ichi, Meredith Guiliani, James Brierley, Ricardo R. Terra, Ray Osarogiagbon, Luis M. Montuenga, Hong Wei Wang, Françoise Galateau, Jim Mo Goo, Bill Travis, José María Matilla, Carolle St. Pierre, Ma Teresa Tzukazan, Nicholas Girard, Andreas Rimmer, Prasad S. Adusumilli, Xavier D’Journo, Donald E. Low, Adam Rosenthal 
TL;DR: R descriptors have prognostic relevance with R(un) survival stratifying between R0 and R1, and a detailed evaluation of R factor is of particular importance in the design and analyses of clinical trials of adjuvant therapies.

77 citations


Journal ArticleDOI
13 Jan 2020
TL;DR: Using three syngeneic models, it is demonstrated that short-term starvation synergizes with PD-1 blockade to inhibit lung cancer progression and metastasis and strongly support the clinical evaluation of IGF-1 modulators in combination with PD -1 blockade.
Abstract: Harnessing the immune system by blocking the programmed cell death protein 1 (PD-1) pathway has been a major breakthrough in non-small-cell lung cancer treatment. Nonetheless, many patients fail to respond to PD-1 inhibition. Using three syngeneic models, we demonstrate that short-term starvation synergizes with PD-1 blockade to inhibit lung cancer progression and metastasis. This antitumor activity was linked to a reduction in circulating insulin-like growth factor 1 (IGF-1) and a downregulation of IGF-1 receptor (IGF-1R) signaling in tumor cells. A combined inhibition of IGF-1R and PD-1 synergistically reduced tumor growth in mice. This effect required CD8 cells, boosted the intratumoral CD8/Treg ratio and led to the development of tumor-specific immunity. In patients with non-small-cell lung cancer, high plasma levels of IGF-1 or high IGF-1R expression in tumors was associated with resistance to anti-PD-1–programmed death-ligand 1 immunotherapy. In conclusion, our data strongly support the clinical evaluation of IGF-1 modulators in combination with PD-1 blockade. Ajona and colleagues report that short-term starvation synergizes with anti-PD-1 blockade to reduce lung tumor growth and metastasis. This antitumor effect is mediated through the reduction of plasma IGF-1 levels and IGF-1R levels on tumor cells.

46 citations


Journal ArticleDOI
TL;DR: The pro-oncogenic lncRNA ALAL-1 is frequently amplified in lung adenocarcinomas and promotes the nuclearization of USP4, affecting TNFα, p53, NF-κB, and TGF-β1 pathways.
Abstract: Cancer is characterized by genomic instability leading to deletion or amplification of oncogenes or tumor suppressors. However, most of the altered regions are devoid of known cancer drivers. Here, we identify lncRNAs frequently lost or amplified in cancer. Among them, we found amplified lncRNA associated with lung cancer-1 (ALAL-1) as frequently amplified in lung adenocarcinomas. ALAL-1 is also overexpressed in additional tumor types, such as lung squamous carcinoma. The RNA product of ALAL-1 is able to promote the proliferation and tumorigenicity of lung cancer cells. ALAL-1 is a TNFα- and NF-κB-induced cytoplasmic lncRNA that specifically interacts with SART3, regulating the subcellular localization of the protein deubiquitinase USP4 and, in turn, its function in the cell. Interestingly, ALAL-1 expression inversely correlates with the immune infiltration of lung squamous tumors, while tumors with ALAL-1 amplification show lower infiltration of several types of immune cells. We have thus unveiled a pro-oncogenic lncRNA that mediates cancer immune evasion, pointing to a new target for immune potentiation.

36 citations


Journal ArticleDOI
TL;DR: Positive PD-L1 expression correlates with a high infiltration by TILs, explaining its association with better prognosis and the type of tumor microenvironment correlated with DSS.
Abstract: The importance of immune tumor microenvironment in the prognosis of patients with head and neck squamous carcinomas (HNSCC) is increasingly recognized. We analyzed the prognostic relevance of PD-L1 and PD-1 expressions in relation to the infiltration by CD8+ and FOXP3+ tumor-infiltrating lymphocytes (TILs). Samples from 372 surgically treated HPV-negative HNSCC patients were evaluated by immunohistochemistry for PD-L1 expression [both tumor proportion score (TPS) and combined proportion score (CPS)], PD-1 expression in immune cells, and density of infiltrating CD8+ and FOXP3+ TILs. PD-L1 expression and CD8+ TIL density were combined to establish the type of tumor microenvironment. 29.5% cases exhibited PD-L1 TPS positivity (≥ 1%), whereas PD-L1 CPS positivity (≥ 1%) was observed in 40% cases. 47.5% cases showed positive PD-1 expression (≥ 1%). PD-L1 and PD-1 positivity correlated with a high density of both CD8+ and FOXP3+ TILs. In univariate analysis, PD-L1 TPS positivity (P = 0.026), PD-L1 CPS positivity (P = 0.004), high density of CD8+ TIL (P = 0.001), and high density of FOXP3+ TIL (P = 0.004) were associated with a better disease-specific survival (DSS). However, in multivariate analysis, only high density of CD8+ TIL was associated with a better DSS (P = 0.002). The type of tumor microenvironment correlated with DSS (P = .008), with the better DSS observed in cases with type I (PD-L1 CPS positivity and high density of CD8+ TIL). High infiltration by CD8+ TIL is associated with better survival outcomes. Positive PD-L1 expression correlates with a high infiltration by TILs, explaining its association with better prognosis.

35 citations



Journal ArticleDOI
TL;DR: The results unveil that the histotype-specific regulation of tumor fibrosis in lung cancer is mediated through differential SMAD3 promoter methylation in TAFs, and provide new mechanistic insights on the selective poor response of SCC-TAFs to nintedanib.
Abstract: The tumor-promoting fibrotic stroma rich in tumor-associated fibroblasts (TAF) is drawing increased therapeutic attention. Intriguingly, a trial with the antifibrotic drug nintedanib in non-small cell lung cancer reported clinical benefits in adenocarcinoma (ADC) but not squamous cell carcinoma (SCC), even though the stroma is fibrotic in both histotypes. Likewise, we reported that nintedanib inhibited the tumor-promoting fibrotic phenotype of TAFs selectively in ADC. Here we show that tumor fibrosis is actually higher in ADC-TAFs than SCC-TAFs in vitro and patient samples. Mechanistically, the reduced fibrosis and nintedanib response of SCC-TAFs was associated with increased promoter methylation of the profibrotic TGFβ transcription factor SMAD3 compared with ADC-TAFs, which elicited a compensatory increase in TGFβ1/SMAD2 activation. Consistently, forcing global DNA demethylation of SCC-TAFs with 5-AZA rescued TGFβ1/SMAD3 activation, whereas genetic downregulation of SMAD3 in ADC-TAFs and control fibroblasts increased TGFβ1/SMAD2 activation, and reduced their fibrotic phenotype and antitumor responses to nintedanib in vitro and in vivo. Our results also support that smoking and/or the anatomic location of SCC in the proximal airways, which are more exposed to cigarette smoke particles, may prime SCC-TAFs to stronger SMAD3 epigenetic repression, because cigarette smoke condensate selectively increased SMAD3 promoter methylation. Our results unveil that the histotype-specific regulation of tumor fibrosis in lung cancer is mediated through differential SMAD3 promoter methylation in TAFs and provide new mechanistic insights on the selective poor response of SCC-TAFs to nintedanib. Moreover, our findings support that patients with ADC may be more responsive to antifibrotic drugs targeting their stromal TGFβ1/SMAD3 activation. SIGNIFICANCE: This study implicates the selective epigenetic repression of SMAD3 in SCC-TAFs in the clinical failure of nintedanib in SCC and supports that patients with ADC may benefit from antifibrotic drugs targeting stromal TGFβ1/SMAD3.

22 citations


Journal ArticleDOI
14 Sep 2020
TL;DR: This paper sets out the potential of biomarker testing, the unfolding precision and range of possible diagnosis and prediction, and the many obstacles to adoption, and urges a European evolution towards an initial minimum testing scenario.
Abstract: Rapid and continuing advances in biomarker testing are not being matched by take-up in health systems, and this is hampering both patient care and innovation. It also risks costing health systems the opportunity to make their services more efficient and, over time, more economical. This paper sets out the potential of biomarker testing, the unfolding precision and range of possible diagnosis and prediction, and the many obstacles to adoption. It offers case studies of biomarker testing in breast, ovarian, prostate, lung, thyroid and colon cancers, and derives specific lessons as to the potential and actual use of each of them. It also draws lessons about how to improve access and alignment, and to remedy the data deficiencies that impede development. And it suggests solutions to outstanding issues - notably including funding and the tangled web of obtaining reimbursement or equivalent coverage that Europe's fragmented health system implies. It urges a European evolution towards an initial minimum testing scenario, which would guarantee universal access to a suite of biomarker tests for the currently most common conditions, and, further into the future, to an optimum testing scenario in which a much wider range of biomarker tests would be introduced and become part of a more sophisticated health system articulated around personalised medicine. For exploiting genomics to the full, it argues the need for a new policy framework for Europe. Biomarker testing is not an issue that can be treated in isolation, since the purpose of testing is to improve health. Its use is therefore always closely linked to specific health challenges and needs to be viewed in the broader policy context in the EU and more widely. The paper is the result of extensive engagement with experts and decision makers to develop the framework, and consequently represents a wide consensus of views on how healthcare systems should respond from push and pull factors at local, national and cross-border and EU level. It contains strong views and clear recommendations springing from the convictions of patients, clinicians, academics, medicines authorities, HTA bodies, payers, the diagnostic, pharmaceutical and ICT industries, and national policy makers.

18 citations


Journal ArticleDOI
TL;DR: Data show that the determination of the expression of FGFR1 or FGFR4 alone is not sufficient to predict anti-FGFR therapy efficacy; complementary determination of N-cadherin expression may further optimise patient selection for this therapeutic strategy.

15 citations


Posted Content
18 Aug 2020
TL;DR: The main issues and proposals that emerged during the previous advisory boards organised by the European Alliance for Personalized Medicine which mainly focus on possible scenarios of harmonisation of both oncogenetic testing and management of cancer patients are shown.
Abstract: Simple Summary The increasing number of data supporting use of a personalized approach in cancer treatment, is changing the path of patient’s management. In the same time, the availability of technologies should allow patients to receive the best test for the specific individual condition. This is theoretically true, when a specific test is designed for the specific disease condition, while it is difficult to implement in the setting of agnostic therapies. Financial sources availability related to the non homogeneous health systems working in the different countries do not allow for an immediate implementation of the technologies and test commercially available. Future perspectives for targeted oncology include tumor-agnostic drugs, which target a given mutation and could be used in treating cancers from multiple organ types. Therefore, the present paper is aimed to both underline a how much important is this new view and also to sensitize the international bodies that supervise health policies at the decision-making level, with the aim of harmonizing cancer treatment pathways in at least all European countries. Abstract Rapid and continuing advances in biomarker testing are not being matched by uptake in health systems, and this is hampering both patient care and innovation. It also risks costing health systems the opportunity to make their services more efficient and, over time, more economical. The potential that genomics has brought to biomarker testing in diagnosis, prediction and research is being realised, pre-eminently in many cancers, but also in an ever-wider range of conditions—notably BRCA1/2 testing in ovarian, breast, pancreatic and prostate cancers. Nevertheless, the implementation of genetic testing in clinical routine setting is still challenging. Development is impeded by country-related heterogeneity, data deficiencies, and lack of policy alignment on standards, approval—and the role of real-world evidence in the process—and reimbursement. The acute nature of the problem is compellingly illustrated by the particular challenges facing the development and use of tumour agnostic therapies, where the gaps in preparedness for taking advantage of this innovative approach to cancer therapy are sharply exposed. Europe should already have in place a guarantee of universal access to a minimum suite of biomarker tests and should be planning for an optimum testing scenario with a wider range of biomarker tests integrated into a more sophisticated health system articulated around personalised medicine. Improving healthcare and winning advantages for Europe’s industrial competitiveness and innovation require an appropriate policy framework—starting with an update to outdated recommendations. We show herein the main issues and proposals that emerged during the previous advisory boards organised by the European Alliance for Personalized Medicine which mainly focus on possible scenarios of harmonisation of both oncogenetic testing and management of cancer patients.

11 citations


Journal ArticleDOI
07 Jul 2020-Cancers
TL;DR: A large-scale bioinformatics tool is developed and validated, which exploits loss-of-function experiments to model the sensitivity of 6237 inhibitors and predict their corresponding biomarkers of sensitivity in 30 tumor types, and suggests a remarkable vulnerability to polo-like kinase 1 (PLK1) inhibition in CREBBP-mutant SCLC cells.
Abstract: The development of predictive biomarkers of response to targeted therapies is an unmet clinical need for many antitumoral agents. Recent genome-wide loss-of-function screens, such as RNA interference (RNAi) and CRISPR-Cas9 libraries, are an unprecedented resource to identify novel drug targets, reposition drugs and associate predictive biomarkers in the context of precision oncology. In this work, we have developed and validated a large-scale bioinformatics tool named DrugSniper, which exploits loss-of-function experiments to model the sensitivity of 6237 inhibitors and predict their corresponding biomarkers of sensitivity in 30 tumor types. Applying DrugSniper to small cell lung cancer (SCLC), we identified genes extensively explored in SCLC, such as Aurora kinases or epigenetic agents. Interestingly, the analysis suggested a remarkable vulnerability to polo-like kinase 1 (PLK1) inhibition in CREBBP-mutant SCLC cells. We validated this association in vitro using four mutated and four wild-type SCLC cell lines and two PLK1 inhibitors (Volasertib and BI2536), confirming that the effect of PLK1 inhibitors depended on the mutational status of CREBBP. Besides, DrugSniper was validated in-silico with several known clinically-used treatments, including the sensitivity of Tyrosine Kinase Inhibitors (TKIs) and Vemurafenib to FLT3 and BRAF mutant cells, respectively. These findings show the potential of genome-wide loss-of-function screens to identify new personalized therapeutic hypotheses in SCLC and potentially in other tumors, which is a valuable starting point for further drug development and drug repositioning projects.

7 citations


Journal ArticleDOI
10 Dec 2020-Cancers
TL;DR: This work combined genomic, transcriptomic, and proteomic approaches to study the mutational status and the expression levels of the SWI/SNF subunits in a panel of 38 lung adenocarcinoma (LUAD) cell lines, finding that theSWI/ SNF complex was mutated in more than 76% of the authors' LUAD cell lines and there was a high variability in the expression of the different SWI-SNFSubunits.
Abstract: Mammalian SWI/SNF (SWitch/Sucrose Non-Fermentable) complexes are ATP-dependent chromatin remodelers whose subunits have emerged among the most frequently mutated genes in cancer. Studying SWI/SNF function in cancer cell line models has unveiled vulnerabilities in SWI/SNF-mutant tumors that can lead to the discovery of new therapeutic drugs. However, choosing an appropriate cancer cell line model for SWI/SNF functional studies can be challenging because SWI/SNF subunits are frequently altered in cancer by various mechanisms, including genetic alterations and post-transcriptional mechanisms. In this work, we combined genomic, transcriptomic, and proteomic approaches to study the mutational status and the expression levels of the SWI/SNF subunits in a panel of 38 lung adenocarcinoma (LUAD) cell lines. We found that the SWI/SNF complex was mutated in more than 76% of our LUAD cell lines and there was a high variability in the expression of the different SWI/SNF subunits. These results underline the importance of the SWI/SNF complex as a tumor suppressor in LUAD and the difficulties in defining altered and unaltered cell models for the SWI/SNF complex. These findings will assist researchers in choosing the most suitable cellular models for their studies of SWI/SNF to bring all of its potential to the development of novel therapeutic applications.

Posted ContentDOI
14 Aug 2020
TL;DR: This competition aims to provide a platform for the development of a new generation of coaches and trainers for the management of youth sports.
Abstract: Denis Horgan, Gennaro Ciliberto, Pierfranco Conte, Giuseppe Curigliano , Luis Seijo, Luis M. Montuenga, Luis Paz-Ares, Marina Garassino, Frederique Penault-Llorca, Fabrizia Galli, Isabelle Ray-Coquard, Denis Querleu, David Baldwin, Susana Banerjee, Peter Riegman, Keith Kerr, Benjamin Horbach, Reinhard Buttner, Hein Van Poppel, Anders Bjartell, Giovanni Codacci-Pisanelli, Benedikt Westphalen, Fabien Calvo, Jasmina Koeva-Balabanova, Stephen Hall, Angelo Paradiso, Dipak Kalra, Christa Cobbaert, Rocio Varea Menendez, Zorana Maravic, Vassiliki Fotaki, Jaafar Bennouna, Estelle Cauchin, Nuria Malats, Iñaki Gutiérrez-Ibarluzea , Benjamin Gannon, Ken Mastris, Chiara Bernini, William Gallagher, Simonetta Buglioni, Alastair Kent, Elisabetta Munzone, Ivica Belina, Jan Van Meerbeeck, Michael Duffy, Elżbietę Sarnowską, Beata Jagielska, Sarah Mee, Ettore Capoluongo