23 results on '"Redin E"'
Search Results
2. YES1 as a Possible Liquid Biopsy Biomarker in Lung Cancer
- Author
-
Otegui, N., Redin, E., Janthus-Lewintre, E., Calabuig, S., Echepare, M., Valencia, K., Leon, S., Serrano, M., Camps, C., Montuenga, L., and Calvo, A.
- Published
- 2023
- Full Text
- View/download PDF
3. PERCEPÇÃO DO BEM-ESTAR ANIMAL NA PECUÁRIA FAMILIAR DE LEITE EM UNAÍ, MINAS GERAIS /PERCEPTION OF ANIMAL WELFARE IN FAMILY DAIRY FARMING IN UNAÍ, MINAS GERAIS
- Author
-
Rezende, N.S., Amaral, R.R., Pereira, A.A., Mota, D.A., Redin, E., and Melo, T.V.
- Published
- 2021
- Full Text
- View/download PDF
4. FP03.04 Dasatinib Improves the Antitumor Activity of Anti-PD-1 in NSCLC Models by Inhibiting Treg Conversion and Proliferation
- Author
-
Redin, E., primary, Garmendia, I., additional, Lozano, T., additional, Serrano, D., additional, Senent, Y., additional, Redrado, M., additional, Villalba, M., additional, De Andrea, C.E., additional, Exposito, F., additional, Ajona, D., additional, Ortiz-Espinosa, S., additional, Remirez, A., additional, Bertolo, C., additional, Sainz, C., additional, García-Pedrero, J.M., additional, Pio, R., additional, Lasarte, J.J., additional, Agorreta, J., additional, Montuenga, L., additional, and Calvo, A., additional
- Published
- 2021
- Full Text
- View/download PDF
5. Cancer Epigenetic Biomarkers in Liquid Biopsy for High Incidence Malignancies
- Author
-
Palanca-Ballester C, Rodriguez-Casanova A, Torres S, Calabuig-Fariñas S, Exposito F, Serrano D, Redin E, Valencia K, Jantus-Lewintre E, Diaz-Lagares A, Montuenga L, Sandoval J, and Calvo A
- Subjects
DNA methylation ,micro-RNAs ,cancer ,epigenetic biomarkers - Abstract
Simple Summary Apart from genetic changes, cancer is characterized by epigenetic alterations, which indicate modifications in the DNA (such as DNA methylation) and histones (such as methylation and acetylation), as well as gene expression regulation by non-coding (nc)RNAs. These changes can be used in biological fluids (liquid biopsies) for diagnosis, prognosis and prediction of cancer drug response. Although these alterations are not widely used as biomarkers in the clinical practice yet, increasing number of commercial kits and clinical trials are expected to prove that epigenetic changes are able to offer valuable information for cancer patients. Early alterations in cancer include the deregulation of epigenetic events such as changes in DNA methylation and abnormal levels of non-coding (nc)RNAs. Although these changes can be identified in tumors, alternative sources of samples may offer advantages over tissue biopsies. Because tumors shed DNA, RNA, and proteins, biological fluids containing these molecules can accurately reflect alterations found in cancer cells, not only coming from the primary tumor, but also from metastasis and from the tumor microenvironment (TME). Depending on the type of cancer, biological fluids encompass blood, urine, cerebrospinal fluid, and saliva, among others. Such samples are named with the general term "liquid biopsy" (LB). With the advent of ultrasensitive technologies during the last decade, the identification of actionable genetic alterations (i.e., mutations) in LB is a common practice to decide whether or not targeted therapy should be applied. Likewise, the analysis of global or specific epigenetic alterations may also be important as biomarkers for diagnosis, prognosis, and even for cancer drug response. Several commercial kits that assess the DNA promoter methylation of single genes or gene sets are available, with some of them being tested as biomarkers for diagnosis in clinical trials. From the tumors with highest incidence, we can stress the relevance of DNA methylation changes in the following genes found in LB: SHOX2 (for lung cancer); RASSF1A, RARB2, and GSTP1 (for lung, breast, genitourinary and colon cancers); and SEPT9 (for colon cancer). Moreover, multi-cancer high-throughput methylation-based tests are now commercially available. Increased levels of the microRNA miR21 and several miRNA- and long ncRNA-signatures can also be indicative biomarkers in LB. Therefore, epigenetic biomarkers are attractive and may have a clinical value in cancer. Nonetheless, validation, standardization, and demonstration of an added value over the common clinical practice are issues needed to be addressed in the transfer of this knowledge from "bench to bedside".
- Published
- 2021
6. EP16.01-009 Two Novel Alternative Cell Line Models for the Study of Multiorganic Metastasis and Immunotherapy in Lung Squamous Cell Carcinoma
- Author
-
Valencia, K., Sainz, C., Redrado, M., Redín, E., Serrano, D., Echepare, M., Calvo, A., and Montuenga, L.M.
- Published
- 2022
- Full Text
- View/download PDF
7. P2.03-38 Identification of a Novel Synthetic Lethal Vulnerability in Non-Small Cell Lung Cancer by Co-Targeting TMPRSS4 and DDR1
- Author
-
Redin, E., primary, Villalba, M., additional, Exposito, F., additional, Pajares, M.J., additional, Sainz, C., additional, Hervas, D., additional, Guruceaga, E., additional, Diaz-Lagares, A., additional, Cirauqui, C., additional, Redrado, M., additional, De Andrea, C., additional, Jantus, E., additional, Camps, C., additional, López, R., additional, Lahoz, A., additional, Montuenga, L., additional, Pio, R., additional, Sandoval, J., additional, and Calvo, A., additional
- Published
- 2019
- Full Text
- View/download PDF
8. P1.03-24 TMPRSS4: A Novel Prognostic Biomarker and Therapeutic Target in NSCLC
- Author
-
Expósito, F., primary, Villalba, M., additional, Pajares, M.J., additional, Redrado, M., additional, Sainz, C., additional, Wistuba, I., additional, Behrens, C., additional, Redin, E., additional, Andrea, C., additional, Cirauquiz, C., additional, Montuenga, L., additional, Pio, R., additional, and Calvo, A., additional
- Published
- 2018
- Full Text
- View/download PDF
9. SATISFAÇÃO DE DISCENTES SOBRE A FORMAÇÃO TÉCNICA E PROFISSIONAL EM INSTITUIÇÕES PÚBLICA E PRIVADA (RIO GRANDE DO SUL, 2013).
- Author
-
REDIN, E., REIMCHE, G. B., LIXINSKI, G. M., STIVANIN, J. B., SILVA, E. I., and QUADROS, C.
- Abstract
The aim of the study was to evaluate the student satisfaction at the last semester of technical college in Administration and Accounting, and compare two institutions - public and private - of Santa Maria, Rio Grande do Sul, and the course chosen, care, training of teachers, teaching methods, attitude, content and infrastructure. Data were collected through a proposed modified Kano questionnaire. The results show that, in the private institution, attributes lab printing and modernity of computer labs have the greatest impact on student satisfaction. In public institution, the attribute lab printing and teacher updating are considered the greatest impact on satisfaction. In general terms, the Index of American Customer Satisfaction - ACSI -, considering the 19 attributes of this study shows that the private institution has an index of 51.6%, while the public shows a higher rate of 62.1%. Finally, the research shows a greater satisfaction of students in public technical institution, while in private institution sets up a duality between education, human resources and infrastructure. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
10. G9a/DNMT1 co-targeting inhibits non-small cell lung cancer growth and reprograms tumor cells to respond to cancer-drugs through SCARA5 and AOX1.
- Author
-
Exposito F, Redrado M, Serrano D, Calabuig-Fariñas S, Bao-Caamano A, Gallach S, Jantus-Lewintre E, Diaz-Lagares A, Rodriguez-Casanova A, Sandoval J, San Jose-Eneriz E, Garcia J, Redin E, Senent Y, Leon S, Pio R, Lopez R, Oyarzabal J, Pineda-Lucena A, Agirre X, Montuenga LM, Prosper F, and Calvo A
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Histocompatibility Antigens metabolism, Histocompatibility Antigens genetics, Female, Gene Expression Regulation, Neoplastic drug effects, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, DNA (Cytosine-5-)-Methyltransferase 1 genetics, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Lung Neoplasms genetics, Lung Neoplasms metabolism, Cell Proliferation drug effects, Histone-Lysine N-Methyltransferase metabolism, Histone-Lysine N-Methyltransferase genetics
- Abstract
The treatment of non-small cell lung cancer (NSCLC) patients has significantly improved with recent therapeutic strategies; however, many patients still do not benefit from them. As a result, new treatment approaches are urgently needed. In this study, we evaluated the antitumor efficacy of co-targeting G9a and DNMT1 enzymes and its potential as a cancer drug sensitizer. We observed co-expression and overexpression of G9a and DNMT1 in NSCLC, which were associated with poor prognosis. Co-targeting G9a/DNMT1 with the drug CM-272 reduced proliferation and induced cell death in a panel of human and murine NSCLC cell lines. Additionally, the transcriptomes of these cells were reprogrammed to become highly responsive to chemotherapy (cisplatin), targeted therapy (trametinib), and epigenetic therapy (vorinostat). In vivo, CM-272 reduced tumor volume in human and murine cell-derived cancer models, and this effect was synergistically enhanced by cisplatin. The expression of SCARA5 and AOX1 was induced by CM-272, and both proteins were found to be essential for the antiproliferative response, as gene silencing decreased cytotoxicity. Furthermore, the expression of SCARA5 and AOX1 was positively correlated with each other and inversely correlated with G9a and DNMT1 expression in NSCLC patients. SCARA5 and AOX1 DNA promoters were hypermethylated in NSCLC, and SCARA5 methylation was identified as an epigenetic biomarker in tumors and liquid biopsies from NSCLC patients. Thus, we demonstrate that co-targeting G9a/DNMT1 is a promising strategy to enhance the efficacy of cancer drugs, and SCARA5 methylation could serve as a non-invasive biomarker to monitor tumor progression., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. Correction: SMARCA4 controls state plasticity in small cell lung cancer through regulation of neuroendocrine transcription factors and REST splicing.
- Author
-
Redin E, Sridhar H, Zhan YA, Pereira Mello B, Zhong H, Durani V, Sabet A, Manoj P, Linkov I, Qiu J, Koche RP, de Stanchina E, Astorkia M, Betel D, Quintanal-Villalonga Á, and Rudin CM
- Published
- 2024
- Full Text
- View/download PDF
12. Targeting the mSWI/SNF complex in POU2F-POU2AF transcription factor-driven malignancies.
- Author
-
He T, Xiao L, Qiao Y, Klingbeil O, Young E, Wu XS, Mannan R, Mahapatra S, Redin E, Cho H, Bao Y, Kandarpa M, Ching-Yi Tien J, Wang X, Eyunni S, Zheng Y, Kim N, Zheng H, Hou S, Su F, Miner SJ, Mehra R, Cao X, Abbineni C, Samajdar S, Ramachandra M, Dhanasekaran SM, Talpaz M, Parolia A, Rudin CM, Vakoc CR, and Chinnaiyan AM
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Octamer Transcription Factor-3 metabolism, Octamer Transcription Factor-3 genetics, Xenograft Model Antitumor Assays, Signal Transduction, Gene Expression Regulation, Neoplastic, Octamer Transcription Factor-2, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma metabolism, Small Cell Lung Carcinoma pathology, Transcription Factors metabolism, Transcription Factors genetics
- Abstract
The POU2F3-POU2AF2/3 transcription factor complex is the master regulator of the tuft cell lineage and tuft cell-like small cell lung cancer (SCLC). Here, we identify a specific dependence of the POU2F3 molecular subtype of SCLC (SCLC-P) on the activity of the mammalian switch/sucrose non-fermentable (mSWI/SNF) chromatin remodeling complex. Treatment of SCLC-P cells with a proteolysis targeting chimera (PROTAC) degrader of mSWI/SNF ATPases evicts POU2F3 and its coactivators from chromatin and attenuates downstream signaling. B cell malignancies which are dependent on the POU2F1/2 cofactor, POU2AF1, are also sensitive to mSWI/SNF ATPase degraders, with treatment leading to chromatin eviction of POU2AF1 and IRF4 and decreased IRF4 signaling in multiple myeloma cells. An orally bioavailable mSWI/SNF ATPase degrader significantly inhibits tumor growth in preclinical models of SCLC-P and multiple myeloma without signs of toxicity. This study suggests that POU2F-POU2AF-driven malignancies have an intrinsic dependence on the mSWI/SNF complex, representing a therapeutic vulnerability., Competing Interests: Declaration of interests A.M.C. serves on the clinical advisory board of Aurigene Oncology Limited. C.A., S.S., and M.R. are employees of Aurigene Oncology Limited. C.R.V. has received consulting fees from Flare Therapeutics, Roivant Sciences, and C4 Therapeutics; he has served on the advisory boards of KSQ Therapeutics, Syros Pharmaceuticals, and Treeline Biosciences. C.R.V. has also received research funding from Boehringer-Ingelheim and Treeline Biosciences and owns stock in Treeline Biosciences. Aurigene Oncology Limited has filed patent applications on AU-15330 and AU-24118. C.M.R. has consulted regarding oncology drug development with AbbVie, Amgen, Astra Zeneca, D2G, Daiichi Sankyo, Epizyme, Genentech/Roche, Ipsen, Jazz, Kowa, Lilly, Merck, and Syros. C.M.R. serves on the scientific advisory boards of Auron, Bridge Medicines, DISCO, Earli, and Harpoon Therapeutics., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. SMARCA4 controls state plasticity in small cell lung cancer through regulation of neuroendocrine transcription factors and REST splicing.
- Author
-
Redin E, Sridhar H, Zhan YA, Pereira Mello B, Zhong H, Durani V, Sabet A, Manoj P, Linkov I, Qiu J, Koche RP, de Stanchina E, Astorkia M, Betel D, Quintanal-Villalonga Á, and Rudin CM
- Subjects
- Humans, Cell Line, Tumor, Animals, Mice, Gene Expression Regulation, Neoplastic, Repressor Proteins, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma metabolism, Small Cell Lung Carcinoma pathology, Small Cell Lung Carcinoma drug therapy, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Lung Neoplasms drug therapy, DNA Helicases genetics, Transcription Factors genetics, Transcription Factors metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism
- 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., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. CDC7 inhibition impairs neuroendocrine transformation in lung and prostate tumors through MYC degradation.
- Author
-
Quintanal-Villalonga A, Kawasaki K, Redin E, Uddin F, Rakhade S, Durani V, Sabet A, Shafer M, Karthaus WR, Zaidi S, Zhan YA, Manoj P, Sridhar H, Kinyua D, Zhong H, Mello BP, Ciampricotti M, Bhanot UK, Linkov I, Qiu J, Patel RA, Morrissey C, Mehta S, Barnes J, Haffner MC, Socci ND, Koche RP, de Stanchina E, Molina-Pinelo S, Salehi S, Yu HA, Chan JM, and Rudin CM
- Subjects
- Humans, Male, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cell Line, Tumor, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Mice, Animals, Neuroendocrine Tumors genetics, Neuroendocrine Tumors pathology, Neuroendocrine Tumors metabolism, Neuroendocrine Tumors drug therapy, Proteolysis drug effects, Retinoblastoma Binding Proteins genetics, Retinoblastoma Binding Proteins metabolism, Ubiquitin-Protein Ligases, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Prostatic Neoplasms metabolism, Prostatic Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Lung Neoplasms metabolism, Lung Neoplasms drug therapy, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism
- 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., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. PTEN Loss Confers Resistance to Anti-PD-1 Therapy in Non-Small Cell Lung Cancer by Increasing Tumor Infiltration of Regulatory T Cells.
- Author
-
Exposito F, Redrado M, Houry M, Hastings K, Molero-Abraham M, Lozano T, Solorzano JL, Sanz-Ortega J, Adradas V, Amat R, Redin E, Leon S, Legarra N, Garcia J, Serrano D, Valencia K, Robles-Oteiza C, Foggetti G, Otegui N, Felip E, Lasarte JJ, Paz-Ares L, Zugazagoitia J, Politi K, Montuenga L, and Calvo A
- Subjects
- Animals, Humans, Mice, B7-H1 Antigen metabolism, Immunotherapy methods, Phosphatidylinositol 3-Kinases metabolism, Tumor Microenvironment, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms metabolism, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, T-Lymphocytes, Regulatory, Drug Resistance, Neoplasm genetics
- Abstract
Immunotherapy resistance in non-small cell lung cancer (NSCLC) may be mediated by an immunosuppressive microenvironment, which can be shaped by the mutational landscape of the tumor. Here, we observed genetic alterations in the PTEN/PI3K/AKT/mTOR pathway and/or loss of PTEN expression in >25% of patients with NSCLC, with higher frequency in lung squamous carcinomas (LUSC). Patients with PTEN-low tumors had higher levels of PD-L1 and PD-L2 and showed worse progression-free survival when treated with immunotherapy. Development of a Pten-null LUSC mouse model revealed that tumors with PTEN loss were refractory to antiprogrammed cell death protein 1 (anti-PD-1), highly metastatic and fibrotic, and secreted TGFβ/CXCL10 to promote conversion of CD4+ lymphocytes into regulatory T cells (Treg). Human and mouse PTEN-low tumors were enriched in Tregs and expressed higher levels of immunosuppressive genes. Importantly, treatment of mice bearing Pten-null tumors with TLR agonists and anti-TGFβ antibody aimed to alter this immunosuppressive microenvironment and led to tumor rejection and immunologic memory in 100% of mice. These results demonstrate that lack of PTEN causes immunotherapy resistance in LUSCs by establishing an immunosuppressive tumor microenvironment that can be reversed therapeutically., Significance: PTEN loss leads to the development of an immunosuppressive microenvironment in lung cancer that confers resistance to anti-PD-1 therapy, which can be overcome by targeting PTEN loss-mediated immunosuppression., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
16. Cancer Cell-Intrinsic Alterations Associated with an Immunosuppressive Tumor Microenvironment and Resistance to Immunotherapy in Lung Cancer.
- Author
-
Otegui N, Houry M, Arozarena I, Serrano D, Redin E, Exposito F, Leon S, Valencia K, Montuenga L, and Calvo A
- Abstract
Despite the great clinical success of immunotherapy in lung cancer patients, only a small percentage of them (<40%) will benefit from this therapy alone or combined with other strategies. Cancer cell-intrinsic and cell-extrinsic mechanisms have been associated with a lack of response to immunotherapy. The present study is focused on cancer cell-intrinsic genetic, epigenetic, transcriptomic and metabolic alterations that reshape the tumor microenvironment (TME) and determine response or refractoriness to immune checkpoint inhibitors (ICIs). Mutations in KRAS , SKT11 ( LKB1 ), KEAP1 and TP53 and co-mutations of these genes are the main determinants of ICI response in non-small-cell lung cancer (NSCLC) patients. Recent insights into metabolic changes in cancer cells that impose restrictions on cytotoxic T cells and the efficacy of ICIs indicate that targeting such metabolic restrictions may favor therapeutic responses. Other emerging pathways for therapeutic interventions include epigenetic modulators and DNA damage repair (DDR) pathways, especially in small-cell lung cancer (SCLC). Therefore, the many potential pathways for enhancing the effect of ICIs suggest that, in a few years, we will have much more personalized medicine for lung cancer patients treated with immunotherapy. Such strategies could include vaccines and chimeric antigen receptor (CAR) cells.
- Published
- 2023
- Full Text
- View/download PDF
17. YES1 Is a Druggable Oncogenic Target in SCLC.
- Author
-
Redin E, Garrido-Martin EM, Valencia K, Redrado M, Solorzano JL, Carias R, Echepare M, Exposito F, Serrano D, Ferrer I, Nunez-Buiza A, Garmendia I, García-Pedrero JM, Gurpide A, Paz-Ares L, Politi K, Montuenga LM, and Calvo A
- Subjects
- Humans, Mice, Animals, Cell Line, Tumor, Oncogenes, Cell Proliferation genetics, Apoptosis, Carcinogenesis genetics, Proto-Oncogene Proteins c-yes genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Small Cell Lung Carcinoma drug therapy, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology
- Abstract
Introduction: SCLC is an extremely aggressive subtype of lung cancer without approved targeted therapies. Here we identified YES1 as a novel targetable oncogene driving SCLC maintenance and metastasis., Methods: Association between YES1 levels and prognosis was evaluated in SCLC clinical samples. In vitro functional experiments for proliferation, apoptosis, cell cycle, and cytotoxicity were performed. Genetic and pharmacologic inhibition of YES1 was evaluated in vivo in cell- and patient-derived xenografts and metastasis. YES1 levels were evaluated in mouse and patient plasma-derived exosomes., Results: Overexpression or gain/amplification of YES1 was identified in 31% and 26% of cases, respectively, across molecular subgroups, and was found as an independent predictor of poor prognosis. Genetic depletion of YES1 dramatically reduced cell proliferation, three-dimensional organoid formation, tumor growth, and distant metastasis, leading to extensive apoptosis and tumor regressions. Mechanistically, YES1-inhibited cells revealed alterations in the replisome and DNA repair processes, that conferred sensitivity to irradiation. Pharmacologic blockade with the novel YES1 inhibitor CH6953755 or dasatinib induced marked antitumor activity in organoid models and cell- and patient-derived xenografts. YES1 protein was detected in plasma exosomes from patients and mouse models, with levels matching those of tumors, suggesting that circulating YES1 could represent a biomarker for patient selection/monitoring., Conclusions: Our results provide evidence that YES1 is a new druggable oncogenic target and biomarker to advance the clinical management of a subpopulation of patients with SCLC., (Copyright © 2022 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. YES1: A Novel Therapeutic Target and Biomarker in Cancer.
- Author
-
Garmendia I, Redin E, Montuenga LM, and Calvo A
- Subjects
- Biomarkers, Cell Line, Tumor, Cell Proliferation, Dasatinib pharmacology, Humans, Proto-Oncogene Proteins c-yes metabolism, Neoplasms drug therapy, Neoplasms genetics, src-Family Kinases
- Abstract
YES1 is a nonreceptor tyrosine kinase that belongs to the SRC family of kinases (SFK) and controls multiple cancer signaling pathways. YES1 is amplified and overexpressed in many tumor types, where it promotes cell proliferation, survival, and invasiveness. Therefore, YES1 has been proposed as an emerging target in solid tumors. In addition, studies have shown that YES1 is a prognostic biomarker and a predictor of dasatinib activity. Several SFKs-targeting drugs have been developed, and some of them have reached clinical trials. However, these drugs have encountered challenges to their utilization in the clinical practice in unselected patients due to toxicity and lack of efficacy. In the case of YES1, novel specific inhibitors have been developed and tested in preclinical models, with impressive antitumor effects. In this review, we summarize the structure and activation of YES1 and describe its role in cancer as a target and prognostic and companion biomarker. We also address the efficacy of SFKs inhibitors that are currently in clinical trials, highlighting the main hindrances for their clinical use. Current available information strongly suggests that inhibiting YES1 in tumors with high expression of this protein is a promising strategy against cancer., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
19. Innate and Adaptive Responses of Intratumoral Immunotherapy with Endosomal Toll-Like Receptor Agonists.
- Author
-
Andón FT, Leon S, Ummarino A, Redin E, Allavena P, Serrano D, Anfray C, and Calvo A
- Abstract
Toll-like receptors (TLRs) are natural initial triggers of innate and adaptive immune responses. With the advent of cancer immunotherapy, nucleic acids engineered as ligands of endosomal TLRs have been investigated for the treatment of solid tumors. Despite promising results, their systemic administration, similarly to other immunotherapies, raises safety issues. To overcome these problems, recent studies have applied the direct injection of endosomal TLR agonists in the tumor and/or draining lymph nodes, achieving high local drug exposure and strong antitumor response. Importantly, intratumoral delivery of TLR agonists showed powerful effects not only against the injected tumors but also often against uninjected lesions (abscopal effects), resulting in some cases in cure and antitumoral immunological memory. Herein, we describe the structure and function of TLRs and their role in the tumor microenvironment. Then, we provide our vision on the potential of intratumor versus systemic delivery or vaccination approaches using TLR agonists, also considering the use of nanoparticles to improve their targeting properties. Finally, we collect the preclinical and clinical studies applying intratumoral injection of TLR agonists as monotherapies or in combination with: (a) other TLR or STING agonists; (b) other immunotherapies; (c) radiotherapy or chemotherapy; (d) targeted therapies.
- Published
- 2022
- Full Text
- View/download PDF
20. Circulating Levels of the Interferon-γ-Regulated Chemokines CXCL10/CXCL11, IL-6 and HGF Predict Outcome in Metastatic Renal Cell Carcinoma Patients Treated with Antiangiogenic Therapy.
- Author
-
Esteban E, Exposito F, Crespo G, Lambea J, Pinto A, Puente J, Arranz JA, Redrado M, Rodriguez-Antona C, de Andrea C, Lopez-Brea M, Redin E, Rodriguez A, Serrano D, Garcia J, Grande E, Castellano D, and Calvo A
- Abstract
Sunitinib and pazopanib are standard first-line treatments for patients with metastatic renal cell carcinoma (mRCC). Nonetheless, as the number of treatment options increases, there is a need to identify biomarkers that can predict drug efficacy and toxicity. In this prospective study we evaluated a set of biomarkers that had been previously identified within a secretory signature in mRCC patients. This set includes tumor expression of c-Met and serum levels of HGF, IL-6, IL-8, CXCL9, CXCL10 and CXCL11. Our cohort included 60 patients with mRCC from 10 different Spanish hospitals who received sunitinib ( n = 51), pazopanib ( n = 4) or both ( n = 5). Levels of biomarkers were studied in relation to response rate, progression-free survival (PFS) and overall survival (OS). High tumor expression of c-Met and high basal serum levels of HGF, IL-6, CXCL11 and CXCL10 were significantly associated with reduced PFS and/or OS. In multivariable Cox regression analysis, CXCL11 was identified as an independent biomarker predictive of shorter PFS and OS, and HGF was an independent predictor of reduced PFS. Correlation analyses using our cohort of patients and patients from TCGA showed that HGF levels were significantly correlated with those of IL-6, CXCL11 and CXCL10. Bioinformatic protein-protein network analysis revealed a significant interaction between these proteins, all this suggesting a coordinated expression and secretion. We also developed a prognostic index that considers this group of biomarkers, where high values in mRCC patients can predict higher risk of relapse (HR 5.28 [2.32-12.0], p < 0.0001). In conclusion, high plasma HGF, CXCL11, CXCL10 and IL-6 levels are associated with worse outcome in mRCC patients treated with sunitinib or pazopanib. Our findings also suggest that these factors may constitute a secretory cluster that acts coordinately to promote tumor growth and resistance to antiangiogenic therapy.
- Published
- 2021
- Full Text
- View/download PDF
21. Immune Cell Infiltrates and Neutrophil-to-Lymphocyte Ratio in Relation to Response to Chemotherapy and Prognosis in Laryngeal and Hypopharyngeal Squamous Cell Carcinomas.
- Author
-
Sánchez-Canteli M, Juesas L, Redin E, Calvo A, López F, Astudillo A, Montuenga LM, García-Pedrero JM, and Rodrigo JP
- Abstract
Our goal was to assess the correlation of immune parameters with the response to induction chemotherapy (ICT) in head and neck squamous cell carcinoma (HNSCC) patients. Pretreatment biopsies from 64 patients with HNSCC that received ICT were assessed for PD-L1 protein expression and density of CD8+ and FOXP3+ tumor infiltrating lymphocytes (TIL). In addition, the neutrophil-to-lymphocyte ratio (NLR) was calculated from pretreatment whole blood counts. In total, 55% of cases exhibited PD-L1 combined proportion score (CPS) positivity (≥1% stained cells). PD-L1 CPS positivity correlated with a high density of both CD8+ ( p = 0.01) and FOXP3+ ( p < 0.001) TILs. There was no correlation between PD-L1 expression or TIL density and NLR values. In univariate analyses, the absence of PD-L1 CPS expression ( p = 0.042) and a high NLR ( p = 0.034) were significantly correlated with response to ICT. Neither CD8+ TIL ( p = 0.99) nor FOXP3+ TIL densities ( p = 0.71) were associated with response to ICT. In multivariate analysis, only a high NLR was associated with response to ICT (HR = 4.06, 95% CI = 1.06-15.5, p = 0.04). In addition, a high NLR was also independently associated with lower disease-specific ( p = 0.03) and overall survival rates ( p = 0.04), particularly in the subset of patients who received definitive surgical treatment. These results suggest that NLR could emerge as a predictive biomarker of response to ICT.
- Published
- 2021
- Full Text
- View/download PDF
22. SRC family kinase (SFK) inhibitor dasatinib improves the antitumor activity of anti-PD-1 in NSCLC models by inhibiting Treg cell conversion and proliferation.
- Author
-
Redin E, Garmendia I, Lozano T, Serrano D, Senent Y, Redrado M, Villalba M, De Andrea CE, Exposito F, Ajona D, Ortiz-Espinosa S, Remirez A, Bertolo C, Sainz C, Garcia-Pedrero J, Pio R, Lasarte J, Agorreta J, Montuenga LM, and Calvo A
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung enzymology, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Drug Resistance, Neoplasm, Female, Humans, Lung Neoplasms enzymology, Lung Neoplasms immunology, Lung Neoplasms pathology, Lymphocytes, Tumor-Infiltrating enzymology, Lymphocytes, Tumor-Infiltrating immunology, Mice, Mice, 129 Strain, Phenotype, Programmed Cell Death 1 Receptor metabolism, Proto-Oncogene Proteins c-yes metabolism, Signal Transduction, T-Lymphocytes, Regulatory enzymology, T-Lymphocytes, Regulatory immunology, Tumor Microenvironment, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, Cell Proliferation drug effects, Dasatinib pharmacology, Immune Checkpoint Inhibitors pharmacology, Lung Neoplasms drug therapy, Lymphocytes, Tumor-Infiltrating drug effects, Programmed Cell Death 1 Receptor antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-yes antagonists & inhibitors, T-Lymphocytes, Regulatory drug effects
- Abstract
Introduction: The use of immune-checkpoint inhibitors has drastically improved the management of patients with non-small cell lung cancer (NSCLC), but innate and acquired resistances are hurdles needed to be solved. Immunomodulatory drugs that can reinvigorate the immune cytotoxic activity, in combination with antiprogrammed cell death 1 (PD-1) antibody, are a great promise to overcome resistance. We evaluated the impact of the SRC family kinases (SFKs) on NSCLC prognosis, and the immunomodulatory effect of the SFK inhibitor dasatinib, in combination with anti-PD-1, in clinically relevant mouse models of NSCLC., Methods: A cohort of patients from University Clinic of Navarra (n=116) was used to study immune infiltrates by multiplex immunofluorescence (mIF) and YES1 protein expression in tumor samples. Publicly available resources (TCGA, Km Plotter, and CIBERSORT) were used to study patient's survival based on expression of SFKs and tumor infiltrates. Syngeneic NSCLC mouse models 393P and UNSCC680AJ were used for in vivo drug testing., Results: Among the SFK members, YES1 expression showed the highest association with poor prognosis. Patients with high YES1 tumor levels also showed high infiltration of CD4+/FOXP3+ cells (regulatory T cells (Tregs)), suggesting an immunosuppressive phenotype. After testing for YES1 expression in a panel of murine cell lines, 393P and UNSCC680AJ were selected for in vivo studies. In the 393P model, dasatinib+anti-PD-1 treatment resulted in synergistic activity, with 87% tumor regressions and development of immunological memory that impeded tumor growth when mice were rechallenged. In vivo depletion experiments further showed that CD8+ and CD4+ cells are necessary for the therapeutic effect of the combination. The antitumor activity was accompanied by a very significant decrease in the number of Tregs, which was validated by mIF in tumor sections. In the UNSCC680AJ model, the antitumor effects of dasatinib+anti-PD-1 were milder but similar to the 393P model. In in vitro assays, we demonstrated that dasatinib blocks proliferation and transforming growth factor beta-driven conversion of effector CD4+ cells into Tregs through targeting of phospholymphocyte-specific protein tyrosine kinase and downstream effectors pSTAT5 and pSMAD3., Conclusions: YES1 protein expression is associated with increased numbers of Tregs in patients with NSCLC. Dasatinib synergizes with anti-PD-1 to impair tumor growth in NSCLC experimental models. This study provides the preclinical rationale for the combined use of dasatinib and PD-1/programmed death-ligand 1 blockade to improve outcomes of patients with NSCLC., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
23. Identification of a novel synthetic lethal vulnerability in non-small cell lung cancer by co-targeting TMPRSS4 and DDR1.
- Author
-
Villalba M, Redin E, Exposito F, Pajares MJ, Sainz C, Hervas D, Guruceaga E, Diaz-Lagares A, Cirauqui C, Redrado M, Valencia K, de Andrea C, Jantus-Lewintre E, Camps C, Lopez-Lopez R, Lahoz A, Montuenga L, Pio R, Sandoval J, and Calvo A
- Subjects
- Adenocarcinoma pathology, Animals, Apoptosis drug effects, Cell Line, Tumor, Cisplatin toxicity, DNA Methylation, Dasatinib toxicity, Drug Resistance, Neoplasm, Humans, Lung Neoplasms pathology, Mice, Promoter Regions, Genetic, Adenocarcinoma genetics, Biomarkers, Tumor genetics, Discoidin Domain Receptor 1 genetics, Lung Neoplasms genetics, Membrane Proteins genetics, Serine Endopeptidases genetics, Synthetic Lethal Mutations
- Abstract
Finding novel targets in non-small cell lung cancer (NSCLC) is highly needed and identification of synthetic lethality between two genes is a new approach to target NSCLC. We previously found that TMPRSS4 promotes NSCLC growth and constitutes a prognostic biomarker. Here, through large-scale analyses across 5 public databases we identified consistent co-expression between TMPRSS4 and DDR1. Similar to TMPRSS4, DDR1 promoter was hypomethylated in NSCLC in 3 independent cohorts and hypomethylation was an independent prognostic factor of disease-free survival. Treatment with 5-azacitidine increased DDR1 levels in cell lines, suggesting an epigenetic regulation. Cells lacking TMPRSS4 were highly sensitive to the cytotoxic effect of the DDR1 inhibitor dasatinib. TMPRSS4/DDR1 double knock-down (KD) cells, but not single KD cells suffered a G0/G1 cell cycle arrest with loss of E2F1 and cyclins A and B, increased p21 levels and a larger number of cells in apoptosis. Moreover, double KD cells were highly sensitized to cisplatin, which caused massive apoptosis (~40%). In vivo studies demonstrated tumor regression in double KD-injected mice. In conclusion, we have identified a novel vulnerability in NSCLC resulting from a synthetic lethal interaction between DDR1 and TMPRSS4.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.