59 results on '"Álvaro Quintanal-Villalonga"'
Search Results
2. SMARCA4 controls state plasticity in small cell lung cancer through regulation of neuroendocrine transcription factors and REST splicing
- Author
-
Esther Redin, Harsha Sridhar, Yingqian A. Zhan, Barbara Pereira Mello, Hong Zhong, Vidushi Durani, Amin Sabet, Parvathy Manoj, Irina Linkov, Juan Qiu, Richard P. Koche, Elisa de Stanchina, Maider Astorkia, Doron Betel, Álvaro Quintanal-Villalonga, and Charles M. Rudin
- Subjects
Lung Cancer ,SCLC ,Plasticity ,Epigenetics ,Targeted therapies ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Introduction Small Cell Lung Cancer (SCLC) can be classified into transcriptional subtypes with distinct degrees of neuroendocrine (NE) differentiation. Recent evidence supports plasticity among subtypes with a bias toward adoption of low-NE states during disease progression or upon acquired chemotherapy resistance. Here, we identify a role for SMARCA4, the catalytic subunit of the SWI/SNF complex, as a regulator of subtype shift in SCLC. Methods ATACseq and RNAseq experiments were performed in SCLC cells after pharmacological inhibition of SMARCA4. DNA binding of SMARCA4 was characterized by ChIPseq in high-NE SCLC patient derived xenografts (PDXs). Enrichment analyses were applied to transcriptomic data. Combination of FHD-286 and afatinib was tested in vitro and in a set of chemo-resistant SCLC PDXs in vivo. Results SMARCA4 expression positively correlates with that of NE genes in both SCLC cell lines and patient tumors. Pharmacological inhibition of SMARCA4 with FHD-286 induces the loss of NE features and downregulates neuroendocrine and neuronal signaling pathways while activating non-NE factors. SMARCA4 binds to gene loci encoding NE-lineage transcription factors ASCL1 and NEUROD1 and alters chromatin accessibility, enhancing NE programs. Enrichment analysis applied to high-confidence SMARCA4 targets confirmed neuron related pathways as the top GO Biological processes regulated by SMARCA4 in SCLC. In parallel, SMARCA4 also controls REST, a known suppressor of the NE phenotype, by regulating SRRM4-dependent REST transcript splicing. Furthermore, SMARCA4 inhibition drives ERBB pathway activation in SCLC, rendering SCLC tumors sensitive to afatinib. Conclusions This study nominates SMARCA4 as a key regulator of the NE state plasticity and defines a novel therapeutic strategy for SCLC.
- Published
- 2024
- Full Text
- View/download PDF
3. Protocol to dissociate, process, and analyze the human lung tissue using single-cell RNA-seq
- Author
-
Álvaro Quintanal-Villalonga, Joseph M. Chan, Ignas Masilionis, Vianne Ran Gao, Yubin Xie, Viola Allaj, Andrew Chow, John T. Poirier, Dana Pe’er, Charles M. Rudin, and Linas Mazutis
- Subjects
Cancer ,Cell isolation ,Flow cytometry/Mass cytometry ,Molecular biology ,RNAseq ,Sequence analysis ,Science (General) ,Q1-390 - Abstract
Summary: We report a protocol for obtaining high-quality single-cell transcriptomics data from human lung biospecimens acquired from core needle biopsies, fine-needle aspirates, surgical resection, and pleural effusions. The protocol relies upon the brief mechanical and enzymatic disruption of tissue, enrichment of live cells by fluorescence-activated cell sorting (FACS), and droplet-based single-cell RNA sequencing (scRNA-seq). The protocol also details a procedure for analyzing the scRNA-seq data.For complete details on the use and execution of this protocol, please refer to Chan et al. (2021). : Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
- Published
- 2022
- Full Text
- View/download PDF
4. FGFR1 and FGFR4 oncogenicity depends on n-cadherin and their co-expression may predict FGFR-targeted therapy efficacy
- Author
-
Álvaro Quintanal-Villalonga, Irene Ferrer, Elizabeth Guruceaga, Cristina Cirauqui, Ángela Marrugal, Laura Ojeda, Santiago García, Jon Zugazagoitia, Sandra Muñoz-Galván, Fernando Lopez-Rios, Luis Montuenga, Silvestre Vicent, Sonia Molina-Pinelo, Amancio Carnero, and Luis Paz-Ares
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Fibroblast growth factor receptor (FGFR)1 and FGFR4 have been associated with tumorigenesis in a variety of tumour types. As a therapeutic approach, their inhibition has been attempted in different types of malignancies, including lung cancer, and was initially focused on FGFR1-amplified tumours, though with limited success. Methods: In vitro and in vivo functional assessments of the oncogenic potential of downregulated/overexpressed genes in isogenic cell lines were performed, as well as inhibitor efficacy tests in vitro and in vivo in patient-derived xenografts (PDXs). mRNA was extracted from FFPE non-small cell lung cancer samples to determine the prognostic potential of the genes under study. Findings: We provide in vitro and in vivo evidence showing that expression of the adhesion molecule N-cadherin is key for the oncogenic role of FGFR1/4 in non-small cell lung cancer. According to this, assessment of the expression of genes in different lung cancer patient cohorts showed that FGFR1 or FGFR4 expression alone showed no prognostic potential, and that only co-expression of FGFR1 and/or FGFR4 with N-cadherin inferred a poorer outcome. Treatment of high-FGFR1 and/or FGFR4-expressing lung cancer cell lines and patient-derived xenografts with selective FGFR inhibitors showed high efficacy, but only in models with high FGFR1/4 and N-cadherin expression. Interpretation: Our 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. Keywords: FGFR1, FGFR4, N-cadherin, Predictive biomarker, FGFR inhibitors
- Published
- 2020
- Full Text
- View/download PDF
5. MAP17 predicts sensitivity to platinum-based therapy, EGFR inhibitors and the proteasome inhibitor bortezomib in lung adenocarcinoma
- Author
-
Irene Ferrer, Álvaro Quintanal-Villalonga, Sonia Molina-Pinelo, Jose Manuel Garcia-Heredia, Marco Perez, Rocío Suárez, Santiago Ponce-Aix, Luis Paz-Ares, and Amancio Carnero
- Subjects
Biomarkers ,Lung cancer ,PDZK1IP1 ,Treatment efficacy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The high incidence and mortality of lung tumours is a major health problem. Therefore, the identification both of biomarkers predicting efficacy for therapies in use and of novel efficacious therapeutic agents is crucial to increase patient survival. MAP17 (PDZK1IP1) is a small membrane-bound protein whose upregulation is reported as a common feature in tumours from diverse histological origins. Furthermore, MAP17 is correlated with tumour progression. Methods We assessed the expression of MAP17 in preclinical models, including cell lines and patient-derived xenografts (PDXs), assessing its correlation with sensitivity to different standard-of-care drugs in lung adenocarcinoma, as well as novel drugs. At the clinical level, we subsequently correlated MAP17 expression in human tumours with patient response to these therapies. Results We show that MAP17 expression is induced during lung tumourigenesis, particularly in lung adenocarcinomas, and provide in vitro and in vivo evidence that MAP17 levels predict sensitivity to therapies currently under clinical use in adenocarcinoma tumours, including cisplatin, carboplatin and EGFR inhibitors. In addition, we show that MAP17 expression predicts proteasome inhibitor efficacy in this context and that bortezomib, an FDA-approved drug, may be a novel therapeutic approach for MAP17-overexpressing lung adenocarcinomas. Conclusions Our results indicate a potential prognostic role for MAP17 in lung tumours, with particular relevance in lung adenocarcinomas, and highlight the predictive pot0065ntial of this membrane-associated protein for platinum-based therapy and EGFR inhibitor efficacy. Furthermore, we propose bortezomib treatment as a novel and efficacious therapy for lung adenocarcinomas exhibiting high MAP17 expression.
- Published
- 2018
- Full Text
- View/download PDF
6. Impact of Heat Shock Protein 90 Inhibition on the Proteomic Profile of Lung Adenocarcinoma as Measured by Two-Dimensional Electrophoresis Coupled with Mass Spectrometry
- Author
-
Ángela Marrugal, Irene Ferrer, Maria Dolores Pastor, Laura Ojeda, Álvaro Quintanal-Villalonga, Amancio Carnero, Sonia Molina-Pinelo, and Luis Paz-Ares
- Subjects
lung cancer ,proteomic ,chaperones ,HSP90 inhibitors ,Cytology ,QH573-671 - Abstract
Heat shock protein 90 (HSP90) is an important chaperone in lung adenocarcinoma, with relevant protein drivers such as EGFR (epidermal growth factor receptor) and EML4-ALK (echinoderm microtubule-associated protein-like protein4 fused to anaplastic lymphoma kinase) depending on it for their correct function, therefore HSP90 inhibitors show promise as potential treatments for lung adenocarcinoma. To study responses to its inhibition, HSP90 was pharmacologically interrupted by geldanamycin and resorcinol derivatives or with combined inhibition of HSP90 plus HSP70 in lung adenocarcinoma cell lines. Two-dimensional electrophoresis was performed to identify proteomic profiles associated with inhibition which will help to understand the biological basis for the responses. HSP90 inhibition resulted in altered protein profiles that differed according the treatment condition studied. Results revealed 254 differentially expressed proteins after treatments, among which, eukaryotic translation initiation factor3 subunit I (eIF3i) and citrate synthase demonstrated their potential role as response biomarkers. The differentially expressed proteins also enabled signalling pathways involved in responses to be identified; these included apoptosis, serine-glycine biosynthesis and tricarboxylic acid cycle. The proteomic profiles identified here contribute to an improved understanding of HSP90 inhibition and open possibilities for the detection of potential response biomarkers which will be essential to maximize treatment efficacy in lung adenocarcinoma.
- Published
- 2019
- Full Text
- View/download PDF
7. Combination therapy with MDM2 and MEK inhibitors is effective in patient-derived models of lung adenocarcinoma with concurrent oncogenic drivers and MDM2 amplification
- Author
-
Arielle Elkrief, Igor Odintsov, Vladimir Markov, Rebecca Caeser, Pawel Sobczuk, Sam E. Tischfield, Umesh Bhanot, Chad M. Vanderbilt, Emily Cheng, Alexander Drilon, Gregory J. Riely, William W. Lockwood, Elisa de Stanchina, Vijaya G. Tirunagaru, Robert C. Doebele, Álvaro Quintanal-Villalonga, Charles M. Rudin, Romel Somwar, and Marc Ladanyi
- Subjects
Pulmonary and Respiratory Medicine ,Oncology - Published
- 2023
- Full Text
- View/download PDF
8. Inhibition of XPO1 Sensitizes Small Cell Lung Cancer to First- and Second-Line Chemotherapy
- Author
-
J. Qiu, Umesh Bhanot, John T. Poirier, Yingqian Zhan, Elisa de Stanchina, Viola Allaj, Hirokazu Taniguchi, Fathema Uddin, Travis J. Hollmann, Jordana Ray-Kirton, Metamia Ciampricotti, Andrew Chow, Marina Asher, Richard Koche, Álvaro Quintanal-Villalonga, Jacklynn V. Egger, Yuan Hao, Joseph M. Chan, Shweta S Chavan, Charles M. Rudin, Michael H.A. Roehrl, Triparna Sen, Michael Offin, Irina Linkov, Nisargbhai S. Shah, and P. Manoj
- Subjects
Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,Receptors, Cytoplasmic and Nuclear ,Karyopherins ,Malignancy ,Article ,Metastasis ,Mice ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Lung cancer ,Cisplatin ,Chemotherapy ,business.industry ,Immunotherapy ,medicine.disease ,Small Cell Lung Carcinoma ,respiratory tract diseases ,Irinotecan ,Oncology ,Cancer research ,business ,Progressive disease ,medicine.drug - Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy characterized by early metastasis and extreme lethality. The backbone of SCLC treatment over the past several decades has been platinum-based doublet chemotherapy, with the recent addition of immunotherapy providing modest benefits in a subset of patients. However, nearly all patients treated with systemic therapy quickly develop resistant disease, and there is an absence of effective therapies for recurrent and progressive disease. Here we conducted CRISPR-Cas9 screens using a druggable genome library in multiple SCLC cell lines representing distinct molecular subtypes. This screen nominated exportin-1, encoded by XPO1, as a therapeutic target. XPO1 was highly and ubiquitously expressed in SCLC relative to other lung cancer histologies and other tumor types. XPO1 knockout enhanced chemosensitivity, and exportin-1 inhibition demonstrated synergy with both first- and second-line chemotherapy. The small molecule exportin-1 inhibitor selinexor in combination with cisplatin or irinotecan dramatically inhibited tumor growth in chemonaïve and chemorelapsed SCLC patient-derived xenografts, respectively. Together these data identify exportin-1 as a promising therapeutic target in SCLC, with the potential to markedly augment the efficacy of cytotoxic agents commonly used in treating this disease. Significance: CRISPR-Cas9 screening nominates exportin-1 as a therapeutic target in SCLC, and exportin-1 inhibition enhances chemotherapy efficacy in patient-derived xenografts, providing a novel therapeutic opportunity in this disease.
- Published
- 2022
- Full Text
- View/download PDF
9. Multiomic Analysis of Lung Tumors Defines Pathways Activated in Neuroendocrine Transformation
- Author
-
Yingqian Zhan, Jason C. Chang, Irina Linkov, Shweta S Chavan, Travis J. Hollmann, Jordana Ray-Kirton, Joseph M. Chan, Metamia Ciampricotti, Triparna Sen, Sam E. Tischfield, J. Qiu, Maysun Hasan, Helena A. Yu, Joachim Silber, Richard Koche, Helen Won, Marina K. Baine, Brian Loomis, Umesh Bhanot, Charles M. Rudin, Natasha Rekhtman, Andrew Chow, Fathema Uddin, Jacklynn V. Egger, Álvaro Quintanal-Villalonga, Sonali Sinha, Fanli Meng, Michael H.A. Roehrl, Christine A. Iacobuzio-Donahue, P. Manoj, Michael Offin, John T. Poirier, Marina Asher, Elisa de Stanchina, Mark T.A. Donoghue, and Hirokazu Taniguchi
- Subjects
medicine.medical_treatment ,Biology ,medicine.disease ,Targeted therapy ,Transcriptome ,Transformation (genetics) ,Oncology ,medicine ,Cancer research ,Lung cancer ,Protein kinase B ,Reprogramming ,PI3K/AKT/mTOR pathway ,Epigenomics - Abstract
Lineage plasticity is implicated in treatment resistance in multiple cancers. In lung adenocarcinomas (LUAD) amenable to targeted therapy, transformation to small cell lung cancer (SCLC) is a recognized resistance mechanism. Defining molecular mechanisms of neuroendocrine (NE) transformation in lung cancer has been limited by a paucity of pre/posttransformation clinical samples. Detailed genomic, epigenomic, transcriptomic, and protein characterization of combined LUAD/SCLC tumors, as well as pre/posttransformation samples, supports that NE transformation is primarily driven by transcriptional reprogramming rather than mutational events. We identify genomic contexts in which NE transformation is favored, including frequent loss of the 3p chromosome arm. We observed enhanced expression of genes involved in the PRC2 complex and PI3K/AKT and NOTCH pathways. Pharmacologic inhibition of the PI3K/AKT pathway delayed tumor growth and NE transformation in an EGFR-mutant patient-derived xenograft model. Our findings define a novel landscape of potential drivers and therapeutic vulnerabilities of NE transformation in lung cancer. Significance: The difficulty in collection of transformation samples has precluded the performance of molecular analyses, and thus little is known about the lineage plasticity mechanisms leading to LUAD-to-SCLC transformation. Here, we describe biological pathways dysregulated upon transformation and identify potential predictors and potential therapeutic vulnerabilities of NE transformation in the lung. See related commentary by Meador and Lovly, p. 2962. This article is highlighted in the In This Issue feature, p. 2945
- Published
- 2021
- Full Text
- View/download PDF
10. CDC7 inhibition impairs neuroendocrine transformation in lung and prostate tumors through MYC degradation
- Author
-
Alvaro Quintanal-Villalonga, Kenta Kawasaki, Esther Redin, Fathema Uddin, Swanand Rakhade, Vidushi Durani, Amin Sabet, Moniquetta Shafer, Wouter R. Karthaus, Samir Zaidi, Yingqian A. Zhan, Parvathy Manoj, Harsha Sridhar, Dennis Kinyua, Hong Zhong, Barbara P. Mello, Metamia Ciampricotti, Umesh K. Bhanot, Irina Linkov, Juan Qiu, Radhika A. Patel, Colm Morrissey, Sanjoy Mehta, Jesse Barnes, Michael C. Haffner, Nicholas D. Socci, Richard P. Koche, Elisa de Stanchina, Sonia Molina-Pinelo, Sohrab Salehi, Helena A. Yu, Joseph M. Chan, and Charles M. Rudin
- Subjects
Medicine ,Biology (General) ,QH301-705.5 - Abstract
Abstract Neuroendocrine (NE) transformation is a mechanism of resistance to targeted therapy in lung and prostate adenocarcinomas leading to poor prognosis. Up to date, even if patients at high risk of transformation can be identified by the occurrence of Tumor Protein P53 (TP53) and Retinoblastoma Transcriptional Corepressor 1 (RB1) mutations in their tumors, no therapeutic strategies are available to prevent or delay histological transformation. Upregulation of the cell cycle kinase Cell Division Cycle 7 (CDC7) occurred in tumors during the initial steps of NE transformation, already after TP53/RB1 co-inactivation, leading to induced sensitivity to the CDC7 inhibitor simurosertib. CDC7 inhibition suppressed NE transdifferentiation and extended response to targeted therapy in in vivo models of NE transformation by inducing the proteasome-mediated degradation of the MYC Proto-Oncogen (MYC), implicated in stemness and histological transformation. Ectopic overexpression of a degradation-resistant MYC isoform reestablished the NE transformation phenotype observed on targeted therapy, even in the presence of simurosertib. CDC7 inhibition also markedly extended response to standard cytotoxics (cisplatin, irinotecan) in lung and prostate small cell carcinoma models. These results nominate CDC7 inhibition as a therapeutic strategy to constrain lineage plasticity, as well as to effectively treat NE tumors de novo or after transformation. As simurosertib clinical efficacy trials are ongoing, this concept could be readily translated for patients at risk of transformation.
- Published
- 2024
- Full Text
- View/download PDF
11. Abstract 6127: MDM2 inhibition in combination with MEK inhibition in pre-clinical models of lung adenocarcinomas with MDM2 amplification
- Author
-
Arielle Elkrief, Vladimir Markov, Álvaro Quintanal-Villalonga, Rebecca Caeser, Pawel Sobczuk, Emily Cheng, Alexander Drilon, Gregory J. Riely, William W. Lockwood, Elisa de Stanchina, Charles M. Rudin, Igor Odintsov, and Romel Somwar
- Subjects
Cancer Research ,Oncology - Abstract
The eventual development of resistance to single-agent targeted therapies in lung adenocarcinomas (LUAD) is inevitable, and new strategies are needed. We hypothesize that combination therapies aimed at a known driver and a distinct targetable alteration could prolong time on oral targeted therapy. In an analysis of 7636 patients with LUAD who underwent MSK-IMPACT large panel NGS testing, 5.5% (416/7636) harbored MDM2 amplification (MDM2amp), a known mechanism of TP53 inactivation. MDM2amp was over-represented among tumors with alterations in METex14 (34.4%, p Citation Format: Arielle Elkrief, Vladimir Markov, Álvaro Quintanal-Villalonga, Rebecca Caeser, Pawel Sobczuk, Emily Cheng, Alexander Drilon, Gregory J. Riely, William W. Lockwood, Elisa de Stanchina, Charles M. Rudin, Igor Odintsov, Romel Somwar. MDM2 inhibition in combination with MEK inhibition in pre-clinical models of lung adenocarcinomas with MDM2 amplification [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6127.
- Published
- 2023
- Full Text
- View/download PDF
12. Abstract 5185: Intratumoral Escherichia is associated with response to single-agent immune checkpoint inhibition in patients with advanced non-small cell lung cancer
- Author
-
Arielle Elkrief, Anita S. Bowman, Ayyuce Begum Bektas, Wenfei Kang, Katia Manova-Todorova, Jacklynn V. Egger, Hira Rizvi, Daniel Kelly, Eric Chan, Eric Rosiek, Fan Ning, Gregory J. Riely, Álvaro Quintanal Villalonga, Snjezana Dogan, Umesh Bhanot, Mithat Gonen, Matthew D. Hellmann, Adam J. Schoenfeld, Charles M. Rudin, Marc Ladanyi, and Chad M. Vanderbilt
- Subjects
Cancer Research ,Oncology - Abstract
The impact of the intratumoral microbiome on immune checkpoint inhibitor (ICI) efficacy in patients (pts) with non-small cell lung cancer (NSCLC) is unknown. In preclinical studies, the presence of lung intratumoral Escherichia was associated with a proinflammatory tumor microenvironment and decreased metastases within lung tissue. We sought to detect intratumoral bacteria in pts with advanced NSCLC using hybrid capture-based, next generation sequencing (NGS). We studied 849 pts treated with ICI-based therapy who underwent NGS at our center. We extracted unmapped reads from BAM files, and these were queried for bacteria (blastn alignment using the NCBI database). Putative environmental contaminants were subtracted from the analysis using “no template” controls (n=2,539) to exclude possible artifactual false positives. A custom E.Coli fluorescence in situ hybridization (FISH) probe was used to visualize Escherichia within the tumors after co-registration with H&E. In 849 pts, a median of 30 bacterial reads was detected per sample (inter-quartile range (18-85)). Among 68 pts with paired primary/metastatic samples, the bacterial spectra were similar in both sites, suggesting that tumor resident bacteria might travel with cancer cells to distant sites. Antibiotic use within 30 days of tumor sampling was associated with decreased intratumoral bacterial diversity (p=0.023 by Inverse Simpson, p=0.038 by Shannon). Intratumoral Escherichia was associated with better PFS (HR 0.78, 95% CI 0.62-0.98, p=0.036), and OS (HR 0.74, 95% CI 0.58-0.95, p=0.017) in pts treated with single-agent ICI, but not combination Chemo/ICI. In a multivariable model adjusting for prognostic features in NSCLC including PD-L1 tumor proportion score, the presence of intratumoral Escherichia was associated with better PFS (p=0.040) and OS (p=0.045) upon single-agent ICI therapy. Escherichia appeared to be intracellular based on co-registration of FISH staining and serial H&E sections. These findings warrant further investigation of the possible inter-relationships between intratumoral Escherichia, tumor immune micro-environment, and ICI therapeutic outcomes. Citation Format: Arielle Elkrief, Anita S. Bowman, Ayyuce Begum Bektas, Wenfei Kang, Katia Manova-Todorova, Jacklynn V. Egger, Hira Rizvi, Daniel Kelly, Eric Chan, Eric Rosiek, Fan Ning, Gregory J. Riely, Álvaro Quintanal Villalonga, Snjezana Dogan, Umesh Bhanot, Mithat Gonen, Matthew D. Hellmann, Adam J. Schoenfeld, Charles M. Rudin, Marc Ladanyi, Chad M. Vanderbilt. Intratumoral Escherichia is associated with response to single-agent immune checkpoint inhibition in patients with advanced non-small cell lung cancer. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5185.
- Published
- 2023
- Full Text
- View/download PDF
13. A Six-Gene Prognostic and Predictive Radiotherapy-Based Signature for Early and Locally Advanced Stages in Non-Small-Cell Lung Cancer
- Author
-
Javier Peinado-Serrano, Álvaro Quintanal-Villalonga, Sandra Muñoz-Galvan, Eva M. Verdugo-Sivianes, Juan C. Mateos, María J. Ortiz-Gordillo, Amancio Carnero, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), European Commission, Centro de Investigación Biomédica en Red Cáncer (España), Junta de Andalucía, Fundación Científica Asociación Española Contra el Cáncer, and Asociación Española Contra el Cáncer
- Subjects
Cancer Research ,Oncology ,predictive signature ,biomarkers ,radiation oncology ,prognosis ,Predictive signature ,Prognosis ,NSCLC ,Radiation oncology ,Biomarkers - Abstract
[Simple Summary] The search for prognostic and/or predictive gene signatures of the response to radiotherapy treatment can significantly aid clinical decision making. These signatures can condition the fractionation, the total dose to be administered, and/or the combination of systemic treatments and radiation. The ultimate goal is to achieve better clinical results, as well as to minimize the adverse effects associated with current cancer therapies. To this end, we analyzed the intrinsic radiosensitivity of 15 NSCLC lines and found the differences in gene expression levels between radiosensitive and radioresistant lines, resulting in a potentially applicable six-gene signature in NSCLC patients. The six-gene signature had the ability to predict overall survival and progression-free survival (PFS), which could translate into a prediction of the response to the cancer treatment received., [Abstract] Non-small-cell lung cancer (NSCLC) is the leading cause of cancer death worldwide, generating an enormous economic and social impact that has not stopped growing in recent years. Cancer treatment for this neoplasm usually includes surgery, chemotherapy, molecular targeted treatments, and ionizing radiation. The prognosis in terms of overall survival (OS) and the disparate therapeutic responses among patients can be explained, to a great extent, by the existence of widely heterogeneous molecular profiles. The main objective of this study was to identify prognostic and predictive gene signatures of response to cancer treatment involving radiotherapy, which could help in making therapeutic decisions in patients with NSCLC. To achieve this, we took as a reference the differential gene expression pattern among commercial cell lines, differentiated by their response profile to ionizing radiation (radiosensitive versus radioresistant lines), and extrapolated these results to a cohort of 107 patients with NSCLC who had received radiotherapy (among other therapies). We obtained a six-gene signature (APOBEC3B, GOLM1, FAM117A, KCNQ1OT1, PCDHB2, and USP43) with the ability to predict overall survival and progression-free survival (PFS), which could translate into a prediction of the response to the cancer treatment received. Patients who had an unfavorable prognostic signature had a median OS of 24.13 months versus 71.47 months for those with a favorable signature, and the median PFS was 12.65 months versus 47.11 months, respectively. We also carried out a univariate analysis of multiple clinical and pathological variables and a bivariate analysis by Cox regression without any factors that substantially modified the HR value of the proposed gene signature., This work was supported by grants from the Ministerio de Ciencia, Innovación y Universidades (MCIU) Plan Estatal de I+D+I 2018, a la Agencia Estatal de Investigación (AEI) y al Fondo Europeo de Desarrollo Regional (MCIU/AEI/FEDER, UE): RTI2018-097455-B-I00; grant from AEI-MICIU/FEDER (RED2018-102723-T); from CIBER de Cáncer (CB16/12/00275), co-funded by FEDER from Regional Development European Funds (European Union); from Consejeria de Salud (PI-0397-2017) and Project P18-RT-2501 from 2018 competitive research projects call within the scope of PAIDI 2020 co-financed by the European Regional Development Fund (ERDF) from the Regional Ministry of Economic Transformation, Industry, Knowledge and Universities, Junta de Andalucía. Special thanks to the AECC (Spanish Association of Cancer Research) Founding Ref. GC16173720CARR and Fundacion Eugenio Rodriguez Pascual for supporting this work. E.S.-M. was funded by a fellowship from Junta de Andalucia. S.M.-G. was funded by Fundación AECC.
- Published
- 2022
- Full Text
- View/download PDF
14. The Human Tumor Atlas Network: Charting Tumor Transitions across Space and Time at Single-Cell Resolution
- Author
-
Orit Rozenblatt-Rosen, Aviv Regev, Philipp Oberdoerffer, Tal Nawy, Anna Hupalowska, Jennifer E. Rood, Orr Ashenberg, Ethan Cerami, Robert J. Coffey, Emek Demir, Li Ding, Edward D. Esplin, James M. Ford, Jeremy Goecks, Sharmistha Ghosh, Joe W. Gray, Justin Guinney, Sean E. Hanlon, Shannon K. Hughes, E. Shelley Hwang, Christine A. Iacobuzio-Donahue, Judit Jané-Valbuena, Bruce E. Johnson, Ken S. Lau, Tracy Lively, Sarah A. Mazzilli, Dana Pe’er, Sandro Santagata, Alex K. Shalek, Denis Schapiro, Michael P. Snyder, Peter K. Sorger, Avrum E. Spira, Sudhir Srivastava, Kai Tan, Robert B. West, Elizabeth H. Williams, Denise Aberle, Samuel I. Achilefu, Foluso O. Ademuyiwa, Andrew C. Adey, Rebecca L. Aft, Rachana Agarwal, Ruben A. Aguilar, Fatemeh Alikarami, Viola Allaj, Christopher Amos, Robert A. Anders, Michael R. Angelo, Kristen Anton, Jon C. Aster, Ozgun Babur, Amir Bahmani, Akshay Balsubramani, David Barrett, Jennifer Beane, Diane E. Bender, Kathrin Bernt, Lynne Berry, Courtney B. Betts, Julie Bletz, Katie Blise, Adrienne Boire, Genevieve Boland, Alexander Borowsky, Kristopher Bosse, Matthew Bott, Ed Boyden, James Brooks, Raphael Bueno, Erik A. Burlingame, Qiuyin Cai, Joshua Campbell, Wagma Caravan, Hassan Chaib, Joseph M. Chan, Young Hwan Chang, Deyali Chatterjee, Ojasvi Chaudhary, Alyce A. Chen, Bob Chen, Changya Chen, Chia-hui Chen, Feng Chen, Yu-An Chen, Milan G. Chheda, Koei Chin, Roxanne Chiu, Shih-Kai Chu, Rodrigo Chuaqui, Jaeyoung Chun, Luis Cisneros, Graham A. Colditz, Kristina Cole, Natalie Collins, Kevin Contrepois, Lisa M. Coussens, Allison L. Creason, Daniel Crichton, Christina Curtis, Tanja Davidsen, Sherri R. Davies, Ino de Bruijn, Laura Dellostritto, Angelo De Marzo, David G. DeNardo, Dinh Diep, Sharon Diskin, Xengie Doan, Julia Drewes, Stephen Dubinett, Michael Dyer, Jacklynn Egger, Jennifer Eng, Barbara Engelhardt, Graham Erwin, Laura Esserman, Alex Felmeister, Heidi S. Feiler, Ryan C. Fields, Stephen Fisher, Keith Flaherty, Jennifer Flournoy, Angelo Fortunato, Allison Frangieh, Jennifer L. Frye, Robert S. Fulton, Danielle Galipeau, Siting Gan, Jianjiong Gao, Long Gao, Peng Gao, Vianne R. Gao, Tim Geiger, Ajit George, Gad Getz, Marios Giannakis, David L. Gibbs, William E. Gillanders, Simon P. Goedegebuure, Alanna Gould, Kate Gowers, William Greenleaf, Jeremy Gresham, Jennifer L. Guerriero, Tuhin K. Guha, Alexander R. Guimaraes, David Gutman, Nir Hacohen, Sean Hanlon, Casey R. Hansen, Olivier Harismendy, Kathleen A. Harris, Aaron Hata, Akimasa Hayashi, Cody Heiser, Karla Helvie, John M. Herndon, Gilliam Hirst, Frank Hodi, Travis Hollmann, Aaron Horning, James J. Hsieh, Shannon Hughes, Won Jae Huh, Stephen Hunger, Shelley E. Hwang, Heba Ijaz, Benjamin Izar, Connor A. Jacobson, Samuel Janes, Reyka G. Jayasinghe, Lihua Jiang, Brett E. Johnson, Bruce Johnson, Tao Ju, Humam Kadara, Klaus Kaestner, Jacob Kagan, Lukas Kalinke, Robert Keith, Aziz Khan, Warren Kibbe, Albert H. Kim, Erika Kim, Junhyong Kim, Annette Kolodzie, Mateusz Kopytra, Eran Kotler, Robert Krueger, Kostyantyn Krysan, Anshul Kundaje, Uri Ladabaum, Blue B. Lake, Huy Lam, Rozelle Laquindanum, Ashley M. Laughney, Hayan Lee, Marc Lenburg, Carina Leonard, Ignaty Leshchiner, Rochelle Levy, Jerry Li, Christine G. Lian, Kian-Huat Lim, Jia-Ren Lin, Yiyun Lin, Qi Liu, Ruiyang Liu, William J.R. Longabaugh, Teri Longacre, Cynthia X. Ma, Mary Catherine Macedonia, Tyler Madison, Christopher A. Maher, Anirban Maitra, Netta Makinen, Danika Makowski, Carlo Maley, Zoltan Maliga, Diego Mallo, John Maris, Nick Markham, Jeffrey Marks, Daniel Martinez, Robert J. Mashl, Ignas Masilionais, Jennifer Mason, Joan Massagué, Pierre Massion, Marissa Mattar, Richard Mazurchuk, Linas Mazutis, Eliot T. McKinley, Joshua F. McMichael, Daniel Merrick, Matthew Meyerson, Julia R. Miessner, Gordon B. Mills, Meredith Mills, Suman B. Mondal, Motomi Mori, Yuriko Mori, Elizabeth Moses, Yael Mosse, Jeremy L. Muhlich, George F. Murphy, Nicholas E. Navin, Michel Nederlof, Reid Ness, Stephanie Nevins, Milen Nikolov, Ajit Johnson Nirmal, Garry Nolan, Edward Novikov, Brendan O’Connell, Michael Offin, Stephen T. Oh, Anastasiya Olson, Alex Ooms, Miguel Ossandon, Kouros Owzar, Swapnil Parmar, Tasleema Patel, Gary J. Patti, Itsik Pe'er, Tao Peng, Daniel Persson, Marvin Petty, Hanspeter Pfister, Kornelia Polyak, Kamyar Pourfarhangi, Sidharth V. Puram, Qi Qiu, Álvaro Quintanal-Villalonga, Arjun Raj, Marisol Ramirez-Solano, Rumana Rashid, Ashley N. Reeb, Mary Reid, Adam Resnick, Sheila M. Reynolds, Jessica L. Riesterer, Scott Rodig, Joseph T. Roland, Sonia Rosenfield, Asaf Rotem, Sudipta Roy, Charles M. Rudin, Marc D. Ryser, Maria Santi-Vicini, Kazuhito Sato, Deborah Schrag, Nikolaus Schultz, Cynthia L. Sears, Rosalie C. Sears, Subrata Sen, Triparna Sen, Alex Shalek, Jeff Sheng, Quanhu Sheng, Kooresh I. Shoghi, Martha J. Shrubsole, Yu Shyr, Alexander B. Sibley, Kiara Siex, Alan J. Simmons, Dinah S. Singer, Shamilene Sivagnanam, Michal Slyper, Artem Sokolov, Sheng-Kwei Song, Austin Southard-Smith, Avrum Spira, Janet Stein, Phillip Storm, Elizabeth Stover, Siri H. Strand, Timothy Su, Damir Sudar, Ryan Sullivan, Lea Surrey, Mario Suvà, Nadezhda V. Terekhanova, Luke Ternes, Lisa Thammavong, Guillaume Thibault, George V. Thomas, Vésteinn Thorsson, Ellen Todres, Linh Tran, Madison Tyler, Yasin Uzun, Anil Vachani, Eliezer Van Allen, Simon Vandekar, Deborah J. Veis, Sébastien Vigneau, Arastoo Vossough, Angela Waanders, Nikhil Wagle, Liang-Bo Wang, Michael C. Wendl, Robert West, Chi-yun Wu, Hao Wu, Hung-Yi Wu, Matthew A. Wyczalkowski, Yubin Xie, Xiaolu Yang, Clarence Yapp, Wenbao Yu, Yinyin Yuan, Dadong Zhang, Kun Zhang, Mianlei Zhang, Nancy Zhang, Yantian Zhang, Yanyan Zhao, Daniel Cui Zhou, Zilu Zhou, Houxiang Zhu, Qin Zhu, Xiangzhu Zhu, Yuankun Zhu, and Xiaowei Zhuang
- Subjects
Cell ,Genomics ,Computational biology ,Tumor initiation ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,03 medical and health sciences ,Atlases as Topic ,0302 clinical medicine ,Neoplasms ,Tumor Microenvironment ,medicine ,Humans ,Precision Medicine ,030304 developmental biology ,0303 health sciences ,Atlas (topology) ,Cancer ,medicine.disease ,3. Good health ,Human tumor ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Single-Cell Analysis ,Single point ,030217 neurology & neurosurgery - Abstract
Crucial transitions in cancer-including tumor initiation, local expansion, metastasis, and therapeutic resistance-involve complex interactions between cells within the dynamic tumor ecosystem. Transformative single-cell genomics technologies and spatial multiplex in situ methods now provide an opportunity to interrogate this complexity at unprecedented resolution. The Human Tumor Atlas Network (HTAN), part of the National Cancer Institute (NCI) Cancer Moonshot Initiative, will establish a clinical, experimental, computational, and organizational framework to generate informative and accessible three-dimensional atlases of cancer transitions for a diverse set of tumor types. This effort complements both ongoing efforts to map healthy organs and previous large-scale cancer genomics approaches focused on bulk sequencing at a single point in time. Generating single-cell, multiparametric, longitudinal atlases and integrating them with clinical outcomes should help identify novel predictive biomarkers and features as well as therapeutically relevant cell types, cell states, and cellular interactions across transitions. The resulting tumor atlases should have a profound impact on our understanding of cancer biology and have the potential to improve cancer detection, prevention, and therapeutic discovery for better precision-medicine treatments of cancer patients and those at risk for cancer.
- Published
- 2020
- Full Text
- View/download PDF
15. Lineage plasticity in cancer: a shared pathway of therapeutic resistance
- Author
-
Charles M. Rudin, Dana Pe'er, Helena A. Yu, Triparna Sen, Álvaro Quintanal-Villalonga, Joseph M. Chan, and Sawyers Charles L
- Subjects
Male ,0301 basic medicine ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,Lineage (genetic) ,Ubiquitin-Protein Ligases ,Cell Plasticity ,Cell ,Drug resistance ,Adenocarcinoma ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Tumor Microenvironment ,medicine ,Humans ,Cell Lineage ,Epithelial–mesenchymal transition ,Protein Kinase Inhibitors ,SOX Transcription Factors ,Tumor microenvironment ,business.industry ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,Cancer ,Androgen Antagonists ,medicine.disease ,ErbB Receptors ,Neuroendocrine Tumors ,Prostatic Neoplasms, Castration-Resistant ,Retinoblastoma Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Tumor Suppressor Protein p53 ,business ,Proto-Oncogene Proteins c-akt - Abstract
Lineage plasticity, the ability of cells to transition from one committed developmental pathway to another, has been proposed as a source of intratumoural heterogeneity and of tumour adaptation to an adverse tumour microenvironment including exposure to targeted anticancer treatments. Tumour cell conversion into a different histological subtype has been associated with a loss of dependency on the original oncogenic driver, leading to therapeutic resistance. A well-known pathway of lineage plasticity in cancer — the histological transformation of adenocarcinomas to aggressive neuroendocrine derivatives — was initially described in lung cancers harbouring an EGFR mutation, and was subsequently reported in multiple other adenocarcinomas, including prostate cancer in the presence of antiandrogens. Squamous transformation is a subsequently identified and less well-characterized pathway of adenocarcinoma escape from suppressive anticancer therapy. The increased practice of tumour re-biopsy upon disease progression has increased the recognition of these mechanisms of resistance and has improved our understanding of the underlying biology. In this Review, we provide an overview of the impact of lineage plasticity on cancer progression and therapy resistance, with a focus on neuroendocrine transformation in lung and prostate tumours. We discuss the current understanding of the molecular drivers of this phenomenon, emerging management strategies and open questions in the field. Lineage plasticity is a source of intratumoural heterogeneity and enables tumour adaptation to an adverse tumour microenvironment, eventually leading to therapeutic resistance. The authors of this Review provide an overview of the impact of lineage plasticity on cancer progression and therapy resistance, with a focus on neuroendocrine transformation in lung and prostate tumours, and discuss emerging management strategies and open questions in the field.
- Published
- 2020
- Full Text
- View/download PDF
16. Targeting Lysine-Specific Demethylase 1 Rescues Major Histocompatibility Complex Class I Antigen Presentation and Overcomes Programmed Death-Ligand 1 Blockade Resistance in SCLC
- Author
-
Evelyn M. Nguyen, Hirokazu Taniguchi, Joseph M. Chan, Yingqian A. Zhan, Xiaoping Chen, Juan Qiu, Elisa de Stanchina, Viola Allaj, Nisargbhai S. Shah, Fathema Uddin, Parvathy Manoj, Michael Liu, Sheng F. Cai, Ross Levine, Álvaro Quintanal-Villalonga, Triparna Sen, Andrew Chow, and Charles M. Rudin
- Subjects
Pulmonary and Respiratory Medicine ,Histone Demethylases ,Antigen Presentation ,Mice ,Lung Neoplasms ,Oncology ,Antigens, Neoplasm ,Histocompatibility Antigens Class I ,Animals ,Genes, MHC Class I ,Humans ,Small Cell Lung Carcinoma ,B7-H1 Antigen - Abstract
SCLC is a highly aggressive neuroendocrine tumor that is characterized by early acquired therapeutic resistance and modest benefit from immune checkpoint blockade (ICB). Repression of the major histocompatibility complex class I (MHC-I) represents a key mechanism driving resistance to T cell-based immunotherapies.We evaluated the role of the lysine-specific demethylase 1 (LSD1) as a determinant of MHC-I expression, functional antigen presentation, and immune activation in SCLC in vitro and in vivo through evaluation of both human SCLC cell lines and immunocompetent mouse models.We found that targeted inhibition of LSD1 in SCLC restores MHC-I cell surface expression and transcriptionally activates genes encoding the antigen presentation pathway. LSD1 inhibition further activates interferon signaling, induces tumor-intrinsic immunogenicity, and sensitizes SCLC cells to MHC-I-restricted T cell cytolysis. Combination of LSD1 inhibitor with ICB augments the antitumor immune response in refractory SCLC models. Together, these data define a role for LSD1 as a potent regulator of MHC-I antigen presentation and provide rationale for combinatory use of LSD1 inhibitors with ICB to improve therapeutic response in SCLC.Epigenetic silencing of MHC-I in SCLC contributes to its poor response to ICB. Our study identifies a previously uncharacterized role for LSD1 as a regulator of MHC-I antigen presentation in SCLC. LSD1 inhibition enables MHC-I-restricted T cell cytolysis, induces immune activation, and augments the antitumor immune response to ICB in SCLC.
- Published
- 2022
17. Comprehensive molecular characterization of lung tumors implicates AKT and MYC signaling in adenocarcinoma to squamous cell transdifferentiation
- Author
-
Marina Asher, Jason C. Chang, Yingqian Zhan, Natasha Rekhtman, Hirokazu Taniguchi, Richard Koche, Brian Houck-Loomis, Elisa de Stanchina, Triparna Sen, Charles M. Rudin, Fanli Meng, Irina Linkov, Jacklynn D. Egger, Umesh Bhanot, Nisargbhai S. Shah, Fathema Uddin, Shweta S. Chavan, Michael H.A. Roehrl, Helena A. Yu, Joseph M. Chan, Michael Offin, Viola Allaj, P. Manoj, Katia Ventura, J. Qiu, Jordana Ray-Kirton, Metamia Ciampricotti, Maysun Hasan, Andrew Chow, and Álvaro Quintanal-Villalonga
- Subjects
Cancer Research ,medicine.medical_treatment ,Adenocarcinoma of Lung ,Context (language use) ,Treatment resistance ,Biology ,Targeted therapy ,Proto-Oncogene Proteins c-myc ,Phosphatidylinositol 3-Kinases ,Mice, Inbred NOD ,Carcinoma, Non-Small-Cell Lung ,medicine ,Animals ,Humans ,Diseases of the blood and blood-forming organs ,Osimertinib ,Molecular Biology ,Protein kinase B ,RC254-282 ,Squamous transdifferentiation ,PI3K/AKT/mTOR pathway ,Research ,Lineage plasticity ,Transdifferentiation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Hematology ,medicine.disease ,Oncology ,Cell Transdifferentiation ,Carcinoma, Squamous Cell ,Cancer research ,Adenocarcinoma ,RC633-647.5 ,Transcriptome ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Background Lineage plasticity, the ability to transdifferentiate among distinct phenotypic identities, facilitates therapeutic resistance in cancer. In lung adenocarcinomas (LUADs), this phenomenon includes small cell and squamous cell (LUSC) histologic transformation in the context of acquired resistance to targeted inhibition of driver mutations. LUAD-to-LUSC transdifferentiation, occurring in up to 9% of EGFR-mutant patients relapsed on osimertinib, is associated with notably poor prognosis. We hypothesized that multi-parameter profiling of the components of mixed histology (LUAD/LUSC) tumors could provide insight into factors licensing lineage plasticity between these histologies. Methods We performed genomic, epigenomics, transcriptomics and protein analyses of microdissected LUAD and LUSC components from mixed histology tumors, pre-/post-transformation tumors and reference non-transformed LUAD and LUSC samples. We validated our findings through genetic manipulation of preclinical models in vitro and in vivo and performed patient-derived xenograft (PDX) treatments to validate potential therapeutic targets in a LUAD PDX model acquiring LUSC features after osimertinib treatment. Results Our data suggest that LUSC transdifferentiation is primarily driven by transcriptional reprogramming rather than mutational events. We observed consistent relative upregulation of PI3K/AKT, MYC and PRC2 pathway genes. Concurrent activation of PI3K/AKT and MYC induced squamous features in EGFR-mutant LUAD preclinical models. Pharmacologic inhibition of EZH1/2 in combination with osimertinib prevented relapse with squamous-features in an EGFR-mutant patient-derived xenograft model, and inhibition of EZH1/2 or PI3K/AKT signaling re-sensitized resistant squamous-like tumors to osimertinib. Conclusions Our findings provide the first comprehensive molecular characterization of LUSC transdifferentiation, suggesting putative drivers and potential therapeutic targets to constrain or prevent lineage plasticity.
- Published
- 2021
- Full Text
- View/download PDF
18. Signatures of plasticity, metastasis, and immunosuppression in an atlas of human small cell lung cancer
- Author
-
Umesh Bhanot, David R. Jones, Thomas Walle, Ojasvi Chaudhary, Triparna Sen, Besnik Qeriqi, Linas Mazutis, Ignas Masilionis, Elisa de Stanchina, Dana Pe'er, W. Victoria Lai, Marissa Mattar, Dig Vijay Kumar Yarlagadda, Andrew Chow, Marina K. Baine, Fathema Uddin, Arvin Ruiz, Yanyun Li, Álvaro Quintanal-Villalonga, Natasha Rekhtman, Jacklynn V. Egger, Tal Nawy, Joseph M. Chan, John T. Poirier, Charles M. Rudin, Matthew J. Bott, Michael Offin, Amber Bahr, Viola Allaj, Travis J. Hollmann, Metamia Ciampricotti, James L. Wang, Vianne R. Gao, and Yubin Xie
- Subjects
Cancer Research ,Myeloid ,Lung Neoplasms ,medicine.medical_treatment ,Cell Plasticity ,Biology ,Malignancy ,Article ,Metastasis ,Immune system ,medicine ,Humans ,Neoplasm Metastasis ,neoplasms ,Lung ,Phospholipase C gamma ,Sequence Analysis, RNA ,Gene Expression Profiling ,Immunosuppression ,medicine.disease ,Prognosis ,Phenotype ,Small Cell Lung Carcinoma ,Survival Analysis ,humanities ,respiratory tract diseases ,metastasis ,myeloid ,PLCG2 ,SCLC ,scRNA-seq ,single cell ,tumor atlas ,medicine.anatomical_structure ,Oncology ,Cancer research ,Adenocarcinoma ,Single-Cell Analysis - Abstract
Summary Small cell lung cancer (SCLC) is an aggressive malignancy that includes subtypes defined by differential expression of ASCL1, NEUROD1, and POU2F3 (SCLC-A, -N, and -P, respectively). To define the heterogeneity of tumors and their associated microenvironments across subtypes, we sequenced 155,098 transcriptomes from 21 human biospecimens, including 54,523 SCLC transcriptomes. We observe greater tumor diversity in SCLC than lung adenocarcinoma, driven by canonical, intermediate, and admixed subtypes. We discover a PLCG2-high SCLC phenotype with stem-like, pro-metastatic features that recurs across subtypes and predicts worse overall survival. SCLC exhibits greater immune sequestration and less immune infiltration than lung adenocarcinoma, and SCLC-N shows less immune infiltrate and greater T cell dysfunction than SCLC-A. We identify a profibrotic, immunosuppressive monocyte/macrophage population in SCLC tumors that is particularly associated with the recurrent, PLCG2-high subpopulation.
- Published
- 2021
19. Regional and clonal T cell dynamics at single cell resolution in immune checkpoint blockade
- Author
-
Charles M. Rudin, Daniel K. Wells, Taha Merghoub, P. Manoj, Triparna Sen, H. J. Woo, Marissa Mattar, Jennifer L. Sauter, Joy A. Pai, Hira Rizvi, Mark T.A. Donoghue, Viola Allaj, Matthew D. Hellmann, Ansuman T. Satpathy, Jamie E. Chaft, Nisargbhai S. Shah, Helen Won, Andrew J. Plodkowski, Marina K. Baine, E. De Stanchina, Fathema Uddin, Brian Houck-Loomis, Álvaro Quintanal-Villalonga, Jedd D. Wolchok, Andrew Chow, and Joseph M. Chan
- Subjects
medicine.anatomical_structure ,Single cell sequencing ,T cell ,T-cell receptor ,Cell ,Cancer cell ,Cancer research ,medicine ,Cytotoxic T cell ,Biology ,CD8 ,Immune checkpoint - Abstract
Paired T cell receptor and RNA single cell sequencing (scTCR/RNA-seq) has allowed for enhanced resolution of clonal T cell dynamics in cancer. Here, we report a scTCR/RNA-seq dataset of 162,062 single T cells from 31 tissue regions, including tumor, adjacent normal tissues, and lymph nodes (LN), from three patients who underwent resections for progressing lung cancers after immune checkpoint blockade (ICB). We found marked regional heterogeneity in tumor persistence that was associated with heterogeneity in CD4 and CD8 T cell phenotypes; regions with persistent cancer cells were enriched for follicular helper CD4 T cells (TFH), regulatory T cells (Treg), and exhausted CD8 T cells. Clonal analysis demonstrated that highly-expanded T cell clones were predominantly of the CD8 subtype, were ubiquitously present across all sampled regions, found in the peripheral circulation, and expressed gene signatures of ‘large’ and ‘dual-expanded’ clones that have been predictive of response to ICB. Longitudinal tracking of CD8 T cell clones in the peripheral blood revealed that the persistence of ubiquitous CD8 T cell clones, as well as phenotypically distinct clones with tumor-reactive features, correlated with systemic tumor control. Finally, tracking CD8 T cell clones across tissues revealed the presence of TCF-1+precursor exhausted CD8 T cells in tumor draining LNs that were clonally linked to expanded exhausted CD8 T cells in tumors. Altogether, this comprehensive scTCR/RNA-seq dataset with regional, longitudinal, and clonal resolution provides fundamental insights into the tissue distribution, persistence, and differentiation trajectories of ICB-responsive T cells that underlie clinical responses to ICB.
- Published
- 2021
- Full Text
- View/download PDF
20. An optimized NGS sample preparation protocol for in vitro CRISPR screens
- Author
-
Fathema Uddin, John T. Poirier, Charles M. Rudin, Álvaro Quintanal-Villalonga, Triparna Sen, and Corrin A. Wohlhieter
- Subjects
Protocol (science) ,Science (General) ,General Immunology and Microbiology ,Computer science ,General Neuroscience ,Computational biology ,Human cell ,High Throughput Screening ,General Biochemistry, Genetics and Molecular Biology ,Calculation methods ,Q1-390 ,CRISPR ,Sample preparation ,Molecular Biology ,Cancer - Abstract
Summary This standardized protocol describes the preparation of PCR amplified and purified samples from human cell lines passaged and collected from CRISPR screening. High-quality samples can be used to perform next-generation sequencing (NGS) to uncover changes in sgRNA abundance from the timepoint at which library-transduced cells are selected to the timepoint when the screen is ended. Here, we describe proper calculation methods for library representation and show how to overcome potential issues often encountered by researchers. For complete information on the use and execution of this protocol, please refer to Wohlhieter et al. (2020) .
- Published
- 2021
21. Abstract 859: Characterizing SMARCA4/STK11/KEAP1 co-mutant lung adenocarcinoma
- Author
-
Emily Costa, Corrin Wohlheiter, Samuel Tischfield, Alister Funnell, JT Poirier, Álvaro Quintanal Villalonga, Triparna Sen, and Charles Rudin
- Subjects
Cancer Research ,Oncology - Abstract
Lung adenocarcinoma (LUAD), constituting 50% of non-small cell lung cancer, is classically defined by the presence of driver mutations in oncogenes such as KRAS, EGFR, and BRAF. While the development of driver-targeted therapies has significantly improved survival in subsets of patients with LUAD, studies examining tumor sequencing data and clinical outcomes have shown that intra-driver genetic and phenotypic heterogeneity underlie differential patient responses to these therapies. In particular, co-occurring alterations in the genes SMARCA4 (BRG1), STK11 (LKB1), and KEAP1 are predictive of exceptionally poor prognosis and worse overall survival in metastatic LUAD patients. SMARCA4 loss is also a hallmark of undifferentiated, highly chemoresistant NSCLC tumors. While these co-mutations most often appear in KRAS-driven tumors, their association with poor outcomes is maintained regardless of driver status. Studies exploring the molecular features of SMARCA4-deficient and STK11/KEAP1 co-mutant LUAD have yielded translational insights specific to these subsets, establishing a rationale to characterize the unique biology of SMARCA4/STK11/KEAP1 (BLK) triple-mutant LUAD. To this end, we are profiling BLK LUAD through a combination of genomic, transcriptomic, epigenomic, and phenotypic assays in in vitro models. We have generated paired isogenic in vitro models of BLK LUAD via CRISPR-Cas9 knockout. Phenotypic analyses of these cell lines, whose engineered genotypes model varying combinations of SMARCA4, KEAP1, and STK11 loss, have revealed growth rate differences in vitro, and pairwise comparisons of RNA-sequencing in these lines show downregulation of lung differentiation markers and upregulation of metastasis-associated genes concomitant with SMARCA4 loss. In conclusion, we have generated isogenic preclinical models showing molecular and phenotypic features consistent with observations reported in BLK LUADs. Further molecular and functional characterization of these models may reveal mechanistic insights into the biology of this subset of aggressive tumors. Citation Format: Emily Costa, Corrin Wohlheiter, Samuel Tischfield, Alister Funnell, JT Poirier, Álvaro Quintanal Villalonga, Triparna Sen, Charles Rudin. Characterizing SMARCA4/STK11/KEAP1 co-mutant lung adenocarcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 859.
- Published
- 2022
- Full Text
- View/download PDF
22. Rlf-Mycl Gene Fusion Drives Tumorigenesis and Metastasis in a Mouse Model of Small Cell Lung Cancer
- Author
-
Triantafyllia R. Karakousi, William M. Rideout, Allison Richards, Andrea Ventura, Mark T.A. Donoghue, Janneke E. Jaspers, Triparna Sen, Elliot H. Akama-Garren, Trudy G. Oliver, Tyler Jacks, Faruk Erdem Kombak, Anastasia-Maria Zavitsanou, John T. Poirier, Metamia Ciampricotti, Álvaro Quintanal-Villalonga, Francisco J. Sánchez-Rivera, Viola Allaj, P. Manoj, Danilo Maddalo, Thales Papagiannakopoulos, Rebecca Caeser, Emily A. Costa, Angeliki Karatza, Kyle B. Spainhower, and Charles M. Rudin
- Subjects
Lung Neoplasms ,Somatic cell ,Carcinogenesis ,Telomere-Binding Proteins ,Genes, myc ,Biology ,medicine.disease_cause ,Article ,Metastasis ,Fusion gene ,Proto-Oncogene Proteins c-myc ,Mice ,Cell Line, Tumor ,Gene expression ,medicine ,CRISPR ,Animals ,neoplasms ,Cas9 ,medicine.disease ,Small Cell Lung Carcinoma ,humanities ,respiratory tract diseases ,Oncology ,Tumor progression ,Cancer research ,Gene Fusion - Abstract
Small cell lung cancer (SCLC) has limited therapeutic options and an exceptionally poor prognosis. Understanding the oncogenic drivers of SCLC may help define novel therapeutic targets. Recurrent genomic rearrangements have been identified in SCLC, most notably an in-frame gene fusion between RLF and MYCL found in up to 7% of the predominant ASCL1-expressing subtype. To explore the role of this fusion in oncogenesis and tumor progression, we used CRISPR/Cas9 somatic editing to generate a Rlf–Mycl-driven mouse model of SCLC. RLF–MYCL fusion accelerated transformation and proliferation of murine SCLC and increased metastatic dissemination and the diversity of metastatic sites. Tumors from the RLF–MYCL genetically engineered mouse model displayed gene expression similarities with human RLF–MYCL SCLC. Together, our studies support RLF–MYCL as the first demonstrated fusion oncogenic driver in SCLC and provide a new preclinical mouse model for the study of this subtype of SCLC. Significance: The biological and therapeutic implications of gene fusions in SCLC, an aggressive metastatic lung cancer, are unknown. Our study investigates the functional significance of the in-frame RLF–MYCL gene fusion by developing a Rlf–Mycl-driven genetically engineered mouse model and defining the impact on tumor growth and metastasis. This article is highlighted in the In This Issue feature, p. 2945
- Published
- 2021
23. Identification of Predictive Biomarkers of Response to HSP90 Inhibitors in Lung Adenocarcinoma
- Author
-
Ángela Marrugal, Álvaro Quintanal-Villalonga, Sonia Molina-Pinelo, Irene Ferrer, David Gomez-Sanchez, Luis Paz-Ares, María Dolores Pastor, and Laura Ojeda
- Subjects
0301 basic medicine ,Proteomics ,Lung Neoplasms ,Oncología ,lcsh:Chemistry ,chemistry.chemical_compound ,0302 clinical medicine ,Benzoquinones ,polycyclic compounds ,Neumología ,lcsh:QH301-705.5 ,Spectroscopy ,biology ,Chemistry ,General Medicine ,Geldanamycin ,Hsp90 ,Computer Science Applications ,Radicicol ,030220 oncology & carcinogenesis ,Adenocarcinoma ,HSP90 inhibitors ,protein biomarkers predictive of response ,lung adenocarcinoma ,Bioquímica ,Lactams, Macrocyclic ,Adenocarcinoma of Lung ,Antineoplastic Agents ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,PDSS2 ,Heat shock protein ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Humans ,HSP90 Heat-Shock Proteins ,Physical and Theoretical Chemistry ,Molecular Biology ,Topoisomerase ,Organic Chemistry ,medicine.disease ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,A549 Cells ,Chaperone (protein) ,biology.protein ,Cancer research ,Molecular Chaperones - Abstract
Heat shock protein 90 (HSP90) plays an essential role in lung adenocarcinoma, acting as a key chaperone involved in the correct functioning of numerous highly relevant protein drivers of this disease. To this end, HSP90 inhibitors have emerged as promising therapeutic strategies, even though responses to them have been limited to date. Given the need to maximize treatment efficacy, the objective of this study was to use isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomic techniques to identify proteins in human lung adenocarcinoma cell lines whose basal abundances were correlated with response to HSP90 inhibitors (geldanamycin and radicicol derivatives). From the protein profiles identified according to response, the relationship between lactate dehydrogenase B (LDHB) and DNA topoisomerase 1 (TOP1) with respect to sensitivity and resistance, respectively, to geldanamycin derivatives is noteworthy. Likewise, rhotekin (RTKN) and decaprenyl diphosphate synthase subunit 2 (PDSS2) were correlated with sensitivity and resistance to radicicol derivatives. We also identified a relationship between resistance to HSP90 inhibition and the p53 pathway by glucose deprivation. In contrast, arginine biosynthesis was correlated with sensitivity to HSP90 inhibitors. Further study of these outcomes could enable the development of strategies to improve the clinical efficacy of HSP90 inhibition in patients with lung adenocarcinoma.
- Published
- 2021
24. Single cell profiling reveals novel tumor and myeloid subpopulations in small cell lung cancer
- Author
-
Marissa Mattar, James L. Wang, Vianne R. Gao, Yubin Xie, Linas Mazutis, Charles M. Rudin, Dig Vk Yarlagadda, Jacklynn V. Egger, Thomas Walle, John T. Poirier, David R. Jones, Tal Nawy, Umesh Bhanot, Ignas Masilionis, Arvin Ruiz, Ojasvi Chaudhary, Triparna Sen, Dana Pe'er, Matthew J. Bott, Michael Offin, Joseph M. Chan, W. Victoria Lai, Viola Allaj, Fathema Uddin, Andrew Chow, Travis J. Hollmann, Metamia Ciampricotti, and Álvaro Quintanal-Villalonga
- Subjects
Myeloid ,Cell ,Biology ,medicine.disease ,Malignancy ,respiratory tract diseases ,Metastasis ,Transcriptome ,ASCL1 ,medicine.anatomical_structure ,medicine ,Cancer research ,Adenocarcinoma ,CD8 - Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy that includes subtypes defined by differential expression of ASCL1, NEUROD1, and POU2F3 (SCLC-A, -N, and -P, respectively), which are associated with distinct therapeutic vulnerabilities. To define the heterogeneity of tumors and their associated microenvironments across subtypes, we sequenced 54,523 cellular transcriptomes from 21 human biospecimens. Our single-cell SCLC atlas reveals tumor diversity exceeding lung adenocarcinoma, driven by canonical, intermediate, and admixed subtypes. We discovered a PLCG2-high tumor cell population with stem-like, pro-metastatic features that recurs across subtypes and predicts worse overall survival, and manipulation of PLCG2 expression in cells confirms correlation with key metastatic markers. Treatment and subtype are associated with substantial phenotypic changes in the SCLC immune microenvironment, with greater T-cell dysfunction in SCLC-N than SCLC-A. Moreover, the recurrent, PLCG2-high subclone is associated with exhausted CD8+ T-cells and a pro-fibrotic, immunosuppressive monocyte/macrophage population, suggesting possible tumor-immune coordination to promote metastasis.
- Published
- 2020
- Full Text
- View/download PDF
25. Concurrent Mutations in STK11 and KEAP1 Promote Ferroptosis Protection and SCD1 Dependence in Lung Cancer
- Author
-
Elisa de Stanchina, Corrin A. Wohlhieter, Charles M. Rudin, Omar Hayatt, Fathema Uddin, Axel Martin, Nisargbhai S. Shah, Mark T.A. Donoghue, Christopher H. Hulton, U. Bhanot, Triparna Sen, Marina Asher, Natasha Rekhtman, John T. Poirier, Allison Richards, Álvaro Quintanal-Villalonga, Darren J. Buonocore, Kathryn C. Arbour, and Ronglai Shen
- Subjects
0301 basic medicine ,Lung Neoplasms ,LKB1 ,AKR1C1 ,STK11 ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,AMP-Activated Protein Kinase Kinases ,medicine ,Humans ,CRISPR ,Lung cancer ,Gene ,lcsh:QH301-705.5 ,Kelch-Like ECH-Associated Protein 1 ,medicine.disease ,KEAP1 ,ferroptosis ,030104 developmental biology ,Drug development ,lcsh:Biology (General) ,Mutation ,Cancer research ,Adenocarcinoma ,Stearoyl-CoA Desaturase ,030217 neurology & neurosurgery ,SCD1 - Abstract
SUMMARY Concurrent loss-of-function mutations in STK11 and KEAP1 in lung adenocarcinoma (LUAD) are associated with aggressive tumor growth, resistance to available therapies, and early death. We investigated the effects of coordinate STK11 and KEAP1 loss by comparing co-mutant with single mutant and wild-type isogenic counterparts in multiple LUAD models. STK11/KEAP1 co-mutation results in significantly elevated expression of ferroptosis-protective genes, including SCD and AKR1C1/2/3, and resistance to pharmacologically induced ferroptosis. CRISPR screening further nominates SCD (SCD1) as selectively essential in STK11/KEAP1 co-mutant LUAD. Genetic and pharmacological inhibition of SCD1 confirms the essentiality of this gene and augments the effects of ferroptosis induction by erastin and RSL3. Together these data identify SCD1 as a selective vulnerability and a promising candidate for targeted drug development in STK11/KEAP1 co-mutant LUAD., Graphical Abstract, In Brief Wohlhieter et al. explore the global changes in gene expression and oncogenic signaling pathways driven by concurrent loss of function in two tumor suppressor genes, STK11 and KEAP1. They identify a molecular vulnerability, in which co-mutant cells depend on ferroptosis protective mechanisms for survival, and highlight SCD1 as an essential gene and promising drug target.
- Published
- 2020
26. Processing of human surgical samples for single-cell sequencing v1
- Author
-
Álvaro Quintanal-Villalonga, not provided Viola Allaj, not provided Andrew Chow, not provided Nisarg Shah, not provided Linas Mazutis, John Thomas Poirier, Triparna Sen, Dana Pe'er, and Charles M. Rudin
- Subjects
Single cell sequencing ,Computational biology ,Biology - Abstract
Human clinical samples pose a challenge for single-cell RNA-seq (scRNA-seq) and single-nucleus ATAC-seq (snATAC-seq) analysis, due to their heterogeneity in cell type composition and degree of degradation or necrosis. Moreover, although some tissues can be sampled by surgical resection, most biospecimens are derived from core needle biopsies or fine needle aspirates, and must be allocated to pressing needs such as pathological analysis, thereby severely limiting the number of cells available for analysis. We have developed a protocol for obtaining robust, high-quality single-cell sequencing data from human biospecimens from a variety of biopsy types, enabling greater access to clinical cohorts. A critical factor is to limit ischemic time and transport to the lab to under an hour, and to restrict subsequent processing time to under 3 hours. This protocol uses a commercial tissue disruptor for dissociation. It was optimized on diverse human lung tumor samples, including pleural effusions, but is widely applicable to normal and diseased human clinical tissues from different organ sites.
- Published
- 2020
- Full Text
- View/download PDF
27. FGFR1 and FGFR4 oncogenicity depends on n-cadherin and their coexpression may predict FGFR-targeted therapy efficacy
- Author
-
Santiago García, Luis Paz-Ares, Fernando Lopez-Rios, Jon Zugazagoitia, Sonia Molina-Pinelo, Laura Ojeda, Irene Ferrer, Sandra Muñoz-Galván, Elizabeth Guruceaga, Luis M. Montuenga, Silvestre Vicent, Cristina Cirauqui, Álvaro Quintanal-Villalonga, Amancio Carnero, Ángela Marrugal, [Quintanal-Villalonga,Á, Ferrer,I, Cirauqui,C, Marrugal,Á, Ojeda,L, García,S, Zugazagoitia,J, Paz-Ares,L] H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigacion Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain. [Quintanal-Villalonga,Á] Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. [Ferrer,I, Muñoz-Galván,S, Lopez-Rios,F, Montuenga,L, Vicent,S, Molina-Pinelo,S, Carnero,A, Paz-Ares,L] CIBERONC, Madrid, Spain. [Guruceaga,E] Bioinformatics Unit, Centre for Applied Medical Research (CIMA), Pamplona, Spain. [Guruceaga,E] PROTEORED, Madrid, Spain. [Zugazagoitia,J, Paz-Ares,L] Medical Oncology Department, Hospital Universitario Doce de Octubre. Madrid, Spain. [Muñoz-Galván,S, Carnero,A] Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain. [Lopez-Rios,F] Laboratorio de Dianas Terapeuticas, Hospital Universitario HM Sanchinarro, Madrid, Spain. [Montuenga,L, Vicent,S] Program in Solid Tumors, Centre for Applied Medical Research (CIMA), Pamplona, Spain. [Montuenga,L, Vicent,S] Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain. [Montuenga,L, Vicent,S] IdiSNA, Navarra Institute for Health Research, Pamplona, SPAIN. [Paz-Ares,L] Medical School, Universidad Complutense, Madrid, Spain., This work was funded by the Community of Madrid, the ISCIII cofunded by FEDER from Regional Development European Funds (European Union), the Spanish Ministry of Economy and Competitiveness, the Mutua Madrilena Foundation, the Ministry of Health and Social ~ Welfare of Junta de Andalucía, the AECC scientific foundation and the Spanish Ministry of Education, Culture and Sport. L.P.A. was funded by the Community of Madrid, CAM, (B2017/BMD3884), ISCIII (PIE15/00076, PI17/00778 and DTS17/00089) and CIBERONC (CD16/12/00442), and co-funded by FEDER from Regional Development European Funds (European Union). A.C. was funded by grants from the Spanish Ministry of Economy and Competitiveness Plan Estatal de I+D+I 2018 cofunded by FEDER: RTI2018-097455-B-I00, CIBER de Cancer (CB16/ 12/00275), co-funded by FEDER from Regional Development European Funds. S.M.P. is funded by the Mutua Madrilena Foundation ~ (2014) Ministry of Health and Social Welfare of Junta de Andalucía (PI-0046-2012, Nicolas Monardes Program C-0040-2016), ISCIII (PI17/00033), and co-funded by FEDER from Regional Development European Funds (European Union). I.F. is funded by the AECC scientific foundation (AIO2015) and Consejería de Igualdad, Salud y Políticas Sociales de la Junta de Andalucía (PI-0029-2013) and ISCIII (PI16/ 01311) and co-funded by FEDER from Regional Development European Funds (European Union). A.Q. is funded by ISCIII (FI12/00429). L. O. is funded by the Spanish Ministry of Education, Culture and Sport (FPU13/02595). S.V. is supported by the Spanish Ministry of Economy and Competitiveness (MINECO, SAF2017-89944-R)., Comunidad de Madrid, Instituto de Salud Carlos III, European Commission, Ministerio de Economía y Competitividad (España), Fundación Mutua Madrileña, Junta de Andalucía, Fundación Científica Asociación Española Contra el Cáncer, Ministerio de Educación, Cultura y Deporte (España), European Regional Development Fund, Regional Government of Andalusia (España), Asociación Española Contra el Cáncer, European Regional Development Fund (ERDF/FEDER), Fundación Mutua Madrileña Automovilista, Gobierno de Andalucia, and Asociacion Espanola Contra el Cancer (AECC)
- Subjects
0301 basic medicine ,Anatomy::Cells::Cells, Cultured::Cell Line::Cell Line, Tumor [Medical Subject Headings] ,Lung Neoplasms ,Research paper ,medicine.medical_treatment ,lcsh:Medicine ,medicine.disease_cause ,Piperazines ,Targeted therapy ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Primates::Haplorhini::Catarrhini::Hominidae::Humans [Medical Subject Headings] ,Mice ,0302 clinical medicine ,Lung neoplasms ,Carcinoma, Non-Small-Cell Lung ,Phenomena and Processes::Physiological Phenomena::Pharmacological Phenomena::Drug Resistance::Drug Resistance, Neoplasm [Medical Subject Headings] ,Tumor Cells, Cultured ,Organisms::Eukaryota::Animals [Medical Subject Headings] ,FGFR inhibitors ,Receptor tipo 1 de factor de crecimiento de fibroblastos ,Chemicals and Drugs::Enzymes and Coenzymes::Enzymes::Transferases::Phosphotransferases::Phosphotransferases (Alcohol Group Acceptor)::Protein Kinases::Protein-Tyrosine Kinases::Receptor Protein-Tyrosine Kinases::Receptor, Fibroblast Growth Factor, Type 1 [Medical Subject Headings] ,lcsh:R5-920 ,Diseases::Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms::Bronchial Neoplasms::Carcinoma, Bronchogenic::Carcinoma, Non-Small-Cell Lung [Medical Subject Headings] ,Receptor tipo 4 de factor de crecimiento de fibroblastos ,Anatomy::Cells::Cells, Cultured::Tumor Cells, Cultured [Medical Subject Headings] ,General Medicine ,Cadherins ,Chemicals and Drugs::Enzymes and Coenzymes::Enzymes::Transferases::Phosphotransferases::Phosphotransferases (Alcohol Group Acceptor)::Protein Kinases::Protein-Tyrosine Kinases::Receptor Protein-Tyrosine Kinases::Receptor, Fibroblast Growth Factor, Type 4 [Medical Subject Headings] ,Predictive biomarker ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Benzamides ,Female ,lcsh:Medicine (General) ,Neoplasias pulmonares ,Mice, Nude ,Antineoplastic Agents ,Diseases::Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms [Medical Subject Headings] ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Chemicals and Drugs::Biological Factors::Biological Markers [Medical Subject Headings] ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 4 ,Receptor, Fibroblast Growth Factor, Type 1 ,Lung cancer ,N-cadherin ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Rodentia::Muridae::Murinae::Mice [Medical Subject Headings] ,business.industry ,Fibroblast growth factor receptor 1 ,lcsh:R ,Chemicals and Drugs::Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [Medical Subject Headings] ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Rodentia::Muridae::Murinae::Mice::Mice, Mutant Strains::Mice, Nude [Medical Subject Headings] ,Fibroblast growth factor receptor 4 ,Chemicals and Drugs::Amino Acids, Peptides, and Proteins::Proteins::Glycoproteins::Membrane Glycoproteins::Cell Adhesion Molecules::Cadherins [Medical Subject Headings] ,medicine.disease ,Isogenic human disease models ,stomatognathic diseases ,030104 developmental biology ,Biomarcadores ,FGFR1 ,Check Tags::Female [Medical Subject Headings] ,Chemicals and Drugs::Heterocyclic Compounds::Heterocyclic Compounds, 1-Ring::Azoles::Pyrazoles [Medical Subject Headings] ,Drug Resistance, Neoplasm ,Cancer research ,FGFR4 ,Pyrazoles ,Chemicals and Drugs::Heterocyclic Compounds::Heterocyclic Compounds, 1-Ring::Piperazines [Medical Subject Headings] ,Carcinogenesis ,business ,Chemicals and Drugs::Organic Chemicals::Amides::Benzamides [Medical Subject Headings] ,Biomarkers ,Cadherinas - Abstract
[Background] Fibroblast growth factor receptor (FGFR)1 and FGFR4 have been associated with tumorigenesis in a variety of tumour types. As a therapeutic approach, their inhibition has been attempted in different types of malignancies, including lung cancer, and was initially focused on FGFR1-amplified tumours, though with limited success., [Methods] In vitro and in vivo functional assessments of the oncogenic potential of downregulated/overexpressed genes in isogenic cell lines were performed, as well as inhibitor efficacy tests in vitro and in vivo in patient-derived xenografts (PDXs). mRNA was extracted from FFPE non-small cell lung cancer samples to determine the prognostic potential of the genes under study., [Findings] We provide in vitro and in vivo evidence showing that expression of the adhesion molecule N-cadherin is key for the oncogenic role of FGFR1/4 in non-small cell lung cancer. According to this, assessment of the expression of genes in different lung cancer patient cohorts showed that FGFR1 or FGFR4 expression alone showed no prognostic potential, and that only co-expression of FGFR1 and/or FGFR4 with N-cadherin inferred a poorer outcome. Treatment of high-FGFR1 and/or FGFR4-expressing lung cancer cell lines and patient-derived xenografts with selective FGFR inhibitors showed high efficacy, but only in models with high FGFR1/4 and N-cadherin expression., [Interpretation] Our 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., This work was funded by the Community of Madrid, the ISCIII co-funded by FEDER from Regional Development European Funds (European Union), the Spanish Ministry of Economy and Competitiveness, the Mutua Madrileña Foundation, the Ministry of Health and Social Welfare of Junta de Andalucía, the AECC scientific foundation and the Spanish Ministry of Education, Culture and Sport.
- Published
- 2020
28. OA07.01 Signatures of Plasticity and Immunosuppression in a Single-Cell Atlas of Human Small Cell Lung Cancer
- Author
-
Vianne R. Gao, Linas Mazutis, Travis J. Hollmann, Yubin Xie, Joseph M. Chan, Dana Pe'er, Metamia Ciampricotti, Tal Nawy, V. Lai, Ignas Masilionis, David R. Jones, Matthew J. Bott, Ojasvi Chaudhary, Thomas Walle, Michael Offin, Triparna Sen, Andrew Chow, Álvaro Quintanal-Villalonga, Jacklynn V. Egger, Charles M. Rudin, and Viola Allaj
- Subjects
Pulmonary and Respiratory Medicine ,medicine.anatomical_structure ,Oncology ,Atlas (topology) ,business.industry ,medicine.medical_treatment ,Cell ,Cancer research ,medicine ,Immunosuppression ,Non small cell ,business - Published
- 2021
- Full Text
- View/download PDF
29. MA16.03 CRISPR Screen Reveals XPO1 as a Therapeutic Target Strongly Sensitizing to First and Second Line Therapy in Small Cell Lung Cancer
- Author
-
Yingqian Zhan, Joseph M. Chan, E. De Stanchina, Fathema Uddin, Andrew Chow, Jacklynn V. Egger, U. Bhanot, Viola Allaj, Álvaro Quintanal-Villalonga, Triparna Sen, John T. Poirier, Michael Offin, Yuan Hao, Hirokazu Taniguchi, P. Manoj, Shweta S. Chavan, Charles M. Rudin, J. Qiu, and Nisargbhai S. Shah
- Subjects
Pulmonary and Respiratory Medicine ,Second-line therapy ,XPO1 ,Oncology ,business.industry ,Cancer research ,Medicine ,CRISPR ,Non small cell ,business - Published
- 2021
- Full Text
- View/download PDF
30. 1MO Multi-omic characterization of lung tumors identify AKT and EZH2 as potential therapeutic targets in adenocarcinoma-to-squamous transdifferentiation
- Author
-
Yingqian Zhan, Nisargbhai S. Shah, J. Qiu, P. Manoj, Natasha Rekhtman, Hiroya Taniguchi, Viola Allaj, Metamia Ciampricotti, U. Bhanot, Charles M. Rudin, Brian Houck-Loomis, Maysun Hasan, Jacklynn V. Egger, Álvaro Quintanal-Villalonga, E. De Stanchina, Fathema Uddin, Richard Koche, Shweta S Chavan, Helena Alexandra Yu, and Triparna Sen
- Subjects
Lung ,business.industry ,EZH2 ,Transdifferentiation ,Hematology ,medicine.disease ,Omics ,medicine.anatomical_structure ,Oncology ,Cancer research ,Medicine ,Adenocarcinoma ,business ,Protein kinase B - Published
- 2021
- Full Text
- View/download PDF
31. MA11.06 Multi-Omic Characterization of Lung Tumors Implicates AKT and MYC Signaling in Adenocarcinoma to Squamous Cell Transdifferentiation
- Author
-
Charles M. Rudin, Helena Alexandra Yu, Jason C. Chang, Joseph M. Chan, Maysun Hasan, Brian Houck-Loomis, Andrew Chow, Álvaro Quintanal-Villalonga, Richard Koche, Triparna Sen, Jacklynn V. Egger, Yingqian Zhan, J. Qiu, M. Offin, P. Manoj, U. Bhanot, E. De Stanchina, Fathema Uddin, Viola Allaj, Nisargbhai S. Shah, Shweta S Chavan, Natasha Rekhtman, and Hiroya Taniguchi
- Subjects
Pulmonary and Respiratory Medicine ,Lung ,medicine.anatomical_structure ,Oncology ,business.industry ,Cell Transdifferentiation ,medicine ,Cancer research ,Adenocarcinoma ,medicine.disease ,business ,Protein kinase B - Published
- 2021
- Full Text
- View/download PDF
32. 2MO XPO1 inhibition strongly sensitizes to first-line and second-line therapy in small cell lung cancer
- Author
-
Travis J. Hollmann, E. De Stanchina, Fathema Uddin, Metamia Ciampricotti, Triparna Sen, Y. Hao, Jacklynn V. Egger, J. Qiu, Yingqian Zhan, John T. Poirier, Hirokazu Taniguchi, Richard Koche, Andrew Chow, Álvaro Quintanal-Villalonga, Viola Allaj, Joseph M. Chan, Michael Offin, Nisargbhai S. Shah, Charles M. Rudin, and P. Manoj
- Subjects
XPO1 ,Second-line therapy ,Oncology ,business.industry ,First line ,Cancer research ,Medicine ,Hematology ,Non small cell ,business - Published
- 2021
- Full Text
- View/download PDF
33. 1800O Multi-omic characterization of lung tumors implicates AKT and MYC signaling in adenocarcinoma to squamous cell transdifferentiation
- Author
-
Metamia Ciampricotti, Triparna Sen, Maysun Hasan, Jacklynn V. Egger, P. Manoj, Viola Allaj, Yingqian Zhan, Helena Alexandra Yu, E. De Stanchina, J. Qiu, Fathema Uddin, Joseph M. Chan, Nisargbhai S. Shah, Charles M. Rudin, Natasha Rekhtman, Shweta S. Chavan, U. Bhanot, Andrew Chow, Álvaro Quintanal-Villalonga, and Hirokazu Taniguchi
- Subjects
Lung ,medicine.anatomical_structure ,Oncology ,business.industry ,Cell Transdifferentiation ,medicine ,Cancer research ,Adenocarcinoma ,Hematology ,medicine.disease ,business ,Protein kinase B - Published
- 2021
- Full Text
- View/download PDF
34. Epigenetics of lung cancer: a translational perspective
- Author
-
Álvaro Quintanal-Villalonga, Sonia Molina-Pinelo, Junta de Andalucía, and European Commission
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Combination therapy ,Treatment of lung cancer ,Bioinformatics ,Epigenesis, Genetic ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Epigenetics ,Practice Patterns, Physicians' ,Lung cancer ,Survival rate ,Epigenomics ,Clinical Trials as Topic ,business.industry ,Precision oncology ,General Medicine ,medicine.disease ,Clinical trial ,030104 developmental biology ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Molecular Medicine ,Immunotherapy ,business - Abstract
[Background] Lung cancer remains the most common cause of cancer-related death, with a 5-year survival rate of only 18%. In recent years, the development of targeted pharmacological agents and immunotherapies has substantially increased the survival of a subset of patients. However, most patients lack such efficacious therapy and are, thus, treated with classical chemotherapy with poor clinical outcomes. Therefore, novel therapeutic strategies are urgently needed. In recent years, the development of epigenetic assays and their application to cancer research have highlighted the relevance of epigenetic regulation in the initiation, development, progression and treatment of lung cancer., [Conclusions] A variety of epigenetic modifications do occur at different steps of lung cancer development, some of which are key to tumor progression. The rise of cutting-edge technologies such as single cell epigenomics is, and will continue to be, crucial for uncovering epigenetic events at a single cell resolution, leading to a better understanding of the biology underlying lung cancer development and to the design of novel therapeutic options. This approach has already led to the development of strategies involving single agents or combined agents targeting epigenetic modifiers, currently in clinical trials. Here, we will discuss the epigenetics of every step of lung cancer development, as well as the translation of these findings into clinical applications., SMP was supported by the Consejería de Salud y Bienestar Social of Junta de Andalucía through the “‘Nicolás Monardes’ program [C-0040-2016] and ISCIII (PI17/00033) and cofounder by FEDER from Regional Development European Funds (European Union).”
- Published
- 2019
35. FGFR4 increases EGFR oncogenic signaling in lung adenocarcinoma, and their combined inhibition is highly effective
- Author
-
Amancio Carnero, Irene Ferrer, Ana Belén Enguita, Luis Paz-Ares, Rocío Suárez, Patricia Yagüe, Santiago Ponce-Aix, Laura Ojeda-Márquez, Sonia Molina-Pinelo, Álvaro Quintanal-Villalonga, Ángela Marrugal, Comunidad de Madrid, Instituto de Salud Carlos III, European Commission, Centro de Investigación Biomédica en Red Cáncer (España), Asociación Española Contra el Cáncer, Junta de Andalucía, Ministerio de Economía y Competitividad (España), Fundación Mutua Madrileña, and Ministerio de Educación, Cultura y Deporte (España)
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Treatment resistance ,Piperazines ,Cohort Studies ,Mice ,0302 clinical medicine ,Medicine ,Epidermal growth factor receptor ,EGFR inhibitors ,biology ,Drug Synergism ,ErbB Receptors ,Oncology ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Benzamides ,Adenocarcinoma ,Drug Therapy, Combination ,Female ,Signal Transduction ,Pulmonary and Respiratory Medicine ,EGFR ,Mice, Nude ,Adenocarcinoma of Lung ,Antineoplastic Agents ,Context (language use) ,Respiratory Mucosa ,Erlotinib Hydrochloride ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 4 ,Lung cancer ,Neoplasm Staging ,business.industry ,Fibroblast growth factor receptor 4 ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,TKIs ,Combination treatment ,Cancer research ,biology.protein ,FGFR4 ,Pyrazoles ,business - Abstract
[Objectives] Lung adenocarcinoma accounts for approximately half of lung cancer cases. Twenty to 50% of tumors of this type harbor mutations affecting epidermal growth factor receptor (EGFR) expression or activity, which can be therapeutically targeted. EGFR inhibitors in this context exhibit high efficacy and are currently used in the clinical setting. However, not all adenocarcinomas harboring EGFR mutations respond to therapy, so predictive biomarkers of therapeutic outcomes, as well as novel therapies sensitizing these tumors to EGFR inhibition, are needed., [Materials and methods] We performed in vitro gene overexpression/silencing and tumorigenic surrogate assays, as well as in vitro and in vivo combination treatments with Fibroblast Growth Factor Receptor (FGFR)/EGFR inhibitors. At the clinical level, we determined FGFR4 expression levels in tumors from patients treated with EGFR inhibitors and correlated these with treatment response., [Results] We describe a cooperative interaction between EGFR and FGFR4, which results in their reciprocal activation with pro-oncogenic consequences in vitro and in vivo. This cooperation is independent of EGFR activating mutations and increases resistance to different EGFR inhibitors. At the therapeutic level, we provide evidence of the synergistic effects of the combination of EGFR and FGFR inhibitors in high FGFR4-expressing, EGFR-activated tumors in vitro and in vivo. Correlated with these results, we found that patients treated with EGFR inhibitors relapse earlier when their tumors exhibit high FGFR4 expression., [Conclusions] We propose a novel predictive biomarker for EGFR-targeted therapy, and a highly efficacious combinatory therapeutic strategy to treat EGFR-dependent; this may may extend the use of appropriate inhibitors beyond EGFR-mutated adenocarcinoma patients., L.P.A. was funded by the Comunidad de Madrid, CAM, (B2017/BMD3884), ISCIII (PI14/01964, PIE15/00076, PI17/00778 and DTS17/00089) and CIBERONC (CD16/12/00442), and co-funded by FEDER from Regional Development European Funds (European Union). I.F. is funded by the AECC (AIO2015) and Consejería de Igualdad, Salud y Políticas Sociales de la Junta de Andalucía (PI-0029-2013) and ISCIII (PI16/01311), and co-funded by FEDER from Regional Development European Funds (European Union). AC was supported by grants from the Spanish Ministry of Economy and Competitiveness Plan Estatal de I + D+I 2013–2016, ISCIII (PI15/00045) and CIBERONC (CD16/12/00275), and co-funded by FEDER from Regional Development European Funds (European Union). S.M.P. is funded by the Consejería de Salud y Bienestar Social (PI-0046-2012), the Fundación Mutua Madrileña (2014) and ISCIII (PI17/00033). A.Q is funded by the ISCIII (FI12/00429). L.O. is funded by the Ministerio de Educación, Cultura y Deporte (FPU13/02595).
- Published
- 2019
36. Concurrent RB1 and TP53 Alterations Define a Subset of EGFR-Mutant Lung Cancers at risk for Histologic Transformation and Inferior Clinical Outcomes
- Author
-
Alexander V Penson, Gregory J. Riely, Álvaro Quintanal-Villalonga, Marc Ladanyi, Megan Tenet, Mark G. Kris, Matthew D. Hellmann, Yahya Daneshbod, Joseph M. Chan, Hira Rizvi, Maria E. Arcila, Helena A. Yu, Charles M. Rudin, Dana Pe'er, Michael Offin, Ronglai Shen, and Natasha Rekhtman
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Adult ,Male ,Lung Neoplasms ,Ubiquitin-Protein Ligases ,Cell ,Population ,Somatic hypermutation ,Adenocarcinoma of Lung ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,medicine ,Biomarkers, Tumor ,Humans ,education ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,education.field_of_study ,Lung ,business.industry ,Kinase ,Incidence (epidemiology) ,Cytidine deaminase ,Middle Aged ,Prognosis ,Small Cell Lung Carcinoma ,respiratory tract diseases ,Discontinuation ,ErbB Receptors ,Survival Rate ,Retinoblastoma Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,business ,Follow-Up Studies - Abstract
Introduction EGFR-mutant lung cancers are clinically and genomically heterogeneous with concurrent RB transcriptional corepressor 1 (RB1)/tumor protein p53 (TP53) alterations identifying a subset at increased risk for small cell transformation. The genomic alterations that induce lineage plasticity are unknown. Methods Patients with EGFR/RB1/TP53-mutant lung cancers, identified by next-generation sequencing from 2014 to 2018, were compared to patients with untreated, metastatic EGFR-mutant lung cancers without both RB1 and TP53 alterations. Time to EGFR–tyrosine kinase inhibitor discontinuation, overall survival, SCLC transformation rate, and genomic alterations were evaluated. Results Patients with EGFR/RB1/TP53-mutant lung cancers represented 5% (43 of 863) of EGFR-mutant lung cancers but were uniquely at risk for transformation (7 of 39, 18%), with no transformations in EGFR-mutant lung cancers without baseline TP53 and RB1 alterations. Irrespective of transformation, patients with EGFR/TP53/RB1-mutant lung cancers had a shorter time to discontinuation than EGFR/TP53- and EGFR-mutant –only cancers (9.5 versus 12.3 versus 36.6 months, respectively, p = 2 × 10-9). The triple-mutant population had a higher incidence of whole-genome doubling compared to NSCLC and SCLC at large (80% versus 34%, p Conclusions EGFR/TP53/RB1-mutant lung cancers are at unique risk of histologic transformation, with 25% presenting with de novo SCLC or eventual small cell transformation. Triple-mutant lung cancers are enriched in whole-genome doubling and Activation-induced cytidine deaminase/apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like hypermutation which may represent early genomic determinants of lineage plasticity.
- Published
- 2019
37. Direct genome editing of patient-derived xenografts using CRISPR-Cas9 enables rapid in vivo functional genomics
- Author
-
Christopher H. Hulton, Emily A. Costa, Elisa de Stanchina, Álvaro Quintanal-Villalonga, John T. Poirier, Nisargbhai S. Shah, Charles M. Rudin, and Glenn Heller
- Subjects
Cancer Research ,0303 health sciences ,Cas9 ,Computational biology ,Biology ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Genome editing ,In vivo ,030220 oncology & carcinogenesis ,Genetically Engineered Mouse ,CRISPR ,Gene ,Functional genomics ,Human cancer ,030304 developmental biology - Abstract
Patient-derived xenografts (PDXs) constitute a powerful set of preclinical models for in vivo cancer research, reflecting the spectrum of genomic alterations and therapeutic liabilities of human cancers1-4. In contrast to either cancer cell lines or genetically engineered mouse models, the utility of PDXs has been limited by the inability to perform targeted genome editing of these tumors. To address this limitation, we have generated a lentiviral platform for CRISPR-Cas9 editing of PDXs using a tightly regulated, inducible Cas9 vector that does not require in vitro culture for selection of transduced cells. We demonstrate the utility of this platform in PDXs (1) to analyze genetic dependencies by targeted gene disruption and (2) to analyze mechanisms of acquired drug resistance by site-specific gene editing using templated homology-directed repair. This flexible system has broad application to other explant models and substantially augments the utility of PDXs as genetically programmable models of human cancer.
- Published
- 2019
- Full Text
- View/download PDF
38. FGFR1 Cooperates with EGFR in Lung Cancer Oncogenesis, and Their Combined Inhibition Shows Improved Efficacy
- Author
-
Ángela Marrugal, Irene Ferrer, Rocío Suárez, Sonia Molina-Pinelo, Santiago Ponce-Aix, Álvaro Quintanal-Villalonga, Ana Belén Enguita, Laura Ojeda-Márquez, Luis Paz-Ares, Esther Conde, Amancio Carnero, Cristina Cirauqui, and Instituto de Biomedicina de Sevilla (IBIS)
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,Carcinogenesis ,Piperazines ,Mice ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,EGFR inhibitors ,Aniline Compounds ,Drug Synergism ,ErbB Receptors ,Combined inhibition ,Oncology ,030220 oncology & carcinogenesis ,Benzamides ,Adenocarcinoma ,Female ,Erlotinib ,Tyrosine kinase ,medicine.drug ,Pulmonary and Respiratory Medicine ,FGFR Inhibition ,EGFR ,Mice, Nude ,Transfection ,03 medical and health sciences ,Erlotinib Hydrochloride ,Gefitinib ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Lung cancer ,Protein Kinase Inhibitors ,Acrylamides ,business.industry ,Phenylurea Compounds ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Enzyme Activation ,stomatognathic diseases ,Cooperation ,030104 developmental biology ,Pyrimidines ,FGFR1 ,Cancer research ,Pyrazoles ,business - Abstract
Introduction: There is substantial evidence for the onco- genic effects of fi broblast growth factor receptor 1 (FGFR1) in many types of cancer, including lung cancer, but the role of this receptor has not been addressed speci fi cally in lung adenocarcinoma. Methods: We performed FGFR1 and EGFR overexpression and co-overexpression assays in adenocarcinoma and in inmortalized lung cell lines, and we also carried out surrogateandinteractionassays.Weperformedmono- therapy and combination EGFR /FGFR inhibitor sensitivity assays in vitro and in vivo in cell line – and patient- derived xenografts. We determined FGFR1 mRNA expression in a cohort of patients with anti – EGFR ther- apy – treated adenocarcinoma. Results: We have reported a cooperative interaction between FGFR1 and EGFR in this context, resulting in increased EGFR activation and oncogenic signaling. We have provided in vitro and in vivo evidence indicating that FGFR1 expression in- creases tumorigenicity in cells with high EGFR activation in EGFR-mutated and EGFR wild-type models. At the clinical level, we have shown that high FGFR1 expression levels pre- dict higher resistance to erlotinib or ge fi tinib in a cohort of patients with tyrosine kinase inhibitor – treated EGFR-mutated and EGFR wild-type lung adenocarcinoma. Dual EGFR and FGFR inhibition in FGFR1-over expressing, EGFR-activated models shows synergistic effects on tumor growth in vitro and in cell line – and patient-derived xenografts, suggesting that patients with tumors bearing these characteristics may bene fi t from combined EGFR/FGFR inhibition. Conclusion: These results support the extended the use of EGFR inhibitors beyond monotherapy in the EGFR-mutated adenocarcinoma setting in combination with FGFR in- hibitors for selected patients with increased FGFR1 over- expression and EGFR activation. ISCIII PI14/01964 PIE15/00076 PI17/00778 DTS17/00089 PI15/00045 PI17/00033 PI16/01311 FI12/00429 CIBERONC CD16/12/00442 FEDER CD16/12/00442 PI16/01311 Spanish Ministry of Economy and Competitiveness PI15/00045 Ministry of Health and Social Welfare of Junta de Andalucía PI-0046-2012 C-0040-2016 Ministry of Equality, Health and Social Policies of the Junta de Andalucía PI- 0029-2013 Comunidad de Madrid B2017/BMD3884 Ministry of Education, Culture and Sports FPU13/02595
- Published
- 2019
39. A patent review of FGFR4 selective inhibition in cancer (2007-2018)
- Author
-
Irene Ferrer, Luis Paz-Ares, Álvaro Quintanal-Villalonga, Sonia Molina-Pinelo, Instituto de Salud Carlos III, European Commission, Centro de Investigación Biomédica en Red Cáncer (España), Roche, Fundación Lilly, MSD, BMS College of Engineering, AstraZeneca, Boehringer Ingelheim Fonds, Pfizer, Takeda Pharmaceutical Company, Novartis, Merck Serono, Amgem, Asociación Española Contra el Cáncer, and Junta de Andalucía
- Subjects
Carcinoma, Hepatocellular ,Inhibitor ,Hepatocellular carcinoma ,Antineoplastic Agents ,Apoptosis ,Selective inhibition ,01 natural sciences ,Receptor tyrosine kinase ,Patents as Topic ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 4 ,Cell Proliferation ,Cancer ,Pharmacology ,biology ,Chemistry ,Liver Neoplasms ,General Medicine ,Fibroblast growth factor receptor 4 ,medicine.disease ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,030220 oncology & carcinogenesis ,Drug Design ,Cancer research ,biology.protein ,FGFR4 - Abstract
[Introduction] FGFR4 is a tyrosine kinase receptor which, under physiological conditions, is activated upon ligand binding in a highly regulated manner. This triggers downstream signaling related to proliferation and apoptosis resistance as well as other physiological processes. Many molecular alterations of the receptor and its ligands, specially FGF19, have been reported in several types of cancer, with special relevance in hepatocellular carcinoma. In addition, these have also been detected in other solid malignancies, including lung, breast, or colon cancer, among others., [Areas covered] This review covers patent literature on specific FGFR4 inhibitors and their applications, published from 2007 to June 2018., [Expert opinion] FGFR4 inhibition has gained relevance in oncology. A considerable number of patents disclosing different approaches to inhibit this receptor have been reported, displaying promising preclinical results for different cancer models. Currently, the safety and preliminary efficacy of several small molecule inhibitors targeting FGFR4 are under early phase clinical assessment, mainly in hepatocellular carcinoma patients. If positive results are derived from these trials, they will open the door for the application of FGFR4 small molecule inhibitors to a wide population of tumors of different types that harbor FGFR4-FGF19 signaling dysregulation., [Trial registration] ClinicalTrials.gov identifier: NCT02325739., [Trial registration] ClinicalTrials.gov identifier: NCT02834780., [Trial registration] ClinicalTrials.gov identifier: NCT03144661., [Trial registration] ClinicalTrials.gov identifier: NCT02508467., L Paz Ares was supported by ISCIII (PI14/01964, PIE15/00076, PI17/00778 and DTS17/00089) and CIBERONC (CD16/12/00442), and co-funded by FEDER from Regional Development European Funds (European Union). L Paz Ares also reports personal fees from Roche, Lilly, MSD, BMS, Astra Zeneca, Boehringer Ing., Pfizer, Takeda, Novartis, Merck Serono and Amgem, outside the submitted work. In addition, L Paz Ares has patent P201730928 and patent PCT/ES2018/070502 pending. I Ferrer was supported by AECC (AIO2015) and Consejería de Igualdad, Salud y Políticas Sociales de la Junta de Andalucía (PI-0029-2013) and ISCIII (PI16/01311) and cofunded by FEDER from Regional Development European Funds (European Union). Also, I Ferrer has patent P201730928 and patent PCT/ES2018/070502 pending. S Molina-Pinelo was supported by the Consejería de Salud y Bienestar Social of Junta de Andalucía through the ‘“Nicolás Monardes”’ program [C-0040-2016] and ISCIII (PI17/00033) and co-funded by FEDER from Regional Development European Funds (European Union). Also, S Molina-Pinelo has patent P201730928 and patent PCT/ES2018/070502 pending.
- Published
- 2019
40. Impact of Heat Shock Protein 90 Inhibition on the Proteomic Profile of Lung Adenocarcinoma as Measured by Two-Dimensional Electrophoresis Coupled with Mass Spectrometry
- Author
-
Álvaro Quintanal-Villalonga, Maria Dolores Pastor, Ángela Marrugal, Laura Ojeda, Irene Ferrer, Amancio Carnero, Sonia Molina-Pinelo, Luis Paz-Ares, Comunidad de Madrid, European Regional Development Fund, Fundación Mutua Madrileña, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (España), European Commission, Junta de Andalucía, Asociación Española Contra el Cáncer, Ministerio de Economía y Competitividad (España), Fundación Científica Asociación Española Contra el Cáncer, and Ministerio de Educación, Cultura y Deporte (España)
- Subjects
0301 basic medicine ,Proteomics ,Lung Neoplasms ,Lactams, Macrocyclic ,Adenocarcinoma of Lung ,Article ,Mass Spectrometry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Heat shock protein ,Cell Line, Tumor ,Chaperones ,medicine ,Benzoquinones ,Anaplastic lymphoma kinase ,chaperones ,Humans ,Electrophoresis, Gel, Two-Dimensional ,HSP90 Heat-Shock Proteins ,Protein Interaction Maps ,lcsh:QH301-705.5 ,proteomic ,HSP90 inhibitors ,Proteomic Profile ,biology ,Proteomic ,General Medicine ,Resorcinols ,Geldanamycin ,medicine.disease ,Hsp90 ,Hsp70 ,Gene Expression Regulation, Neoplastic ,lung cancer ,030104 developmental biology ,Biochemistry ,chemistry ,lcsh:Biology (General) ,A549 Cells ,030220 oncology & carcinogenesis ,Chaperone (protein) ,biology.protein ,Adenocarcinoma ,Lung cancer - Abstract
Heat shock protein 90 (HSP90) is an important chaperone in lung adenocarcinoma, with relevant protein drivers such as EGFR (epidermal growth factor receptor) and EML4-ALK (echinoderm microtubule-associated protein-like protein4 fused to anaplastic lymphoma kinase) depending on it for their correct function, therefore HSP90 inhibitors show promise as potential treatments for lung adenocarcinoma. To study responses to its inhibition, HSP90 was pharmacologically interrupted by geldanamycin and resorcinol derivatives or with combined inhibition of HSP90 plus HSP70 in lung adenocarcinoma cell lines. Two-dimensional electrophoresis was performed to identify proteomic profiles associated with inhibition which will help to understand the biological basis for the responses. HSP90 inhibition resulted in altered protein profiles that differed according the treatment condition studied. Results revealed 254 differentially expressed proteins after treatments, among which, eukaryotic translation initiation factor3 subunit I (eIF3i) and citrate synthase demonstrated their potential role as response biomarkers. The differentially expressed proteins also enabled signalling pathways involved in responses to be identified; these included apoptosis, serine-glycine biosynthesis and tricarboxylic acid cycle. The proteomic profiles identified here contribute to an improved understanding of HSP90 inhibition and open possibilities for the detection of potential response biomarkers which will be essential to maximize treatment efficacy in lung adenocarcinoma., L.P.A. was funded by the Comunidad de Madrid, CAM, (B2017/BMD3884), ISCIII (PIE15/00076, PI17/00778 and DTS17/00089) and CIBERONC (CB16/12/00442), and co-funded by FEDER from Regional Development European Funds (European Union). S.M.P. is funded by the Fundación Mutua Madrileña (2014) Ministry of Health and Social Welfare of Junta de Andalucía (PI-0046-2012, Nicolas Monardes Program C-0040-2016), ISCIII (PI17/00033), and co-funded by FEDER from Regional Development European Funds (European Union). I.F. is funded by the AECC (AIO2015) and ISCIII (PI16/01311), and co-funded by FEDER from Regional Development European Funds (European Union). AC was funded by grants from the Spanish Ministry of Economy and Competitiveness Plan Estatal de I+D+I 2018 co-funded by FEDER: RTI2018-097455-B-I00; CIBER de Cáncer (CB16/12/00275), co-funded by FEDER from Regional Development European Funds. Especial thanks to the Fundación AECC. L.O. is funded by the Ministerio de Educación, Cultura y Deporte (FPU13/02595).
- Published
- 2019
41. Signatures of plasticity and immunosuppression in a single-cell atlas of human small cell lung cancer
- Author
-
Charles M. Rudin, Jacklynn V. Egger, Thomas Walle, Travis Hollman, Matthew J. Bott, Marissa Mattar, Tal Nawy, Michael Offin, Dana Pe'er, Vianne R. Gao, Linas Mazutis, W. Victoria Victoria Lai, Ojasvi Chaudhary, Triparna Sen, Andrew Chow, Álvaro Quintanal-Villalonga, Joseph M. Chan, Viola Allaj, and Ignas Masilionis
- Subjects
Cancer Research ,Atlas (topology) ,business.industry ,medicine.medical_treatment ,Cell ,Immunosuppression ,Malignancy ,medicine.disease ,respiratory tract diseases ,ASCL1 ,medicine.anatomical_structure ,Oncology ,NEUROD1 ,medicine ,Cancer research ,Non small cell ,Differential expression ,business ,neoplasms - Abstract
8509 Background: Small cell lung cancer (SCLC) is an aggressive malignancy that includes subtypes defined by differential expression of ASCL1, NEUROD1, and POU2F3 (SCLC-A, -N, and -P, respectively), which are associated with distinct therapeutic vulnerabilities. The emerging consensus on SCLC subtypes has led to new questions, such as whether subtypes are associated with different disease stages, metastatic potential, or immune microenvironments; whether there is plasticity between subtypes; and whether novel SCLC phenotypes exist. Single cell RNA sequencing (scRNA-seq) offers a unique opportunity to address these questions by dissecting intratumoral transcriptional heterogeneity and the surrounding tumor microenvironment (TME). However, efforts to apply this technology to human SCLC tumors have been limited, as these tumors are infrequently resected. Methods: We have optimized protocols to process both surgical resections and biopsies to construct the first single-cell atlas of SCLC patient tumors (N = 21), with comparative lung adenocarcinoma (LUAD) and normal lung data. We leverage computational methods including diffusion maps and non-negative matrix factorization to perform a deep annotation of SCLC phenotypes and the surrounding immune TME. We perform validation experiments using flow cytometry, Vectra, and immunohistochemistry in independent SCLC cohorts, as well as genetic manipulation in preclinical SCLC models. Results: Our data reveals substantial transcriptional heterogeneity in SCLC both within and across tumors and confirms a pro-metastatic gene program in SCLC-N subtype characterized by epithelial-mesenchymal transformation and axonogenesis. Beyond known subtypes, we discover a PLCG2-high tumor cell population with stem-like, pro-metastatic features that recurs across subtypes and predicts significantly worse overall survival. Manipulation of PLCG2 expression in cells confirms correlation with key metastatic markers. Treatment and subtype are associated with substantial phenotypic changes in the SCLC immune microenvironment, with greater T-cell dysfunction in SCLC-N than SCLC-A. Moreover, the recurrent, PLCG2-high subclone is associated with exhausted CD8+ T-cells and a pro-fibrotic, immunosuppressive monocyte/macrophage population, suggesting possible tumor-immune coordination to promote metastasis. Conclusions: This atlas of SCLC illustrates how canonical subtypes and a novel PLCG2-high recurrent tumor subclone enlist diverse gene programs to create tumor heterogeneity and facilitate metastasis in a profoundly immunosuppressed TME. Our dataset provides further insight into tumor and immune biology in SCLC at single-cell resolution, with potential implications for design of novel targeted therapies and immunotherapeutic approaches.
- Published
- 2021
- Full Text
- View/download PDF
42. Publisher Correction: Lineage plasticity in cancer: a shared pathway of therapeutic resistance
- Author
-
Dana Pe'er, Sawyers Charles L, Triparna Sen, Álvaro Quintanal-Villalonga, Joseph M. Chan, Charles M. Rudin, and Helena A. Yu
- Subjects
Lineage (genetic) ,Oncology ,Tumour heterogeneity ,business.industry ,Medicine ,Cancer ,Computational biology ,Therapeutic resistance ,business ,medicine.disease - Published
- 2020
- Full Text
- View/download PDF
43. MAP17 predicts sensitivity to platinum-based therapy, EGFR inhibitors and the proteasome inhibitor bortezomib in lung adenocarcinoma
- Author
-
José M. García-Heredia, Luis Paz-Ares, Amancio Carnero, Sonia Molina-Pinelo, Rocío Suárez, Santiago Ponce-Aix, Marco Perez, Álvaro Quintanal-Villalonga, Irene Ferrer, Ministerio de Economía y Competitividad (España), Centro de Investigación Biomedica en Red - CIBER, Asociación Española Contra el Cáncer, Instituto de Salud Carlos III, Regional Government of Andalusia (España), and Universidad de Sevilla. Departamento de Bioquímica Vegetal y Biología Molecular
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Lung Neoplasms ,PDZK1IP1 ,Kaplan-Meier Estimate ,Bortezomib ,chemistry.chemical_compound ,0302 clinical medicine ,EGFR inhibitors ,Aged, 80 and over ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Prognosis ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Female ,Lung cancer ,Proteasome Inhibitors ,medicine.drug ,Adult ,Context (language use) ,Adenocarcinoma of Lung ,lcsh:RC254-282 ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Humans ,Protein Kinase Inhibitors ,Aged ,Cisplatin ,business.industry ,Research ,Membrane Proteins ,medicine.disease ,Xenograft Model Antitumor Assays ,Carboplatin ,030104 developmental biology ,chemistry ,Treatment efficacy ,Proteasome inhibitor ,Cancer research ,Receptor, Epidermal Growth Factor ,business ,Biomarkers - Abstract
BACKGROUND: The high incidence and mortality of lung tumours is a major health problem. Therefore, the identification both of biomarkers predicting efficacy for therapies in use and of novel efficacious therapeutic agents is crucial to increase patient survival. MAP17 (PDZK1IP1) is a small membrane-bound protein whose upregulation is reported as a common feature in tumours from diverse histological origins. Furthermore, MAP17 is correlated with tumour progression. METHODS: We assessed the expression of MAP17 in preclinical models, including cell lines and patient-derived xenografts (PDXs), assessing its correlation with sensitivity to different standard-of-care drugs in lung adenocarcinoma, as well as novel drugs. At the clinical level, we subsequently correlated MAP17 expression in human tumours with patient response to these therapies. RESULTS: We show that MAP17 expression is induced during lung tumourigenesis, particularly in lung adenocarcinomas, and provide in vitro and in vivo evidence that MAP17 levels predict sensitivity to therapies currently under clinical use in adenocarcinoma tumours, including cisplatin, carboplatin and EGFR inhibitors. In addition, we show that MAP17 expression predicts proteasome inhibitor efficacy in this context and that bortezomib, an FDA-approved drug, may be a novel therapeutic approach for MAP17-overexpressing lung adenocarcinomas. CONCLUSIONS: Our results indicate a potential prognostic role for MAP17 in lung tumours, with particular relevance in lung adenocarcinomas, and highlight the predictive pot0065ntial of this membrane-associated protein for platinum-based therapy and EGFR inhibitor efficacy. Furthermore, we propose bortezomib treatment as a novel and efficacious therapy for lung adenocarcinomas exhibiting high MAP17 expression. The authors thank the donors, the HUVR-IBiS Biobank (Andalusian Public Health System Biobank and ISCIII-Red de Biobancos PT13/0010/0056), and the Hospital 12 de Octubre Biobank for the human specimens used in this study. Sí
- Published
- 2018
44. Histology-dependent prognostic role of pERK and p53 protein levels in early-stage non-small cell lung cancer
- Author
-
Luis Paz-Ares, Rocío Suárez, Sonia Molina-Pinelo, Irene Ferrer, Mariló Mediano, Andrés Carranza-Carranza, Álvaro Quintanal-Villalonga, Ricardo Melendez, Amancio Carnero, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Fundación Mutua Madrileña, European Regional Development Fund, Asociación Española Contra el Cáncer, and Junta de Andalucía
- Subjects
0301 basic medicine ,Oncology ,p53 ,medicine.medical_specialty ,Context (language use) ,medicine.disease_cause ,NSCLC ,03 medical and health sciences ,0302 clinical medicine ,pERK ,Internal medicine ,medicine ,Stage (cooking) ,Lung cancer ,business.industry ,biomarkers ,Histology ,medicine.disease ,030104 developmental biology ,Clinical research ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Immunohistochemistry ,Carcinogenesis ,business ,prognostic ,Research Paper - Abstract
Lung tumors represent a major health problem. In early stage NSCLC tumors, surgical resection is the preferred treatment, but 30-55% of patients will relapse within 5 years after surgery. Thus, the identification of prognostic biomarkers in early stage NSCLC patients, especially those which are therapeutically addressable, is crucial to enhance survival of these patients. We determined the immunohistochemistry expression of key proteins involved in tumorigenesis and oncogenic signaling, p53, EGFR, pAKT and pERK, and correlated their expression level to clinicopathological characteristics and patient outcome. We found EGFR expression is higher in the squamous cell carcinomas than in adenocarcinomas (p=0.043), and that nuclear p53 staining correlated with lower differentiated squamous tumors (p=0.034). Regarding the prognostic potential of the expression of these proteins, high pERK levels proved to be an independent prognostic factor for overall (p, The authors thank the donors and the HUVR-IBiS Biobank (Andalusian Public Health System Biobank and ISCIII-Red de Biobancos PT13/0010/0056) for the human specimens used in this study.
- Published
- 2017
45. Coordinated downregulation of Spinophilin and the catalytic subunits of PP1, PPP1CA/B/C, contributes to a worse prognosis in lung cancer
- Author
-
Blanca Felipe-Abrio, Sandra Muñoz-Galván, Álvaro Quintanal-Villalonga, Luis M. Montuenga, Lola E. Navas, Luis Paz-Ares, Juan J. Marin, Irene Ferrer, Amancio Carnero, José M. García-Heredia, Sonia Molina-Pinelo, Eva M. Verdugo-Sivianes, Javier Peinado, Ministerio de Economía y Competitividad (España), European Commission, Junta de Andalucía, Instituto de Salud Carlos III, Asociación Española Contra el Cáncer, Fundación BBVA, Centro de Investigación Biomédica en Red Cáncer (España), [Verdugo-Sivianes, Eva M.] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Navas, Lola] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Molina-Pinelo, Sonia] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Peinado, Javier] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Garcia-Heredia, Jose M.] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Felipe-Abrio, Blanca] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Munoz-Galvan, Sandra] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Marin, Juan J.] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Carnero, Amancio] Univ Seville, Inst Biomed Sevilla IBIS, Hosp Univ Virgen del Rocio, CSIC, Seville, Spain, [Verdugo-Sivianes, Eva M.] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Navas, Lola] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Molina-Pinelo, Sonia] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Ferrer, Irene] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Garcia-Heredia, Jose M.] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Felipe-Abrio, Blanca] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Munoz-Galvan, Sandra] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Marin, Juan J.] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Montuenga, Luis] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Paz-Ares, Luis] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Carnero, Amancio] Inst Salud Carlos III, CIBER Canc, Pabellon 11,Planta 0, Madrid, Spain, [Ferrer, Irene] Inst Invest Hosp 12 Octubre, CNIO Lung Canc Clin Res Unit H120, Madrid, Spain, [Quintanal-Villalonga, Alvaro] Inst Invest Hosp 12 Octubre, CNIO Lung Canc Clin Res Unit H120, Madrid, Spain, [Paz-Ares, Luis] Inst Invest Hosp 12 Octubre, CNIO Lung Canc Clin Res Unit H120, Madrid, Spain, [Ferrer, Irene] CNIO, Madrid, Spain, [Quintanal-Villalonga, Alvaro] CNIO, Madrid, Spain, [Paz-Ares, Luis] CNIO, Madrid, Spain, [Peinado, Javier] Hosp Univ Virgen del Rocio, Radiat Oncol Dept, Seville, Spain, [Garcia-Heredia, Jose M.] Univ Seville, Dept Vegetal Biochem & Mol Biol, Seville, Spain, [Marin, Juan J.] Univ Seville, Dept Predict Med & Publ Hlth, Seville, Spain, [Montuenga, Luis] Ctr Appl Med Res CIMA, Program Solid Tumors & Biomarkers, Pamplona, Spain, Spanish Ministry of Economy and Competitivity, Plan Estatal de I+D+I, ISCIII, CIBER de Cancer, FEDER from Regional Development European Funds (European Union), Consejeria de Ciencia e Innovacion, Consejeria de Salud of the Junta de Andalucia, AECC Foundation, and Universidad de Sevilla. Departamento de Bioquímica Vegetal y Biología Molecular
- Subjects
Genetic-linkage analysis ,0301 basic medicine ,Scaffold protein ,Cells in-vitro ,Spinophilin ,Binding-protein ,Protein degradation ,Bioinformatics ,Tumor-suppressor spinophilin ,Dendritic spines ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Familial breast ,Lung cancer ,Breast-cancer ,therapy ,Bortezomib ,Chemistry ,Poor-prognosis ,medicine.disease ,PP1 ,Chromosome 17q12-21 ,lung cancer ,030104 developmental biology ,Oncology ,Retinoblastoma protein ,030220 oncology & carcinogenesis ,Cancer research ,Adenocarcinoma ,Biomarker (medicine) ,Phosphorylation ,biomarker ,Therapy ,Biomarkers ,Research Paper ,medicine.drug - Abstract
The scaffold protein Spinophilin (Spinophilin, PPP1R9B) is one of the regulatory subunits of phosphatase-1 (PP1), directing it to distinct subcellular locations and targets. The loss of Spinophilin reduces PP1 targeting to pRb, thereby maintaining higher levels of phosphorylated pRb. Spinophilin is absent or reduced in approximately 40% of human lung tumors, correlating with the malignant grade. However, little is known about the relevance of the coordinated activity or presence of Spinophilin and its reported catalytic partners in the prognosis of lung cancer. In the present work, we show that the downregulation of Spinophilin, either by protein or mRNA, is related to a worse prognosis in lung tumors. This effect is more relevant in squamous cell carcinoma, SCC, than in adenocarcinoma. Downregulation of Spinophilin is related to a decrease in the levels of its partners PPP1CA/B/C, the catalytic subunits of PP1. A decrease in these subunits is also related to prognosis in SCC and, in combination with a decrease in Spinophilin, are markers of a poor prognosis in these tumors. The analysis of the genes that correlate to Spinophilin in lung tumors showed clear enrichment in ATP biosynthesis and protein degradation GO pathways. The analysis of the response to several common and pathway-related drugs indicates a direct correlation between the Spinophilin/PPP1Cs ratio and the response to oxaliplatin and bortezomib. This finding indicates that this ratio may be a good predictive biomarker for the activity of the drugs in these tumors with a poor prognosis., This work was funded by grants from the Spanish Ministry of Economy and Competitivity, Plan Estatal de I+D+I 2013-2016, ISCIII (Fis: PI15/00045) and CIBER de Cáncer (CB16/12/00275, CB16/12/00443, CB16/12/00442), co-funded by FEDER from Regional Development European Funds (European Union), Consejeria de Ciencia e Innovacion (CTS-1848) and Consejeria de Salud of the Junta de Andalucia (PI-0096-2014). Especial thanks to the AECC Foundation for also supporting this work. This work has also been made possible by the Fundacion BBVA.
- Published
- 2017
46. The FGFR4-388arg Variant Promotes Lung Cancer Progression by N-Cadherin Induction
- Author
-
Álvaro Quintanal-Villalonga, Ángela Marrugal, Amancio Carnero, Laura Ojeda-Márquez, Santiago Ponce-Aix, Sonia Molina-Pinelo, Irene Ferrer, Patricia Yagüe, Ana Salinas, Luis Paz-Ares, Instituto de Salud Carlos III, Centros de Investigación Biomédica en Red (España), Red Temática de Investigación Cooperativa en Cáncer (España), European Commission, Ministerio de Economía y Competitividad (España), Junta de Andalucía, Fundación Mutua Madrileña, Asociación Española Contra el Cáncer, Ministerio de Educación, Cultura y Deporte (España), Centro de Investigación Biomedica en Red - CIBER, and Regional Government of Andalusia (España)
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,lcsh:Medicine ,Vimentin ,medicine.disease_cause ,FACTOR RECEPTOR 4 ,STAT3 ,Mice ,0302 clinical medicine ,HEPATOCELLULAR-CARCINOMA ,Carcinoma, Non-Small-Cell Lung ,EPITHELIAL-MESENCHYMAL TRANSITION ,PROGNOSTIC-SIGNIFICANCE ,Medicine ,lcsh:Science ,Regulation of gene expression ,Mutation ,Multidisciplinary ,biology ,Nuclear Proteins ,Cadherins ,Prognosis ,Phenotype ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,SURVIVAL ,Disease Progression ,Heterografts ,Female ,Mitogen-Activated Protein Kinases ,STAT3 Transcription Factor ,EXTRAHEPATIC CHOLANGIOCARCINOMA ,Epithelial-Mesenchymal Transition ,Mice, Nude ,Article ,03 medical and health sciences ,Antigens, CD ,Cell Line, Tumor ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 4 ,Lung cancer ,business.industry ,Cadherin ,lcsh:R ,Twist-Related Protein 1 ,Cancer ,Fibroblast growth factor receptor 4 ,medicine.disease ,Survival Analysis ,FGFR4 GLY388ARG POLYMORPHISM ,030104 developmental biology ,CELLS ,biology.protein ,Cancer research ,lcsh:Q ,Snail Family Transcription Factors ,ARG(388) ALLELE ,business - Abstract
The FGFR4-388Arg variant has been related to poor prognosis in several types of cancer, including lung cancer. The mechanism underlying this association has not been addressed in detail in patients with this pathology. Here, we report that this FGFR4 variant induces MAPK and STAT3 activation and causes pro-oncogenic effects in NSCLC in vitro and in vivo. This variant induces the expression of EMT-related genes, such as N-cadherin, vimentin, Snail1 and Twist1. Indeed, the induction of N-cadherin protein expression by this variant is essential for its pro-tumorigenic role. The presence of the FGFR4-388Arg variant correlates with higher N-cadherin expression levels in clinical NSCLC samples and with poorer outcome in patients with FGFR expression. These results support the prognostic role of this FGFR variant in lung cancer and show that these effects may be mediated by the induction of N-cadherin expression and an EMT phenotype., L.P.A. was funded by ISCIII (PI14/01964 and PIE15/00076), CIBER (CB16/12/00442) and RTICC (R12/0036/0028) and co-funded by European Union (ERDF/ESF, “Investing in your future”). The laboratory of A.C. was supported by grants from the Spanish Ministry of Economy and Competitiveness, PN I+D+I 2008-2011, PE I+D+I 2013-2016, ISCIII (PI15/00045 and CB16/12/00275), Consejeria de Ciencia e Innovacion (CTS-1848) and Consejeria de Salud of the Junta de Andalucia (PI-0096-2014). S.M.P. is funded by Consejería de Salud y Bienestar Social (PI-0046-2012), ISCIII (PI17/00033) and co-funded by European Union (ERDF/ESF, “Investing in your future”), and Fundación Mutua Madrileña (2014). I.F. is funded by AECC (AIO2015) and Consejería de Igualdad, Salud y Políticas Sociales de la Junta de Andalucía (PI-0029-2013) and FIS (PI16/01311). A.Q. is funded by ISCIII (FI12/00429). L.O. is funded by Ministerio de Educación, Cultura y Deporte (FPU13/02595).
- Published
- 2017
47. Tyrosine Kinase Receptor Landscape in Lung Cancer: Therapeutical Implications
- Author
-
Luis Paz-Ares, Sonia Molina-Pinelo, Irene Ferrer, and Álvaro Quintanal-Villalonga
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,Clinical Biochemistry ,Receptor Protein-Tyrosine Kinases ,Disease ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Genetics ,medicine ,Biomarkers, Tumor ,Humans ,Lung cancer ,Molecular Biology ,Survival rate ,Protein Kinase Inhibitors ,lcsh:R5-920 ,biology ,business.industry ,Biochemistry (medical) ,Advanced stage ,General Medicine ,Omics ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,business ,lcsh:Medicine (General) ,Corrigendum - Abstract
Lung cancer is a heterogeneous disease responsible for the most cases of cancer-related deaths. The majority of patients are clinically diagnosed at advanced stages, with a poor survival rate. For this reason, the identification of oncodrivers and novel biomarkers is decisive for the future clinical management of this pathology. The rise of high throughput technologies popularly referred to as “omics” has accelerated the discovery of new biomarkers and drivers for this pathology. Within them, tyrosine kinase receptors (TKRs) have proven to be of importance as diagnostic, prognostic, and predictive tools and, due to their molecular nature, as therapeutic targets. Along this review, the role of TKRs in the different lung cancer histologies, research on improvement of anti-TKR therapy, and the current approaches to manage anti-TKR resistance will be discussed.
- Published
- 2016
48. IL-11 and CCL-1: Novel Protein Diagnostic Biomarkers of Lung Adenocarcinoma in Bronchoalveolar Lavage Fluid (BALF)
- Author
-
Sonia Molina-Pinelo, Luis Paz-Ares, Rocío García-Carboner, Ana Nogal, José Luis López-Campos, Maria Delores Pastor, Beatrice Romero-Romero, Irene Ferrer, Amancio Carnero, Ricardo Melendez, Álvaro Quintanal-Villalonga, Maria de Miguel, and Jesus Corral
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Male ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Adenocarcinoma of Lung ,Adenocarcinoma ,03 medical and health sciences ,Chemokine CCL1 ,0302 clinical medicine ,medicine ,Biomarkers, Tumor ,Humans ,Lung cancer ,Prospective cohort study ,Inflammation ,COPD ,Lung ,medicine.diagnostic_test ,biology ,business.industry ,Bronchoalveolar lavage fluid ,Biomarker ,medicine.disease ,Interleukin-11 ,respiratory tract diseases ,030104 developmental biology ,Bronchoalveolar lavage ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Screening ,Biomarker (medicine) ,Cytokines ,Female ,Antibody ,business ,Bronchoalveolar Lavage Fluid - Abstract
Lung cancer (LC) and chronic obstructive pulmonary disease (COPD) are smoking-related diseases, with the presence of COPD itself increasing the risk for development of LC, probably owing to underlying inflammation. LC is typically detected at late stages of the disease and carries a poor prognosis. There is an unmet need for methods to facilitate the early detection of LC in high-risk subjects such as smokers.The expression of inflammatory proteins in bronchoalveolar lavage fluid (BALF) samples was studied by antibody arrays in a prospective cohort of 60 smokers of more than 30 pack-years divided into four groups (control, patients with LC, patients with COPD, and patients with LC plus COPD). Relevant biomarkers were validated by Western blot. Additional validation with enzyme-linked immunosorbent assay (ELISA) was carried out on two independent controlled cohorts of 139 patients (control, patients with LC, patients with COPD, and patients with LC plus COPD) and 160 patients (control and patients with LC of all histological types).A total of 16 differentially expressed proteins in samples from patients with LC, COPD, and LC plus COPD were identified by antibody arrays and validated by Western blot and ELISA. C-C motif chemokine ligand 1 (CCL-1) and interleukin-11 (IL)-11 were selectively expressed in samples from patients with adenocarcinoma with or without COPD (p0.005). These proteins exhibited a remarkable diagnostic performance for lung adenocarcinoma in an independent cohort of 139 patients. Receiver operating characteristic curves showed that the optimum diagnostic cutoff value for IL-11 was 42 pg/mL (area under the curve = 0.93 [95% confidence interval: 0.896-0.975], sensitivity 90%, specificity 86%), whereas for CCL-1 it was 39.5 pg/mL (0.83 [95% confidence interval: 0.749-0.902], sensitivity 83%, and specificity 74%). Further validation of the ELISA biomarkers at the aforementioned cutoffs was performed in an additional cohort of 160 patients (20 controls, 66 patients with LC, and 74 patients with LC plus COPD). There was a significant correlation between BALF levels of IL-11 and CCL-1 (rIL-11 and CCL-1 are highly specific biomarkers with great accuracy for the diagnosis of lung adenocarcinoma in BALF specimens. Further study of these proteins as markers for the early diagnosis and screening of plasma and other biological materials is warranted.
- Published
- 2016
49. Corrigendum to 'Tyrosine Kinase Receptor Landscape in Lung Cancer: Therapeutical Implications'
- Author
-
Álvaro Quintanal-Villalonga, Luis Paz-Ares, Sonia Molina-Pinelo, and Irene Ferrer
- Subjects
0301 basic medicine ,lcsh:R5-920 ,biology ,business.industry ,Biochemistry (medical) ,Clinical Biochemistry ,Review Article ,General Medicine ,medicine.disease ,Receptor tyrosine kinase ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,030220 oncology & carcinogenesis ,Genetics ,biology.protein ,Cancer research ,Medicine ,lcsh:Medicine (General) ,business ,Lung cancer ,Molecular Biology - Abstract
Lung cancer is a heterogeneous disease responsible for the most cases of cancer-related deaths. The majority of patients are clinically diagnosed at advanced stages, with a poor survival rate. For this reason, the identification of oncodrivers and novel biomarkers is decisive for the future clinical management of this pathology. The rise of high throughput technologies popularly referred to as “omics” has accelerated the discovery of new biomarkers and drivers for this pathology. Within them, tyrosine kinase receptors (TKRs) have proven to be of importance as diagnostic, prognostic, and predictive tools and, due to their molecular nature, as therapeutic targets. Along this review, the role of TKRs in the different lung cancer histologies, research on improvement of anti-TKR therapy, and the current approaches to manage anti-TKR resistance will be discussed.
- Published
- 2018
- Full Text
- View/download PDF
50. Abstract 2589: Novel predictor of FGFR1 inhibition efficacy in non-small cell lung cancer
- Author
-
Álvaro Quintanal-Villalonga, Irene Ferrer, Ángela Marrugal, Laura Ojeda-Márquez, Jon Zugazagoita, Laura García-Redondo, Fernando López-Ríos, Luis Montuenga, Sonia Molina-Pinelo, Amancio Carnero, and Luis Paz-Ares
- Subjects
Cancer Research ,business.industry ,Fibroblast growth factor receptor 1 ,FGFR Inhibition ,Cell ,Cancer ,medicine.disease ,medicine.disease_cause ,stomatognathic diseases ,medicine.anatomical_structure ,Oncology ,In vivo ,Fibroblast growth factor receptor ,medicine ,Cancer research ,Lung cancer ,Carcinogenesis ,business - Abstract
Fibroblast growth factor receptor (FGFR)1 has been associated with tumorigenesis in a variety of tumors, including lung cancer. FGFR1 amplification is detected in up to 20% of lung squamous cell carcinomas, and was initially related to tumorigenesis and to response to FGFR inhibitors in preclinical models of this pathology. These data encouraged the establishment of FGFR1 amplification as the main eligibility criterion for inclusion of patients in FGFR inhibitors clinical trials. However, the use of FGFR inhibitors in FGFR1-amplified tumors has shown limited success, with only 5-10% of selected patients showing partial response, suggesting that this genomic aberration may not be a good predictive biomarker for the efficacy of these inhibitors. In this work, we provide in vitro and in vivo data showing that the oncogenic effects of FGFR1 expression depend on the expression of the adhesion molecule N-cadherin in non-small cell lung cancer (NSCLC). In line with these results, FGFR1 expression correlates with poorer prognosis only in tumors with high N-cadherin levels in two independent NSCLC patient cohorts. We observed that high FGFR1 expression alone is not sufficient to predict FGFR inhibition efficacy in lung cancer cell lines and patient-derived xenografts, with only high FGFR1- and high N-cadherin-expressing models responding to selective FGFR inhibitors. Altogether, our data show that the determination of the expression of FGFR1 alone is not sufficient to predict FGFR inhibition efficacy. The co-determination of N-cadherin expression may optimize patient selection for this therapeutic strategy. Citation Format: Álvaro Quintanal-Villalonga, Irene Ferrer, Ángela Marrugal, Laura Ojeda-Márquez, Jon Zugazagoita, Laura García-Redondo, Fernando López-Ríos, Luis Montuenga, Sonia Molina-Pinelo, Sonia Molina-Pinelo, Amancio Carnero, Luis Paz-Ares. Novel predictor of FGFR1 inhibition efficacy in non-small cell lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2589.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.