311 results on '"J. Albanell"'
Search Results
102. Publisher Correction: MSK1 regulates luminal cell differentiation and metastatic dormancy in ER + breast cancer.
- Author
-
Gawrzak S, Rinaldi L, Gregorio S, Arenas EJ, Salvador F, Urosevic J, Figueras-Puig C, Rojo F, Del Barco Barrantes I, Cejalvo JM, Palafox M, Guiu M, Berenguer-Llergo A, Symeonidi A, Bellmunt A, Kalafatovic D, Arnal-Estapé A, Fernández E, Müllauer B, Groeneveld R, Slobodnyuk K, Stephan-Otto Attolini C, Saura C, Arribas J, Cortes J, Rovira A, Muñoz M, Lluch A, Serra V, Albanell J, Prat A, Nebreda AR, Benitah SA, and Gomis RR
- Abstract
In the version of this Article originally published, the boxes framing the two plots in Fig. 1g were misaligned from the axes due to a technical error. This has now been corrected in all versions of the Article.
- Published
- 2018
- Full Text
- View/download PDF
103. Manic Fringe deficiency imposes Jagged1 addiction to intestinal tumor cells.
- Author
-
López-Arribillaga E, Rodilla V, Colomer C, Vert A, Shelton A, Cheng JH, Yan B, Gonzalez-Perez A, Junttila MR, Iglesias M, Torres F, Albanell J, Villanueva A, Bigas A, Siebel CW, and Espinosa L
- Subjects
- Animals, Biomarkers, Tumor metabolism, Carcinogenesis metabolism, Carcinogenesis pathology, Cell Proliferation, Epithelial Cells metabolism, Epithelial Cells pathology, Glucosyltransferases, Humans, Ligands, Mice, Models, Biological, Prognosis, Receptor, Notch1 metabolism, Signal Transduction, Spheroids, Cellular metabolism, Spheroids, Cellular pathology, Stem Cells metabolism, Transcription, Genetic, Hexosyltransferases metabolism, Intestinal Neoplasms metabolism, Intestinal Neoplasms pathology, Intracellular Signaling Peptides and Proteins metabolism, Jagged-1 Protein metabolism, Membrane Proteins metabolism, Proteins metabolism
- Abstract
Delta ligands regulate Notch signaling in normal intestinal stem cells, while Jagged1 activates Notch in intestinal adenomas carrying active β-catenin. We used the Apc
Min/+ mouse model, tumor spheroid cultures, and patient-derived orthoxenografts to address this divergent ligand-dependent Notch function and its implication in disease. We found that intestinal-specific Jag1 deletion or antibody targeting Jag1 prevents tumor initiation in mice. Addiction to Jag1 is concomitant with the absence of Manic Fringe (MFNG) in adenoma cells, and its ectopic expression reverts Jag1 dependence. In 239 human colorectal cancer patient samples, MFNG imposes a negative correlation between Jag1 and Notch, being high Jag1 in the absence of MFNG predictive of poor prognosis. Jag1 antibody treatment reduces patient-derived tumor orthoxenograft growth without affecting normal intestinal mucosa. Our data provide an explanation to Jag1 dependence in cancer, and reveal that Jag1-Notch1 interference provides therapeutic benefit in a subset of colorectal cancer and FAP syndrome patients.- Published
- 2018
- Full Text
- View/download PDF
104. Biomarkers in breast cancer: A consensus statement by the Spanish Society of Medical Oncology and the Spanish Society of Pathology.
- Author
-
Colomer R, Aranda-López I, Albanell J, García-Caballero T, Ciruelos E, López-García MÁ, Cortés J, Rojo F, Martín M, and Palacios-Calvo J
- Subjects
- Breast Neoplasms genetics, Female, Gene Expression Profiling, Humans, Societies, Medical, Spain, Biomarkers, Tumor genetics, Breast Neoplasms diagnosis, Decision Making, Practice Guidelines as Topic standards
- Abstract
This consensus statement revises and updates the recommendations for biomarkers use in the diagnosis and treatment of breast cancer, and is a joint initiative of the Spanish Society of Medical Oncology and the Spanish Society of Pathology. This expert group recommends determining in all cases of breast cancer the histologic grade and the alpha-estrogen receptor (ER), progesterone receptor, Ki-67 and HER2 status, in order to assist prognosis and establish therapeutic options, including hormone therapy, chemotherapy and anti-HER2 therapy. One of the four available genetic prognostic platforms (MammaPrint
® , Oncotype DX® , Prosigna® or EndoPredict® ) may be used in node-negative ER-positive patients to establish a prognostic category and decide with the patient whether adjuvant treatment may be limited to hormonal therapy. Newer technologies including next-generation sequencing, liquid biopsy, tumour-infiltrating lymphocytes or PD-1 determination are at this point investigational.- Published
- 2018
- Full Text
- View/download PDF
105. Triple negative breast cancer subtypes and pathologic complete response rate to neoadjuvant chemotherapy.
- Author
-
Santonja A, Sánchez-Muñoz A, Lluch A, Chica-Parrado MR, Albanell J, Chacón JI, Antolín S, Jerez JM, de la Haba J, de Luque V, Fernández-De Sousa CE, Vicioso L, Plata Y, Ramírez-Tortosa CL, Álvarez M, Llácer C, Zarcos-Pedrinaci I, Carrasco E, Caballero R, Martín M, and Alba E
- Abstract
Triple negative breast cancer (TNBC) is a heterogeneous disease with distinct molecular subtypes that differentially respond to chemotherapy and targeted agents. The purpose of this study is to explore the clinical relevance of Lehmann TNBC subtypes by identifying any differences in response to neoadjuvant chemotherapy among them. We determined Lehmann subtypes by gene expression profiling in paraffined pre-treatment tumor biopsies from 125 TNBC patients treated with neoadjuvant anthracyclines and/or taxanes +/- carboplatin. We explored the clinicopathological characteristics of Lehmann subtypes and their association with the pathologic complete response (pCR) to different treatments. The global pCR rate was 37%, and it was unevenly distributed within Lehmann's subtypes. Basal-like 1 (BL1) tumors exhibited the highest pCR to carboplatin containing regimens (80% vs 23%, p=0.027) and were the most proliferative (Ki-67>50% of 88.2% vs. 63.7%, p=0.02). Luminal-androgen receptor (LAR) patients achieved the lowest pCR to all treatments (14.3% vs 42.7%, p=0.045 when excluding mesenchymal stem-like (MSL) samples) and were the group with the lowest proliferation (Ki-67≤50% of 71% vs 27%, p=0.002). In our cohort, only tumors with LAR phenotype presented non-basal-like intrinsic subtypes (HER2-enriched and luminal A). TNBC patients present tumors with a high genetic diversity ranging from highly proliferative tumors, likely responsive to platinum-based therapies, to a subset of chemoresistant tumors with low proliferation and luminal characteristics., Competing Interests: CONFLICTS OF INTEREST The authors declare that they have no conflicts of interest.
- Published
- 2018
- Full Text
- View/download PDF
106. Efficacy of Sym004 in Patients With Metastatic Colorectal Cancer With Acquired Resistance to Anti-EGFR Therapy and Molecularly Selected by Circulating Tumor DNA Analyses: A Phase 2 Randomized Clinical Trial.
- Author
-
Montagut C, Argilés G, Ciardiello F, Poulsen TT, Dienstmann R, Kragh M, Kopetz S, Lindsted T, Ding C, Vidal J, Clausell-Tormos J, Siravegna G, Sánchez-Martín FJ, Koefoed K, Pedersen MW, Grandal MM, Dvorkin M, Wyrwicz L, Rovira A, Cubillo A, Salazar R, Desseigne F, Nadal C, Albanell J, Zagonel V, Siena S, Fumi G, Rospo G, Nadler P, Horak ID, Bardelli A, and Tabernero J
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor analysis, Biomarkers, Tumor blood, Biomarkers, Tumor genetics, Circulating Tumor DNA analysis, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology, ErbB Receptors antagonists & inhibitors, Female, Humans, Male, Middle Aged, Neoplasm Metastasis, Survival Analysis, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Circulating Tumor DNA blood, Colorectal Neoplasms blood, Colorectal Neoplasms drug therapy, Drug Resistance, Neoplasm drug effects, Patient Selection, Protein Kinase Inhibitors therapeutic use
- Abstract
Importance: Acquired resistance to anti-EGFR therapy (epidermal growth factor receptor) is frequently due to RAS and EGFR extracellular domain (ECD) mutations in metastatic colorectal cancer (mCRC). Some anti-EGFR-refractory patients retain tumor EGFR dependency potentially targetable by agents such as Sym004, which is a mixture of 2 nonoverlapping monoclonal antibodies targeting EGFR., Objective: To determine if continuous blockade of EGFR by Sym004 has survival benefit., Design, Setting, and Participants: Multicenter, phase 2, randomized, clinical trial comparing 2 regimens of Sym004 with investigator's choice from March 6, 2014, through October 15, 2015. Circulating tumor DNA (ctDNA) was analyzed for biomarker and tracking clonal dynamics during treatment. Participants had wild-type KRAS exon 2 mCRC refractory to standard chemotherapy and acquired resistance to anti-EGFR monoclonal antibodies., Interventions: Participants were randomly assigned in a 1:1:1 ratio to Sym004, 12 mg/kg/wk (arm A), Sym004, 9 mg/kg loading dose followed by 6 mg/kg/wk (arm B), or investigator's choice of treatment (arm C)., Main Outcomes and Measures: Overall survival (OS). Secondary end points included preplanned exploratory biomarker analysis in ctDNA., Results: A total of 254 patients were randomized (intent-to-treat [ITT] population) (median age, 63 [range, 34-91] years; 63% male; n = 160). Median OS in the ITT population was 7.9 months (95% CI, 6.5-9.9 months), 10.3 months (95% CI, 9.0-12.9 months), and 9.6 months (95% CI, 8.3-12.2 months) for arms A, B, and C, respectively (hazard ratio [HR], 1.31; 95% CI, 0.92-1.87 for A vs C; and HR, 0.97; 95% CI, 0.68-1.40 for B vs C). The ctDNA revealed high intrapatient genomic heterogeneity following anti-EGFR therapy. Sym004 effectively targeted EGFR ECD-mutated cancer cells, and a decrease in EGFR ECD ctDNA occurred in Sym004-treated patients. However, this did not translate into clinical benefit in patients with EGFR ECD mutations, likely owing to co-occurring resistance mechanisms. A subgroup of patients was defined by ctDNA (RAS/BRAF/EGFR ECD-mutation negative) associated with improved OS in Sym004-treated patients in arm B compared with arm C (median OS, 12.8 and 7.3 months, respectively)., Conclusions and Relevance: Sym004 did not improve OS in an unselected population of patients with mCRC and acquired anti-EGFR resistance. A prospective clinical validation of Sym004 efficacy in a ctDNA molecularly defined subgroup of patients with refractory mCRC is warranted., Trial Registration: clinicaltrialsregister.eu Identifier: 2013-003829-29.
- Published
- 2018
- Full Text
- View/download PDF
107. [Consensus statement on biomarkers in breast cancer by the Spanish Society of Pathology and the Spanish Society of Medical Oncology].
- Author
-
Palacios Calvo J, Albanell J, Rojo F, Ciruelos E, Aranda-López I, Cortés J, García-Caballero T, Martín M, López-García MÁ, and Colomer R
- Subjects
- Algorithms, Biomarkers, Tumor blood, Breast Neoplasms blood, Breast Neoplasms chemistry, Female, Humans, Biomarkers, Tumor analysis, Breast Neoplasms diagnosis
- Abstract
This consensus statement is a joint initiative of the Spanish Society of Pathology (SEAP) and the Spanish Society of Medical Oncology (SEOM). It revises and updates the recommendations for the use of biomarkers in the diagnosis and treatment of breast cancer. The group of experts recommends that, in all cases of breast cancer, the histologic grade and the alpha-estrogen receptor (ER), progesterone receptor, Ki-67 and HER2 status should be determined, in order to assist prognosis and establish therapeutic options, including hormone therapy, chemotherapy and anti-HER2 therapy. One of the four available genetic prognostic platforms (MammaPrint
® , Oncotype DX® , Prosigna® or EndoPredict® ) may be used in node-negative ER-positive patients to establish a prognostic category and decide, together with the patient, whether adjuvant treatment be limited to hormonal therapy. Newer technologies, including next generation sequencing, liquid biopsy, tumour infiltrating lymphocytes or PD-1 determination, are still investigational., (Copyright © 2018 Sociedad Española de Anatomía Patológica. Publicado por Elsevier España, S.L.U. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
108. First-in-Human Phase I Study of Single-agent Vanucizumab, A First-in-Class Bispecific Anti-Angiopoietin-2/Anti-VEGF-A Antibody, in Adult Patients with Advanced Solid Tumors.
- Author
-
Hidalgo M, Martinez-Garcia M, Le Tourneau C, Massard C, Garralda E, Boni V, Taus A, Albanell J, Sablin MP, Alt M, Bahleda R, Varga A, Boetsch C, Franjkovic I, Heil F, Lahr A, Lechner K, Morel A, Nayak T, Rossomanno S, Smart K, Stubenrauch K, and Krieter O
- Subjects
- Adult, Aged, Angiogenesis Inhibitors pharmacokinetics, Angiogenesis Inhibitors pharmacology, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal, Humanized, Colonic Neoplasms metabolism, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Humans, Kidney Neoplasms metabolism, Male, Maximum Tolerated Dose, Middle Aged, Neovascularization, Pathologic drug therapy, Antibodies, Monoclonal pharmacology, Colonic Neoplasms drug therapy, Kidney Neoplasms drug therapy, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vesicular Transport Proteins antagonists & inhibitors
- Abstract
Purpose: Vanucizumab is an investigational antiangiogenic, first-in-class, bispecific mAb targeting VEGF-A and angiopoietin-2 (Ang-2). This first-in-human study evaluated the safety, pharmacokinetics, pharmacodynamics, and antitumor activity of vanucizumab in adults with advanced solid tumors refractory to standard therapies. Experimental Design: Patients received escalating biweekly (3-30 mg/kg) or weekly (10-30 mg/kg) intravenous doses guided by a Bayesian logistic regression model with overdose control. Results: Forty-two patients were treated. One dose-limiting toxicity, a fatal pulmonary hemorrhage from a large centrally located mediastinal mass judged possibly related to vanucizumab, occurred with the 19 mg/kg biweekly dose. Arterial hypertension (59.5%), asthenia (42.9%), and headache (31%) were the most common toxicities. Seventeen (41%) patients experienced treatment-related grade ≥3 toxicities. Toxicity was generally higher with weekly than biweekly dosing. A MTD of vanucizumab was not reached in either schedule. Pharmacokinetics were dose-linear with an elimination half-life of 6-9 days. All patients had reduced plasma levels of free VEGF-A and Ang-2; most had reductions in K
TRANS (measured by dynamic contrast-enhanced MRI). Two patients (renal cell and colon cancer) treated with 30 mg/kg achieved confirmed partial responses. Ten patients were without disease progression for ≥6 months. A flat-fixed 2,000 mg biweekly dose (phamacokinetically equivalent to 30 mg/kg biweekly) was recommended for further investigation. Conclusions: Biweekly vanucizumab had an acceptable safety and tolerability profile consistent with single-agent use of selective inhibitors of the VEGF-A and Ang/Tie2 pathway. Vanucizumab modulated its angiogenic targets, impacted tumor vascularity, and demonstrated encouraging antitumor activity in this heterogeneous population. Clin Cancer Res; 24(7); 1536-45. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
109. Cancer Genome Interpreter annotates the biological and clinical relevance of tumor alterations.
- Author
-
Tamborero D, Rubio-Perez C, Deu-Pons J, Schroeder MP, Vivancos A, Rovira A, Tusquets I, Albanell J, Rodon J, Tabernero J, de Torres C, Dienstmann R, Gonzalez-Perez A, and Lopez-Bigas N
- Subjects
- Antineoplastic Agents therapeutic use, Biomarkers, Tumor metabolism, Databases, Genetic, Genes, Neoplasm, Humans, Mutation genetics, Neoplasms drug therapy, Genome, Human, Molecular Sequence Annotation, Neoplasms genetics, Software
- Abstract
While tumor genome sequencing has become widely available in clinical and research settings, the interpretation of tumor somatic variants remains an important bottleneck. Here we present the Cancer Genome Interpreter, a versatile platform that automates the interpretation of newly sequenced cancer genomes, annotating the potential of alterations detected in tumors to act as drivers and their possible effect on treatment response. The results are organized in different levels of evidence according to current knowledge, which we envision can support a broad range of oncology use cases. The resource is publicly available at http://www.cancergenomeinterpreter.org .
- Published
- 2018
- Full Text
- View/download PDF
110. MSK1 regulates luminal cell differentiation and metastatic dormancy in ER + breast cancer.
- Author
-
Gawrzak S, Rinaldi L, Gregorio S, Arenas EJ, Salvador F, Urosevic J, Figueras-Puig C, Rojo F, Del Barco Barrantes I, Cejalvo JM, Palafox M, Guiu M, Berenguer-Llergo A, Symeonidi A, Bellmunt A, Kalafatovic D, Arnal-Estapé A, Fernández E, Müllauer B, Groeneveld R, Slobodnyuk K, Stephan-Otto Attolini C, Saura C, Arribas J, Cortes J, Rovira A, Muñoz M, Lluch A, Serra V, Albanell J, Prat A, Nebreda AR, Benitah SA, and Gomis RR
- Subjects
- Adult, Aged, Animals, Biomarkers, Tumor genetics, Bone Neoplasms genetics, Bone Neoplasms pathology, Bone Neoplasms secondary, Breast Neoplasms pathology, Cell Differentiation genetics, Chromatin genetics, Female, Gene Expression Regulation, Neoplastic, Genome, Human genetics, Humans, Mice, Middle Aged, Neoplasm Metastasis, Prognosis, RNA, Small Interfering genetics, Receptors, Estrogen genetics, Xenograft Model Antitumor Assays, Breast Neoplasms genetics, GATA3 Transcription Factor genetics, Hepatocyte Nuclear Factor 3-alpha genetics, Ribosomal Protein S6 Kinases, 90-kDa genetics
- Abstract
For many patients with breast cancer, symptomatic bone metastases appear after years of latency. How micrometastatic lesions remain dormant and undetectable before initiating colonization is unclear. Here, we describe a mechanism involved in bone metastatic latency of oestrogen receptor-positive (ER
+ ) breast cancer. Using an in vivo genome-wide short hairpin RNA screening, we identified the kinase MSK1 as an important regulator of metastatic dormancy in breast cancer. In patients with ER+ breast cancer, low MSK1 expression associates with early metastasis. We show that MSK1 downregulation impairs the differentiation of breast cancer cells, increasing their bone homing and growth capacities. MSK1 controls the expression of genes required for luminal cell differentiation, including the GATA3 and FOXA1 transcription factors, by modulating their promoter chromatin status. Our results indicate that MSK1 prevents metastatic progression of ER+ breast cancer, suggesting that stratifying patients with breast cancer as high or low risk for early relapse based on MSK1 expression could improve prognosis.- Published
- 2018
- Full Text
- View/download PDF
111. Recent Insights into the Development of Preclinical Trastuzumab- Resistant HER2+ Breast Cancer Models.
- Author
-
Gonzalez-Alonso P, Cristobal I, Zazo S, Martin-Aparicio E, Chamizo C, Madoz-Gurpide J, Rovira A, Eroles P, Lluch A, Albanell J, and Rojo F
- Subjects
- Animals, Antineoplastic Agents, Immunological immunology, Biomarkers, Tumor, Cell Line, Tumor, Drug Resistance, Neoplasm, Female, Gene Expression Regulation, Neoplastic, Humans, Receptor, ErbB-2 genetics, Receptor, ErbB-2 immunology, Trastuzumab immunology, Antineoplastic Agents, Immunological therapeutic use, Breast Neoplasms drug therapy, Receptor, ErbB-2 antagonists & inhibitors, Trastuzumab therapeutic use
- Abstract
Background: Overexpression and amplification of the human epidermal growth factor receptor 2 (HER2) occur in 20% of total breast carcinomas. HER2-overexpression is implicated in disease initiation and progression and associated with poor prognosis. Trastuzumab, a humanized monoclonal antibody, is the standard HER2-targeted therapy for early and metastatic HER2-amplified breast cancer patients. Trastuzumab has significantly increased clinical benefit in HER2+ metastatic and adjuvant settings; however, it is not effective for many patients due to primary or acquired resistance to the drug. During the last decade, many studies have revealed a number of novel molecular traits of HER2+ breast cancer, allowing us to uncover the molecular mechanisms involved in trastuzumab resistance and develop strategies to overcome resistance to therapy., Objective: In this review, we comprehensively addressed the current achievements in preclinical studies; we discussed molecular mechanisms of acquired trastuzumab resistance in HER2+ breast cancer models and potential therapeutic approaches based on the molecular features for HER2+ breast cancer., Conclusion: Enhanced understanding of the molecular profiles in HER2+ breast cancer may lead to the identification of novel biomarkers for the development of diagnostic approaches and improvement of therapeutic targets for the prevention and treatment of trastuzumab resistant HER2+ breast cancer., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.)
- Published
- 2018
- Full Text
- View/download PDF
112. CD274 (PDL1) and JAK2 genomic amplifications in pulmonary squamous-cell and adenocarcinoma patients.
- Author
-
Clavé S, Pijuan L, Casadevall D, Taus Á, Gimeno J, Hernández-Llodrà S, Rodríguez-Rivera M, Lorenzo M, Menéndez S, Albanell J, Espinet B, Arriola E, and Salido M
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Gene Amplification, Humans, Janus Kinase 2 biosynthesis, Male, Middle Aged, Retrospective Studies, B7-H1 Antigen genetics, Carcinoma, Non-Small-Cell Lung genetics, Gene Expression Regulation, Neoplastic genetics, Janus Kinase 2 genetics, Lung Neoplasms genetics
- Abstract
Aims: CD274 (PDL1) and JAK2 (9p24.1) gene amplifications have been recently described in pulmonary carcinomas in association with programmed death-ligand 1 (PD-L1) expression. Furthermore, PTEN loss has been explored preclinically in relation to PD-L1 expression. Our aim was to determine whether these genomic alterations affect PD-L1 expression levels in non-small-cell lung cancer., Methods and Results: PD-L1 and PTEN expression determined by immunohistochemistry (IHC), and CD274, JAK2 and PTEN copy number alterations (CNAs) determined by fluorescence in-situ hybridisation, were studied in 171 pulmonary carcinoma specimens. PD-L1 expression was positive in 40 cases (23.3%), and CD274 amplification was present in 14 tumours (8.8%). Concordance between both events was found in 12 of 14 amplified cases (P = 0.0001). We found nine JAK2-amplified cases (5.7%), seven with PD-L1 expression (P = 0.0006). Moreover, six of the seven cases had JAK2 and CD274 coamplification (9p24.1 genomic amplification). Remarkably, the average PD-L1 IHC score was higher in these amplified cases (230 versus 80; P = 0.001). Non-statistical associations were observed between PD-L1 expression and PTEN loss and PTEN deletions., Conclusions: We describe a subset of patients (8.2%) who had 9p24.1 amplifications resulting in high expression of PD-L1. Our results provide evidence for genomic up-regulation of PD-L1 expression in non-small-cell lung cancer., (© 2017 John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
113. Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance in HER2-positive Breast Cancer.
- Author
-
Sabbaghi M, Gil-Gómez G, Guardia C, Servitja S, Arpí O, García-Alonso S, Menendez S, Arumi-Uria M, Serrano L, Salido M, Muntasell A, Martínez-García M, Zazo S, Chamizo C, González-Alonso P, Madoz-Gúrpide J, Eroles P, Arribas J, Tusquets I, Lluch A, Pandiella A, Rojo F, Rovira A, and Albanell J
- Subjects
- Ado-Trastuzumab Emtansine, Animals, Apoptosis drug effects, Apoptosis genetics, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Breast Neoplasms pathology, CDC2 Protein Kinase genetics, CDC2 Protein Kinase metabolism, Cell Line, Tumor, Cyclin B1 metabolism, Disease Models, Animal, Female, G2 Phase Cell Cycle Checkpoints drug effects, G2 Phase Cell Cycle Checkpoints genetics, Humans, Maytansine pharmacology, Mice, Protein Binding, Receptor, ErbB-2 metabolism, Xenograft Model Antitumor Assays, Breast Neoplasms genetics, Cyclin B1 deficiency, Drug Resistance, Neoplasm genetics, Maytansine analogs & derivatives, Receptor, ErbB-2 genetics, Trastuzumab pharmacology
- Abstract
Purpose: Trastuzumab-emtansine (T-DM1) is a standard treatment in advanced HER2-positive breast cancer. However, resistance inevitably occurs. We aimed to identify mechanisms of acquired T-DM1 resistance. Experimental Design: HER2-positive breast cancer cells (HCC1954, HCC1419, SKBR3, and BT474) were treated in a pulse-fashion with T-DM1 to induce a resistant phenotype. Cellular and molecular effects of T-DM1 in parental versus resistant cells were compared. CDK1 kinase activity and cyclin B1 expression were assayed under various conditions. Genetic modifications to up- or downregulate cyclin B1 were conducted. Effects of T-DM1 on cyclin B1 levels, proliferation, and apoptosis were assayed in human HER2 -positive breast cancer explants. Results: We obtained three cell lines with different levels of acquired T-DM1 resistance (HCC1954/TDR, HCC1419/TDR, and SKBR3/TDR cells). HER2 remained amplified in the resistant cells. Binding to HER2 and intracellular uptake of T-DM1 were maintained in resistant cells. T-DM1 induced cyclin B1 accumulation in sensitive but not resistant cells. Cyclin B1 knockdown by siRNA in parental cells induced T-DM1 resistance, while increased levels of cyclin B1 by silencing cdc20 partially sensitized resistant cells. In a series of 18 HER2-positive breast cancer fresh explants, T-DM1 effects on proliferation and apoptosis paralleled cyclin B1 accumulation. Conclusions: Defective cyclin B1 induction by T-DM1 mediates acquired resistance in HER2-positive breast cancer cells. These results support the testing of cyclin B1 induction upon T-DM1 treatment as a pharmacodynamic predictor in HER2-positive breast cancer. Clin Cancer Res; 23(22); 7006-19. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
114. Interplay between Natural Killer Cells and Anti-HER2 Antibodies: Perspectives for Breast Cancer Immunotherapy.
- Author
-
Muntasell A, Cabo M, Servitja S, Tusquets I, Martínez-García M, Rovira A, Rojo F, Albanell J, and López-Botet M
- Abstract
Overexpression of the human epidermal growth factor receptor 2 (HER2) defines a subgroup of breast tumors with aggressive behavior. The addition of HER2-targeted antibodies (i.e., trastuzumab, pertuzumab) to chemotherapy significantly improves relapse-free and overall survival in patients with early-stage and advanced disease. Nonetheless, considerable proportions of patients develop resistance to treatment, highlighting the need for additional and co-adjuvant therapeutic strategies. HER2-specific antibodies can trigger natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity and indirectly enhance the development of tumor-specific T cell immunity; both mechanisms contributing to their antitumor efficacy in preclinical models. Antibody-dependent NK cell activation results in the release of cytotoxic granules as well as the secretion of pro-inflammatory cytokines (i.e., IFNγ and TNFα) and chemokines. Hence, NK cell tumor suppressive functions include direct cytolytic killing of tumor cells as well as the regulation of subsequent antitumor adaptive immunity. Albeit tumors with gene expression signatures associated to the presence of cytotoxic lymphocyte infiltrates benefit from trastuzumab-based treatment, NK cell-related biomarkers of response/resistance to HER2-specific therapeutic antibodies in breast cancer patients remain elusive. Several variables, including (i) the configuration of the patient NK cell repertoire; (ii) tumor molecular features (i.e., estrogen receptor expression); (iii) concomitant therapeutic regimens (i.e., chemotherapeutic agents, tyrosine kinase inhibitors); and (iv) evasion mechanisms developed by progressive breast tumors, have been shown to quantitatively and qualitatively influence antibody-triggered NK cell responses. In this review, we discuss possible interventions for restoring/enhancing the therapeutic activity of HER2 therapeutic antibodies by harnessing NK cell antitumor potential through combinatorial approaches, including immune checkpoint blocking/stimulatory antibodies, cytokines and toll-like receptor agonists.
- Published
- 2017
- Full Text
- View/download PDF
115. Heterogeneity of Tumor and Immune Cell PD-L1 Expression and Lymphocyte Counts in Surgical NSCLC Samples.
- Author
-
Casadevall D, Clavé S, Taus Á, Hardy-Werbin M, Rocha P, Lorenzo M, Menéndez S, Salido M, Albanell J, Pijuan L, and Arriola E
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma surgery, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung surgery, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell surgery, Female, Gene Amplification, Gene Expression Regulation, Neoplastic, Humans, In Situ Hybridization, Fluorescence, Lung Neoplasms genetics, Lung Neoplasms surgery, Lymphocyte Count, Male, Middle Aged, Patient Selection, Retrospective Studies, Tissue Array Analysis, Adenocarcinoma pathology, B7-H1 Antigen genetics, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell pathology, Lung Neoplasms pathology
- Abstract
Background: Immune-checkpoint inhibitors against programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) have shown remarkable therapeutic activity in non-small-cell lung cancer (NSCLC). However, biomarker-based patient selection remains a challenge. Our aim was to assess the heterogeneity of various immune markers between different tumor areas of surgically resected NSCLC specimens., Materials and Methods: We included 94 adenocarcinoma (ADC) and 50 squamous cell carcinoma (SCC) specimens. Two distinct tumor areas of each tumor sample were selected and incorporated into tissue microarrays. PD-L1 expression in tumor cells (TCs) and immune cells (ICs) was assessed using clone SP142 (Ventana). PDL1 gene amplification was assessed using fluorescence in situ hybridization. CD3 and CD8 densities were quantified using digital image-based analysis. Heterogeneity was assessed using kappa agreement index (KI) and intraclass correlation coefficient., Results: Prevalence of PD-L1 expression was 16.8% in TCs and 27.8% in ICs. Eleven tumors (7.6%) showed PDL1 amplification. In ADC, KI of PD-L1 TC and IC expression between cores was 0.465 and 0.260, compared with 0.274 and 0.124 in SCC, respectively. Higher concordance was observed for PDL1 amplification (KI, 0.647 in ADC and KI, 1 in SCC). Eleven (61.1%) of 18 amplified cores showed PD-L1 staining in < 5% of TCs. Intraclass correlation coefficients for CD3 and CD8 were 0.293 and 0.186 in ADC and 0.489 and 0.610 in SCC samples, respectively., Conclusions: We found significant heterogeneity of PD-L1 expression in both ADC and SCC samples, especially in the IC compartment. Heterogeneous expression of PD-L1 could misclassify patients for PD-1/PD-L1-directed therapies., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
116. Effect of MAF amplification on treatment outcomes with adjuvant zoledronic acid in early breast cancer: a secondary analysis of the international, open-label, randomised, controlled, phase 3 AZURE (BIG 01/04) trial.
- Author
-
Coleman R, Hall A, Albanell J, Hanby A, Bell R, Cameron D, Dodwell D, Marshall H, Jean-Mairet J, Tercero JC, Rojo F, Gregory W, and Gomis RR
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms genetics, Breast Neoplasms pathology, Chemotherapy, Adjuvant, Diphosphonates adverse effects, Disease-Free Survival, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Follow-Up Studies, Humans, Imidazoles adverse effects, Kaplan-Meier Estimate, Mastectomy methods, Maximum Tolerated Dose, Middle Aged, Neoplasm Invasiveness pathology, Neoplasm Staging, Proportional Hazards Models, Prospective Studies, Proto-Oncogene Proteins c-maf genetics, Survival Analysis, Treatment Outcome, Zoledronic Acid, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms mortality, Diphosphonates therapeutic use, Imidazoles therapeutic use, Proto-Oncogene Proteins c-maf drug effects
- Abstract
Background: Adjuvant use of bisphosphonates can reduce the incidence of bone metastases in early breast cancer. Recurrence and survival seem to be improved only in postmenopausal patients, but the underlying mechanisms remain unclear. We investigated whether MAF amplification (a biomarker for bone metastasis) in primary tumours could predict the treatment outcomes of adjuvant zoledronic acid., Methods: The study population included patients enrolled in the international, open-label, randomised, controlled, phase 3 AZURE trial at eligible UK sites who had stage II or III breast cancer and who gave consent for use of their primary tumour samples. Patients were randomly assigned (1:1) to receive standard adjuvant systemic therapy alone (control group) or with zoledronic acid every 3-4 weeks for six doses, then every 3-6 months until the end of 5 years. Minimisation took into account the number of involved axillary lymph nodes, clinical tumour stage, oestrogen-receptor status, type and timing of systemic therapy, menopausal status, statin use, and treating centre. The primary endpoint was disease-free survival; the secondary endpoint, invasive-disease-free survival, was the primary disease endpoint for the analysis in this report. MAF amplification was assessed by fluorescence in-situ hybridisation of two cores of breast tumour tissue in a microarray, done in a central laboratory by technicians unaware of treatment assignment. We used multivariate analyses to assess disease outcomes by intention to treat. We also assessed interactions between MAF-positive status and menopausal status on efficacy of zoledronic acid. The AZURE trial is registered with the International Standard Randomised Controlled Trial Registry, number ISRCTN79831382., Findings: 1739 AZURE patients contributed primary tumour samples, of whom 865 (50%) had two assessable cores (445 in the control groups and 420 in the zoledronic acid group). 184 (21%) tumours were MAF positive (85 in the control groups and 99 in the zoledronic acid group) and the remaining tumours were MAF negative. At a median follow-up of 84·6 months (IQR 72·0-95·8), MAF status was not prognostic for invasive-disease-free survival in the control group (MAF-positive vs MAF-negative: hazard ratio [HR] 0·92, 95% CI 0·59-1·41), but was in the zoledronic acid group (0·52, 0·36-0·75). In patients with MAF-negative tumours, zoledronic acid was associated with higher invasive-disease-free survival than was control treatment (HR 0·74, 95% CI 0·56-0·98), but not in patients who had MAF-positive tumours. Additionally, among 121 patients not postmenopausal at randomisation with MAF-positive tumours, zoledronic acid was associated with lower invasive-disease-free survival (HR 2·47, 95% CI 1·23-4·97) and overall survival (2·27, 95% CI 1·04-4·93) than control treatment., Interpretation: MAF status can predict likelihood of benefit from adjuvant zoledronic acid and merits further investigation as a potential companion diagnostic., Funding: Novartis Global and Inbiomotion., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
117. A PAM50-Based Chemoendocrine Score for Hormone Receptor-Positive Breast Cancer with an Intermediate Risk of Relapse.
- Author
-
Prat A, Lluch A, Turnbull AK, Dunbier AK, Calvo L, Albanell J, de la Haba-Rodríguez J, Arcusa A, Chacón JI, Sánchez-Rovira P, Plazaola A, Muñoz M, Paré L, Parker JS, Ribelles N, Jimenez B, Bin Aiderus AA, Caballero R, Adamo B, Dowsett M, Carrasco E, Martín M, Dixon JM, Perou CM, and Alba E
- Subjects
- Aged, Breast Neoplasms genetics, Breast Neoplasms pathology, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Middle Aged, Neoadjuvant Therapy adverse effects, Neoplasm Proteins genetics, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Receptor, ErbB-2 genetics, Receptors, Estrogen genetics, Receptors, Progesterone genetics, Recurrence, Antineoplastic Agents, Hormonal administration & dosage, Breast Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Prognosis
- Abstract
Purpose: Hormone receptor-positive (HR
+ ) breast cancer is clinically and biologically heterogeneous, and subgroups with different prognostic and treatment sensitivities need to be identified. Experimental Design: Research-based PAM50 subtyping and expression of additional genes was performed on 63 patients with HR+ /HER2- disease randomly assigned to neoadjuvant multiagent chemotherapy versus endocrine therapy in a phase II trial. The biology associated with treatment response was used to derive a PAM50-based chemoendocrine score (CES). CES's predictive ability was evaluated in 4 independent neoadjuvant data sets ( n = 675) and 4 adjuvant data sets ( n = 1,505). The association of CES, intrinsic biology, and PAM50 risk of relapse (ROR) was explored across 6,007 tumors. Results: Most genes associated with endocrine sensitivity were also found associated with chemotherapy resistance. In the chemotherapy test/validation data sets, CES was independently associated with pathologic complete response (pCR), even after adjusting for intrinsic subtype. pCR rates of the CES endocrine-sensitive (CES-E), uncertain (CES-U), and chemotherapy-sensitive (CES-C) groups in both data sets combined were 25%, 11%, and 2%, respectively. In the endocrine test/validation data sets, CES was independently associated with response. Compared with ROR, >90% of ROR-low and ROR-high tumors were identified as CES-E and CES-C, respectively; however, each CES group represented >25% of ROR-intermediate disease. In terms of survival outcome, CES-C was associated with poor relapse-free survival in patients with ROR-intermediate disease treated with either adjuvant endocrine therapy only or no adjuvant systemic therapy, but not in patients treated with (neo)adjuvant chemotherapy. Conclusions: CES is a genomic signature capable of estimating chemoendocrine sensitivity in HR+ breast cancer beyond intrinsic subtype and risk of relapse. Clin Cancer Res; 23(12); 3035-44. ©2016 AACR ., (©2016 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
118. CIP2A confirms its prognostic value in triple-negative breast cancer.
- Author
-
Cristóbal I, Zazo S, Torrejón B, Pedregal M, Madoz-Gúrpide J, Lluch A, Eroles P, Rovira A, Albanell J, García-Foncillas J, and Rojo F
- Subjects
- Autoantigens, Biomarkers, Tumor, Breast Neoplasms, Humans, Prognosis, Cell Line, Tumor, Triple Negative Breast Neoplasms
- Published
- 2017
- Full Text
- View/download PDF
119. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients.
- Author
-
Vidal J, Muinelo L, Dalmases A, Jones F, Edelstein D, Iglesias M, Orrillo M, Abalo A, Rodríguez C, Brozos E, Vidal Y, Candamio S, Vázquez F, Ruiz J, Guix M, Visa L, Sikri V, Albanell J, Bellosillo B, López R, and Montagut C
- Subjects
- Antineoplastic Agents therapeutic use, Colorectal Neoplasms blood, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, ErbB Receptors antagonists & inhibitors, Humans, Monitoring, Physiologic methods, Neoplasm Metastasis, Prospective Studies, Retrospective Studies, Colorectal Neoplasms diagnosis, DNA Mutational Analysis methods, DNA, Neoplasm blood, Genes, ras
- Abstract
Background: RAS assessment is mandatory for therapy decision in metastatic colorectal cancer (mCRC) patients. This determination is based on tumor tissue, however, genotyping of circulating tumor (ct)DNA offers clear advantages as a minimally invasive method that represents tumor heterogeneity. Our study aims to evaluate the use of ctDNA as an alternative for determining baseline RAS status and subsequent monitoring of RAS mutations during therapy as a component of routine clinical practice., Patients and Methods: RAS mutational status in plasma was evaluated in mCRC patients by OncoBEAM™ RAS CRC assay. Concordance of results in plasma and tissue was retrospectively evaluated. RAS mutations were also prospectively monitored in longitudinal plasma samples from selected patients., Results: Analysis of RAS in tissue and plasma samples from 115 mCRC patients showed a 93% overall agreement. Plasma/tissue RAS discrepancies were mainly explained by spatial and temporal tumor heterogeneity. Analysis of clinico-pathological features showed that the site of metastasis (i.e. peritoneal, lung), the histology of the tumor (i.e. mucinous) and administration of treatment previous to blood collection negatively impacted the detection of RAS in ctDNA. In patients with baseline mutant RAS tumors treated with chemotherapy/antiangiogenic, longitudinal analysis of RAS ctDNA mirrored response to treatment, being an early predictor of response. In patients RAS wt, longitudinal monitoring of RAS ctDNA revealed that OncoBEAM was useful to detect emergence of RAS mutations during anti-EGFR treatment., Conclusion: The high overall agreement in RAS mutational assessment between plasma and tissue supports blood-based testing with OncoBEAM™ as a viable alternative for genotyping RAS of mCRC patients in routine clinical practice. Our study describes practical clinico-pathological specifications to optimize RAS ctDNA determination. Moreover, OncoBEAM™ is useful to monitor RAS in patients undergoing systemic therapy to detect resistance and evaluate the efficacy of particular treatments., (© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
120. Intrinsic Subtypes and Gene Expression Profiles in Primary and Metastatic Breast Cancer.
- Author
-
Cejalvo JM, Martínez de Dueñas E, Galván P, García-Recio S, Burgués Gasión O, Paré L, Antolín S, Martinello R, Blancas I, Adamo B, Guerrero-Zotano Á, Muñoz M, Nucíforo P, Vidal M, Pérez RM, Chacón López-Muniz JI, Caballero R, Peg V, Carrasco E, Rojo F, Perou CM, Cortés J, Adamo V, Albanell J, Gomis RR, Lluch A, and Prat A
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor biosynthesis, Breast Neoplasms classification, Breast Neoplasms pathology, Estrogens genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Middle Aged, Neoplasm Metastasis, Neoplasm Proteins biosynthesis, Neoplasm Recurrence, Local pathology, Prognosis, Transcriptome, Biomarkers, Tumor genetics, Breast Neoplasms genetics, Neoplasm Proteins genetics, Neoplasm Recurrence, Local genetics, Receptor, ErbB-2 genetics
- Abstract
Biological changes that occur during metastatic progression of breast cancer are still incompletely characterized. In this study, we compared intrinsic molecular subtypes and gene expression in 123 paired primary and metastatic tissues from breast cancer patients. Intrinsic subtype was identified using a PAM50 classifier and χ
2 tests determined the differences in variable distribution. The rate of subtype conversion was 0% in basal-like tumors, 23.1% in HER2-enriched (HER2-E) tumors, 30.0% in luminal B tumors, and 55.3% in luminal A tumors. In 40.2% of cases, luminal A tumors converted to luminal B tumors, whereas in 14.3% of cases luminal A and B tumors converted to HER2-E tumors. We identified 47 genes that were expressed differentially in metastatic versus primary disease. Metastatic tumors were enriched for proliferation-related and migration-related genes and diminished for luminal-related genes. Expression of proliferation-related genes were better at predicting overall survival in metastatic disease (OSmet) when analyzed in metastatic tissue rather than primary tissue. In contrast, a basal-like gene expression signature was better at predicting OSmet in primary disease compared with metastatic tissue. We observed correlations between time to tumor relapse and the magnitude of changes of proliferation, luminal B, or HER2-E signatures in metastatic versus primary disease. Although the intrinsic subtype was largely maintained during metastatic progression, luminal/HER2-negative tumors acquired a luminal B or HER2-E profile during metastatic progression, likely reflecting tumor evolution or acquisition of estrogen independence. Overall, our analysis revealed the value of stratifying gene expression by both cancer subtype and tissue type, providing clinicians more refined tools to evaluate prognosis and treatment. Cancer Res; 77(9); 2213-21. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
121. A definition for aggressive disease in patients with HER-2 negative metastatic breast cancer: an expert consensus of the Spanish Society of Medical Oncology (SEOM).
- Author
-
González A, Lluch A, Aba E, Albanell J, Antón A, Álvarez I, Ayala F, Barnadas A, Calvo L, Ciruelos E, Cortés J, de la Haba J, López-Vega JM, Martínez E, Muñoz M, Peláez I, Redondo A, Rodríguez Á, Rodríguez CA, Ruíz A, and Llombart A
- Subjects
- Consensus, Delphi Technique, Female, Humans, Receptor, ErbB-2, Societies, Medical, Breast Neoplasms classification, Breast Neoplasms pathology, Medical Oncology standards
- Abstract
Purpose: To converge on an expert opinion to define aggressive disease in patients with HER2-negative mBC using a modified Delphi methodology., Methods: A panel of 21 breast cancer experts from the Spanish Society of Medical Oncology agreed upon a survey which comprised 47 questions that were grouped into three sections: relevance for defining aggressive disease, aggressive disease criteria and therapeutic goals. Answers were rated using a 9-point Likert scale of relevance or agreement., Results: Among the 88 oncologists that were invited to participate, 81 answered the first round (92%), 70 answered the second round (80%), and 67 answered the third round (76%) of the survey. There was strong agreement regarding the fact that identifying patients with aggressive disease needs to be adequately addressed to help practitioners to decide the best treatment options for patients with HER2-negative mBC. The factors that were considered to be strongly relevant to classifying patients with aggressive HER2-negative mBC were a high tumor burden, a disease-free interval of less than 12-24 months after surgery, the presence of progressive disease during adjuvant or neoadjuvant chemotherapy and having a triple-negative phenotype. The main therapeutic goals were controlling symptoms, improving quality of life and increasing the time to progression and overall survival., Conclusions: High tumor burden, time to recurrence after prior therapy and having a triple-negative phenotype were the prognostic factors for which the greatest consensus was found for identifying patients with aggressive HER2-negative mBC. Identifying patients with aggressive disease leads to different therapeutic approaches.
- Published
- 2017
- Full Text
- View/download PDF
122. Limitations in predicting PAM50 intrinsic subtype and risk of relapse score with Ki67 in estrogen receptor-positive HER2-negative breast cancer.
- Author
-
Fernandez-Martinez A, Pascual T, Perrone G, Morales S, de la Haba J, González-Rivera M, Galván P, Zalfa F, Amato M, Gonzalez L, Prats M, Rojo F, Manso L, Paré L, Alonso I, Albanell J, Vivancos A, González A, Matito J, González S, Fernandez P, Adamo B, Muñoz M, Viladot M, Font C, Aya F, Vidal M, Caballero R, Carrasco E, Altomare V, Tonini G, Prat A, and Martin M
- Subjects
- Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms genetics, Chemotherapy, Adjuvant, Female, Follow-Up Studies, Gene Expression Profiling, Humans, Incidence, Neoplasm Recurrence, Local epidemiology, Neoplasm Recurrence, Local genetics, Prognosis, Prospective Studies, Biomarkers, Tumor genetics, Breast Neoplasms drug therapy, Neoplasm Recurrence, Local diagnosis, Receptor, ErbB-2 genetics, Receptors, Estrogen genetics, Risk Assessment methods, Tamoxifen therapeutic use
- Abstract
PAM50/Prosigna gene expression-based assay identifies three categorical risk of relapse groups (ROR-low, ROR-intermediate and ROR-high) in post-menopausal patients with estrogen receptor estrogen receptor-positive (ER+)/ HER2-negative (HER2-) early breast cancer. Low risk patients might not need adjuvant chemotherapy since their risk of distant relapse at 10-years is below 10% with endocrine therapy only. In this study, 517 consecutive patients with ER+/HER2- and node-negative disease were evaluated for Ki67 and Prosigna. Most of Luminal A tumors (65.6%) and ROR-low tumors (70.9%) had low Ki67 values (0-10%); however, the percentage of patients with ROR-medium or ROR-high disease within the Ki67 0-10% group was 42.7% (with tumor sizes ≤2 cm) and 33.9% (with tumor sizes > 2 cm). Finally, we found that the optimal Ki67 cutoff for identifying Luminal A or ROR-low tumors was 14%. Ki67 as a surrogate biomarker in identifying Prosigna low-risk outcome patients or Luminal A disease in the clinical setting is unreliable. In the absence of a well-validated prognostic gene expression-based assay, the optimal Ki67 cutoff for identifying low-risk outcome patients or Luminal A disease remains at 14%.
- Published
- 2017
- Full Text
- View/download PDF
123. The role of miR-26a and miR-30b in HER2+ breast cancer trastuzumab resistance and regulation of the CCNE2 gene.
- Author
-
Tormo E, Adam-Artigues A, Ballester S, Pineda B, Zazo S, González-Alonso P, Albanell J, Rovira A, Rojo F, Lluch A, and Eroles P
- Subjects
- Cell Cycle drug effects, Cell Cycle genetics, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Cyclins metabolism, Down-Regulation drug effects, Down-Regulation genetics, Drug Resistance, Neoplasm genetics, Female, Gene Silencing, Humans, MicroRNAs genetics, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Trastuzumab pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Cyclins genetics, Gene Expression Regulation, Neoplastic drug effects, MicroRNAs metabolism, Receptor, ErbB-2 metabolism, Trastuzumab therapeutic use
- Abstract
A subset of HER2+ breast cancer patients manifest clinical resistance to trastuzumab. Recently, miR-26a and miR-30b have been identified as trastuzumab response regulators, and their target gene CCNE2 seems to play an important role in resistance to trastuzumab therapy. Cell viability was evaluated in trastuzumab treated HER2+ BT474 wt (sensitive), BT474r (acquired resistance), HCC1954 (innate resistance), and MDA-MB-231 (HER2-) cell lines, and the expression of miR-26a, miR-30b, and their target genes was measured. BT474 wt cell viability decreased by 60% and miR-26a and miR-30b were significantly overexpressed (~3-fold, p = 0.003 and p = 0.002, respectively) after trastuzumab treatment, but no differences were observed in resistant and control cell lines. Overexpression of miR-30b sensitized BT474r cells to trastuzumab (p = 0.01) and CCNE2, was significantly overexpressed after trastuzumab treatment in BT474r cells (p = 0.032), but no significant changes were observed in sensitive cell line. When CCNE2 was silenced BT474r cell sensitivity to trastuzumab increased (p = 0.03). Thus, the molecular mechanism of trastuzumab action in BT474 cell line may be regulated by miR-26a and miR-30b and CCNE2 overexpression might play an important role in acquired trastuzumab resistance in HER2+ breast cancer given that resistance was diminished when CCNE2 was silenced.
- Published
- 2017
- Full Text
- View/download PDF
124. Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer.
- Author
-
Van Emburgh BO, Arena S, Siravegna G, Lazzari L, Crisafulli G, Corti G, Mussolin B, Baldi F, Buscarino M, Bartolini A, Valtorta E, Vidal J, Bellosillo B, Germano G, Pietrantonio F, Ponzetti A, Albanell J, Siena S, Sartore-Bianchi A, Di Nicolantonio F, Montagut C, and Bardelli A
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents, Immunological pharmacology, Clonal Evolution, Colorectal Neoplasms genetics, Female, Humans, Male, Middle Aged, Mutation, Antineoplastic Agents, Immunological therapeutic use, Colorectal Neoplasms drug therapy, Drug Resistance, Neoplasm genetics, ErbB Receptors antagonists & inhibitors, Genes, erbB-1, Genes, ras
- Abstract
Blockade of the epidermal growth factor receptor (EGFR) with the monoclonal antibodies cetuximab or panitumumab is effective in a subset of colorectal cancers (CRCs), but the emergence of resistance limits the efficacy of these therapeutic agents. At relapse, the majority of patients develop RAS mutations, while a subset acquires EGFR extracellular domain (ECD) mutations. Here we find that patients who experience greater and longer responses to EGFR blockade preferentially develop EGFR ECD mutations, while RAS mutations emerge more frequently in patients with smaller tumour shrinkage and shorter progression-free survival. In circulating cell-free tumour DNA of patients treated with anti-EGFR antibodies, RAS mutations emerge earlier than EGFR ECD variants. Subclonal RAS but not EGFR ECD mutations are present in CRC samples obtained before exposure to EGFR blockade. These data indicate that clonal evolution of drug-resistant cells is associated with the clinical outcome of CRC patients treated with anti-EGFR antibodies., Competing Interests: A.S.-B. is a member of advisory boards for Amgen, Bayer, Lilly and Sanofi. S.S. is a member of advisory boards for Amgen, Roche, Bayer, Sanofi, Eli Lilly and Merck. The remaining authors declare no competing financial interests.
- Published
- 2016
- Full Text
- View/download PDF
125. Generation, characterization, and maintenance of trastuzumab-resistant HER2+ breast cancer cell lines.
- Author
-
Zazo S, González-Alonso P, Martín-Aparicio E, Chamizo C, Cristóbal I, Arpí O, Rovira A, Albanell J, Eroles P, Lluch A, Madoz-Gúrpide J, and Rojo F
- Abstract
Trastuzumab became the therapy of choice for patients with HER2-positive breast cancer in 1998, and it has provided clinical benefit ever since. However, a significant percentage of patients show primary resistance to trastuzumab at diagnosis, and most patients with metastatic disease that initially respond to trastuzumab eventually progress (acquired resistance). Consequently, there is an urgent need to improve our knowledge of the mechanisms governing resistance, so that specific therapeutic strategies can be developed to provide improved efficacy. We generated new cell lines derived from BCCL through extended exposure to trastuzumab. Drug-conditioned populations were authenticated for their molecular profile and their resistance rate was determined. Heterogeneous HER2 amplification was observed across most of the BCCLs, ranging from cells without HER2 amplification to elevated HER2 gene copy numbers in others. Using a phospho-antibody array we analyzed the status of kinase receptors and effectors from different cellular pathways. This revealed that HER2, AKT, and S6RP presented high phosphorylation levels with specific variations between sensitive and resistant populations. In addition, differences in phosphorylation levels for several of those pathways targets were found between sensitive and resistant lines. Furthermore, a biochemical study characterized patterns of molecular alterations similar to those commonly described in breast cancer. Finally, a subcutaneous xenograft murine model confirmed the resistance to trastuzumab of the established cell line. We conclude that these resistant BCCLs can be a valuable tool to gain insight into the mechanisms of acquisition of trastuzumab resistance.
- Published
- 2016
126. c-Jun N-Terminal Kinase Inactivation by Mitogen-Activated Protein Kinase Phosphatase 1 Determines Resistance to Taxanes and Anthracyclines in Breast Cancer.
- Author
-
Rincón R, Zazo S, Chamizo C, Manso R, González-Alonso P, Martín-Aparicio E, Cristóbal I, Cañadas C, Perona R, Lluch A, Eroles P, García-Foncillas J, Albanell J, Rovira A, Madoz-Gúrpide J, and Rojo F
- Subjects
- Antineoplastic Combined Chemotherapy Protocols, Apoptosis drug effects, Biomarkers, Tumor, Breast Neoplasms diagnosis, Breast Neoplasms genetics, Breast Neoplasms mortality, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Cluster Analysis, Dual Specificity Phosphatase 1 genetics, Female, Gene Expression, Gene Expression Profiling, Humans, Kaplan-Meier Estimate, Neoplasm Grading, Prognosis, Proportional Hazards Models, Recurrence, Anthracyclines pharmacology, Antineoplastic Agents pharmacology, Breast Neoplasms metabolism, Drug Resistance, Neoplasm, Dual Specificity Phosphatase 1 metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Taxoids pharmacology
- Abstract
MAPK phosphatase-1 (MKP-1) is overexpressed during malignant transformation of the breast in many patients, and it is usually associated with chemoresistance through interference with JNK-driven apoptotic pathways. Although the molecular settings of the mechanism have been documented, details about the contribution of MKP-1 to the failure of chemotherapeutic interventions are unclear. Transient overexpression of MKP-1 and treatment with JNK-modulating agents in breast carcinoma cells confirmed the mediation of MKP-1 in the resistance to taxanes and anthracyclines in breast cancer, through the inactivation of JNK1/2. We next assessed MKP-1 expression and JNK1/2 phosphorylation status in a large cohort of samples from 350 early breast cancer patients treated with adjuvant anthracycline-based chemotherapy. We detected that MKP-1 overexpression is a recurrent event predominantly linked to dephosphorylation of JNK1/2 with an adverse impact on relapse of the tumor and overall and disease-free survival. Moreover, MKP-1 and p-JNK1/2 determinations in 64 locally advanced breast cancer patients treated with neoadjuvant taxane-based chemotherapy showed an inverse correlation between MKP-1 overexpression (together with JNK1/2 inhibition) and the pathologic response of the tumors. Our results emphasize the importance of MKP-1 as a potential predictive biomarker for a subset of breast cancer patients with worse outcome and less susceptibility to treatment. Mol Cancer Ther; 15(11); 2780-90. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
127. Pooled analysis of prospective European studies assessing the impact of using the 21-gene Recurrence Score assay on clinical decision making in women with oestrogen receptor-positive, human epidermal growth factor receptor 2-negative early-stage breast cancer.
- Author
-
Albanell J, Svedman C, Gligorov J, Holt SD, Bertelli G, Blohmer JU, Rouzier R, Lluch A, and Eiermann W
- Subjects
- Adult, Aged, Aged, 80 and over, Breast Neoplasms pathology, Breast Neoplasms therapy, Europe, Female, Gene Expression Profiling, Humans, Middle Aged, Neoplasm Recurrence, Local drug therapy, Precision Medicine methods, Prospective Studies, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Risk Assessment, Severity of Illness Index, Tumor Burden, Breast Neoplasms drug therapy, Clinical Decision-Making methods
- Abstract
Purpose: The 21-gene Recurrence Score assay (Oncotype DX) provides prognostic/predictive information in oestrogen receptor positive (ER+) early breast cancer, but access/reimbursement has been limited in most European countries in the absence of prospective outcome data. Recently, two large prospective studies and a real-life 5-year outcome study have been reported. We performed a pooled analysis of prospective European impact studies to generate robust data on impact of use in different clinical subgroups., Methods: The analysis included four studies (French, German, Spanish, and British) in ER+ human epidermal growth factor receptor 2-negative breast cancer patients (n = 527). Node-positive patients were excluded., Results: The analysis demonstrated that treatment recommendations changed in 32% of patients post-testing; chemotherapy recommendation rate decreased from 55% to 34%. Change rates in the individual studies ranged from 30% to 37%. The highest change rates were in patients originally recommended chemotherapy and in grade II tumours; there was no subgroup without a treatment recommendation change. Notably, 31% of patients with an intermediate Recurrence Score result had a treatment recommendation change suggesting that testing provides actionable information in this group. With the exception of the German study (where chemotherapy rates remained high [41%] post-testing), between-study variability in treatment recommendations decreased post-testing (chemotherapy: from 36-52% to 26-29%; hormonal therapy: from 48-64% to 71-74%). Physicians' confidence regarding treatment recommendations improved in all the studies after testing., Conclusion: Recurrence Score testing led to changes in adjuvant chemotherapy use in approximately a third of patients, to an overall reduced chemotherapy use, and to more homogeneous decision making., (Copyright © 2016 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
128. Is Metastatic Disease the Best Setting for Cost-Effectiveness Studies?
- Author
-
Colomer R, Pascual T, and Albanell J
- Subjects
- Antibodies, Monoclonal, Humanized, Humans, Quality-Adjusted Life Years, Cetuximab, Cost-Benefit Analysis
- Published
- 2016
- Full Text
- View/download PDF
129. Non-canonical NF-κB pathway activation predicts outcome in borderline oestrogen receptor positive breast carcinoma.
- Author
-
Rojo F, González-Pérez A, Furriol J, Nicolau MJ, Ferrer J, Burgués O, Sabbaghi M, González-Navarrete I, Cristobal I, Serrano L, Zazo S, Madoz J, Servitja S, Tusquets I, Albanell J, Lluch A, Rovira A, and Eroles P
- Subjects
- Adult, Aged, Aged, 80 and over, Cohort Studies, Female, Humans, Middle Aged, Prognosis, Breast Neoplasms metabolism, NF-kappa B metabolism, Receptors, Estrogen metabolism
- Abstract
Background: NF-κB signalling appears deregulated in breast tumours. The purpose of this study was to determine whether the non-canonical NF-κB pathway, is activated in oestrogen receptor positive (ER+) breast cancer, to identify any correlation between its activity and the clinico-pathological phenotype and to explore whether NF-κB2 and RelB subunits and/or any of their target genes might be used as a predictive marker., Methods: Two independent cohorts of ER+ early breast cancer patients treated with adjuvant endocrine therapy were included in the study. Activation of RelB and NF-κB2 subunits was determined in a training set of 121 patients by measuring DNA-binding activities in nuclear extracts from fresh frozen specimens by an ELISA-based assay. Samples of 15 ER- breast cancer patients were also included in the study. In a large validation cohort of 207 patients, nuclear immunostaining of RelB and NF-κB2 on formalin-fixed paraffin-embedded specimens was performed. Statistical correlation within clinico-pathological factors, disease-free survival (DFS) and overall survival (OS) was evaluated. Publicly available gene expression and survival data have been interrogated aimed to identify target genes., Results: Activation of NF-κB2 and RelB was found in 53.7 and 49.2% of the 121 ER+ tumours analysed, with similar levels to ER- breast tumours analysed in parallel for comparisons. In the validation cohort, we obtained a similar proportion of cases with activation of NF-κB2 and RelB (59.9 and 32.4%), with a 39.6% of co-activation. Multiplexing immunofluorescence in breast cancer tissue confirmed an inverse spatial distribution of ER with NF-κB2 and RelB nuclear expression in tumour cells. Interestingly, NF-κB2 and RelB mRNA expression was inversely correlated with ER gene (ESR1) levels (P<0.001, both) and its activation was significantly associated with worse DFS (P=0.005 and P=0.035, respectively) in ER+ breast cancer. Moreover, the co-activation of both subunits showed a stronger association with early relapse (P=0.002) and OS (P=0.001). Finally, higher expression of the non-canonical NF-κB target gene myoglobin was associated with a poor outcome in ER+ breast cancer (DFS, P<0.05)., Conclusions: The non-canonical NF-κB pathway activation is inversely associated with oestrogen receptor expression in ER+ breast cancer and predicts poor survival in this subgroup. The myoglobin gene expression has been identified as a possible surrogate marker of the non-canonical NF-κB pathway activation in these tumours.
- Published
- 2016
- Full Text
- View/download PDF
130. Trastuzumab emtansine (T-DM1) plus docetaxel with or without pertuzumab in patients with HER2-positive locally advanced or metastatic breast cancer: results from a phase Ib/IIa study.
- Author
-
Martin M, Fumoleau P, Dewar JA, Albanell J, Limentani SA, Campone M, Chang JC, Patre M, Strasak A, de Haas SL, Xu J, and Garcia-Saenz JA
- Subjects
- Ado-Trastuzumab Emtansine, Adult, Aged, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms pathology, Disease-Free Survival, Docetaxel, Female, Granulocyte Colony-Stimulating Factor genetics, Humans, Maytansine administration & dosage, Maytansine adverse effects, Maytansine pharmacokinetics, Middle Aged, Neoplasm Metastasis, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Taxoids adverse effects, Taxoids pharmacokinetics, Trastuzumab adverse effects, Trastuzumab pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Breast Neoplasms drug therapy, Maytansine analogs & derivatives, Receptor, ErbB-2 genetics, Taxoids administration & dosage, Trastuzumab administration & dosage
- Abstract
Background: Trastuzumab emtansine (T-DM1) exhibited enhanced antitumor activity when combined with docetaxel or pertuzumab in preclinical studies. This phase Ib/IIa study assessed the feasibility of T-DM1 + docetaxel in patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC) and T-DM1 + docetaxel ± pertuzumab in patients with HER2-positive locally advanced breast cancer (LABC)., Patients and Methods: Phase Ib (part 1) explored dose escalation, with T-DM1 + docetaxel administered for greater than or equal to six cycles in patients with MBC. Phase Ib (part 2) began with the maximum tolerated dose (MTD) identified in part 1. Patients with LABC were administered less than or equal to six cycles of T-DM1 + docetaxel or T-DM1 + docetaxel + pertuzumab. Phase IIa explored the MTDs identified in phase Ib., Results: Administered with T-DM1 (3.6 mg/kg), the docetaxel MTD was 60 mg/m(2) in MBC. In LABC, the MTD was 100 mg/m(2) docetaxel in combination with T-DM1 (3.6 mg/kg), given with granulocyte colony-stimulating factor (G-CSF). Administered with T-DM1 (3.6 mg/kg) + pertuzumab (840 mg, cycle 1; 420 mg, subsequent cycles), the docetaxel MTD in LABC was 75 mg/m(2) with G-CSF support. Neutropenia was the most common grade 3-4 adverse event (AE; MBC, 72% and LABC, 29%). In total, 48% (12/25) of MBC patients and 47% (34/73) of LABC patients experienced AEs requiring dose modification. In MBC (median prior systemic agents = 5), the objective response rate was 80.0% (20/25; 95% confidence interval [CI] 59.3-93.2) and the median progression-free survival was 13.8 months (range, 1.6-33.5). The pathologic complete response (ypT0/is, ypN0) rate in LABC was 60.3% (44/73; 95% CI 48.1-71.5). Pharmacokinetic analyses indicated a low risk of drug-drug interaction between T-DM1 and docetaxel., Conclusions: T-DM1 combined with docetaxel ± pertuzumab appeared efficacious in MBC or LABC; however, nearly half of patients experienced AEs requiring dose reductions with these T-DM1 combinations., Clinicaltrialsgov Identifier: NCT00934856., (© The Author 2016. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
131. The First-in-class Anti-EGFR Antibody Mixture Sym004 Overcomes Cetuximab Resistance Mediated by EGFR Extracellular Domain Mutations in Colorectal Cancer.
- Author
-
Sánchez-Martín FJ, Bellosillo B, Gelabert-Baldrich M, Dalmases A, Cañadas I, Vidal J, Martinez A, Argilés G, Siravegna G, Arena S, Koefoed K, Visa L, Arpí O, Horak ID, Iglesias M, Stroh C, Kragh M, Rovira A, Albanell J, Tabernero J, Bardelli A, and Montagut C
- Subjects
- 3T3 Cells, Animals, Antibodies, Monoclonal immunology, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm drug effects, ErbB Receptors genetics, ErbB Receptors immunology, Humans, Male, Mice, Mice, Inbred BALB C, Mutation, Panitumumab, Signal Transduction drug effects, Xenograft Model Antitumor Assays methods, Antibodies, Monoclonal therapeutic use, Cetuximab therapeutic use, Colorectal Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors
- Abstract
Purpose: Approved anti-EGFR antibodies cetuximab and panitumumab provide significant clinical benefit in patients with metastatic colorectal cancer (MCRC). However, patients ultimately develop disease progression, often driven by acquisition of mutations in the extracellular domain (ECD) of EGFR. Sym004 is a novel 1:1 mixture of two nonoverlapping anti-EGFR mAbs that recently showed promising clinical activity in a phase I trial in MCRC. Our aim was to determine the efficacy of Sym004 to circumvent cetuximab resistance driven by EGFR ECD mutations., Experimental Design: Functional studies were performed to assess drug-receptor binding as well as ligand-dependent activation of individual EGFR mutants in the presence of cetuximab, panitumumab, and Sym004. Cell viability and molecular effects of the drugs were assayed in cetuximab-resistant cell lines and in tumor xenograft models. Efficacy of Sym004 was evaluated in patients progressing to cetuximab that harbored EGFR mutation in the post-cetuximab tumor sample., Results: Contrary to cetuximab and panitumumab, Sym004 effectively bound and abrogated ligand-induced phosphorylation of all individual EGFR mutants. Cells resistant to cetuximab harboring mutations in EGFR maintained sensitivity to Sym004, which was consistent with an effective suppression of EGFR downstream signaling, translating into profound and sustained tumor regression in the xenograft model. As proof-of-principle, a patient with a tumor harboring an EGFR mutation (G465R) following cetuximab therapy benefited from Sym004 therapy., Conclusions: Sym004 is an active drug in MCRC resistant to cetuximab/panitumumab mediated by EGFR mutations. EGFR mutations are potential biomarkers of response to Sym004 to be evaluated in ongoing large clinical trials. Clin Cancer Res; 22(13); 3260-7. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
132. High Proliferation Predicts Pathological Complete Response to Neoadjuvant Chemotherapy in Early Breast Cancer.
- Author
-
Alba E, Lluch A, Ribelles N, Anton-Torres A, Sanchez-Rovira P, Albanell J, Calvo L, García-Asenjo JA, Palacios J, Chacon JI, Ruiz A, De la Haba-Rodriguez J, Segui-Palmer MA, Cirauqui B, Margeli M, Plazaola A, Barnadas A, Casas M, Caballero R, Carrasco E, and Rojo F
- Published
- 2016
- Full Text
- View/download PDF
133. Quality Indicators to Assure and Improve Cancer Care in Spain Using the Delphi Technique.
- Author
-
Camps C, Albanell J, Antón A, Aranda E, Carrato A, Cassinello J, Castellano D, Cruz JJ, Garrido P, Guillem V, Grávalos C, López G, Llorente C, Lorenzo A, Lluch A, Ignacio E, and Díaz-Rubio E
- Subjects
- Aged, Humans, Spain, Delphi Technique, Neoplasms therapy, Quality Indicators, Health Care trends
- Abstract
Background: The quality of cancer care has become a priority for health care systems. The goal of this research was to develop a set of evidence-based quality indicators (QIs) for organization, palliative care, and colorectal, breast, and lung cancers for introducing a system of benchmarking in Spain., Methods: A comprehensive evidence-based literature search was performed to identify potential QIs. An expert panel (the health care quality promotion group) of 9 oncologists identified indicators and evaluated them. A Delphi process involving 58 physicians was used to rank QIs by clinical relevance (validity). The expert panel then evaluated the selected indicators in terms of the feasibility of measuring them in Spanish hospitals, their usefulness for comparisons, their degree of clinical relevance, and their sensitivity to the impact of health care improvements., Results: From the literature review, 99 potential QIs were identified. The Delphi process shortened the list to 72 QIs. A final set of 57 QIs was established by the health care quality promotion group: 12 related to organizational issues, 11 to colorectal cancer, 11 to breast cancer, 12 to lung cancer, and 11 to palliative care. This final set included structure (n=2), process (n=36), and outcome (n=19) indicators., Conclusions: A set of QIs has been developed using a validated Delphi method, meaning that we can be confident of their validity, feasibility, sensitivity, and acceptability. These QIs are to serve as the basis of a strategy for benchmarking across oncology services in Spanish hospitals and should enable us to assure and improve the quality of cancer care., (Copyright © 2016 by the National Comprehensive Cancer Network.)
- Published
- 2016
- Full Text
- View/download PDF
134. FoxA and LIPG endothelial lipase control the uptake of extracellular lipids for breast cancer growth.
- Author
-
Slebe F, Rojo F, Vinaixa M, García-Rocha M, Testoni G, Guiu M, Planet E, Samino S, Arenas EJ, Beltran A, Rovira A, Lluch A, Salvatella X, Yanes O, Albanell J, Guinovart JJ, and Gomis RR
- Subjects
- 4-Butyrolactone analogs & derivatives, 4-Butyrolactone pharmacology, Animals, Biological Transport, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Doxycycline pharmacology, Enzyme Inhibitors pharmacology, Female, Hepatocyte Nuclear Factor 3-alpha antagonists & inhibitors, Hepatocyte Nuclear Factor 3-alpha genetics, Hepatocyte Nuclear Factor 3-beta antagonists & inhibitors, Hepatocyte Nuclear Factor 3-beta genetics, Humans, Lactones pharmacology, Lipase antagonists & inhibitors, Lipase genetics, MCF-7 Cells, Mice, Mice, Nude, Neoplasm Invasiveness, Orlistat, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Signal Transduction, Xenograft Model Antitumor Assays, Breast Neoplasms metabolism, Gene Expression Regulation, Neoplastic, Hepatocyte Nuclear Factor 3-alpha metabolism, Hepatocyte Nuclear Factor 3-beta metabolism, Lipase metabolism, Lipid Metabolism genetics
- Abstract
The mechanisms that allow breast cancer (BCa) cells to metabolically sustain rapid growth are poorly understood. Here we report that BCa cells are dependent on a mechanism to supply precursors for intracellular lipid production derived from extracellular sources and that the endothelial lipase (LIPG) fulfils this function. LIPG expression allows the import of lipid precursors, thereby contributing to BCa proliferation. LIPG stands out as an essential component of the lipid metabolic adaptations that BCa cells, and not normal tissue, must undergo to support high proliferation rates. LIPG is ubiquitously and highly expressed under the control of FoxA1 or FoxA2 in all BCa subtypes. The downregulation of either LIPG or FoxA in transformed cells results in decreased proliferation and impaired synthesis of intracellular lipids.
- Published
- 2016
- Full Text
- View/download PDF
135. AI-related BMD variation in actual practice conditions: A prospective cohort study.
- Author
-
Rodríguez-Sanz M, Prieto-Alhambra D, Servitja S, Garcia-Giralt N, Garrigos L, Rodriguez-Morera J, Albanell J, Martínez-García M, González I, Diez-Perez A, Tusquets I, and Nogués X
- Subjects
- Aged, Aromatase Inhibitors therapeutic use, Bone Density drug effects, Bone Density Conservation Agents therapeutic use, Breast Neoplasms drug therapy, Diphosphonates therapeutic use, Female, Femur Neck physiology, Hip physiology, Humans, Lumbar Vertebrae physiology, Middle Aged, Osteoporosis physiopathology, Osteoporosis prevention & control, Prospective Studies, Aromatase Inhibitors adverse effects, Breast Neoplasms physiopathology, Osteoporosis chemically induced
- Abstract
The aim of the study was to evaluate the progression of bone mineral density (BMD) during 3 years of aromatase inhibitors (AI) therapy in actual practice conditions. This prospective, clinical cohort study of Barcelona-Aromatase induced Bone Loss in Early breast cancer (B-ABLE) assessed BMD changes during 3 years of AI treatment in women with breast cancer. Patients with osteoporosis (T score < -2.5 or T score ≤ -2.0) and a major risk factor and/or prevalent fragility fractures were treated with oral bisphosphonates (BPs). Of 685 women recruited, 179 (26.1%) received BP treatment. By the third year of AI therapy, this group exhibited increased BMD in the lumbar spine (LS; 2.59%) and femoral neck (FN; 2.50%), although the increase was significant only within the first year (LS: 1.99% and FN: 2.04%). Despite BP therapy, however, approximately 15% of these patients lost more than 3% of their baseline bone mass. At 3 years, patients without BP experienced BMD decreases in the LS (-3.10%) and FN (-2.79%). In this group, BMD changes occurred during the first (LS: -1.33% and FN: -1.25%), second (LS: -1.19% and FN: -0.82%), and third (LS: -0.57% and FN: -0.65%) years of AI treatment. Increased BMD (>3%) was observed in just 7.6% and 10.8% of these patients at the LS and FN, respectively. Our data confirm a clinically relevant bone loss associated with AI therapy amongst nonusers of preventative BPs. We further report on the importance of BMD monitoring as well as calcium and 25-hydroxy vitamin D supplementation in these patients., (© 2016 Society for Endocrinology.)
- Published
- 2016
- Full Text
- View/download PDF
136. ROS1 copy number alterations are frequent in non-small cell lung cancer.
- Author
-
Clavé S, Gimeno J, Muñoz-Mármol AM, Vidal J, Reguart N, Carcereny E, Pijuan L, Menéndez S, Taus Á, Mate JL, Serrano S, Albanell J, Espinet B, Arriola E, and Salido M
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Carcinoma, Large Cell metabolism, Carcinoma, Large Cell pathology, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Female, Follow-Up Studies, Humans, Immunoenzyme Techniques, In Situ Hybridization, Fluorescence, Lung Neoplasms metabolism, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Prognosis, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins metabolism, Survival Rate, Tissue Array Analysis, Adenocarcinoma genetics, Carcinoma, Large Cell genetics, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Squamous Cell genetics, DNA Copy Number Variations genetics, Gene Rearrangement, Lung Neoplasms genetics, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins genetics
- Abstract
Objectives: We aimed to determine the prevalence and partners of ROS1 rearrangements, to explore the correlation between FISH and IHC assays, and to investigate clinical implications of ROS1 copy number alterations (CNAs)., Methods: A total of 314 NSCLC patients were screened using ROS1 FISH break-apart probes. Of these, 47 surgical tumors were included in TMAs to analyze ROS1 heterogeneity assessed either by FISH and IHC, and chromosome 6 aneusomy. To characterize ROS1 partners, probes for CD74, EZR, SLC34A2 and SDC3 genes were developed. ROS1 positive FISH cases were screened also by IHC., Results: Five patients were ROS1 positive (1.8%). We identified two known fusion partners in three patients: CD74 and SLC34A2. Four out of five ROS1 rearranged patients were female, never smokers and with adenocarcinoma histology. Rearranged cases were also positive by IHC as well. According to ROS1 CNAs, we found a prevalence of 37.8% gains/amplifications and 25.1% deletions., Conclusions: This study point out the high prevalence of ROS1 CNAs in a large series of NSCLC. ROS1 gains, amplifications and deletions, most of them due to chromosome 6 polysomy or monosomy, were heterogeneous within a tumor and had no impact on overall survival.
- Published
- 2016
- Full Text
- View/download PDF
137. Treatment of HER2 positive advanced breast cancer with T-DM1: A review of the literature.
- Author
-
Martínez MT, Pérez-Fidalgo JA, Martín-Martorell P, Cejalvo JM, Pons V, Bermejo B, Martín M, Albanell J, and Lluch A
- Subjects
- Ado-Trastuzumab Emtansine, Antibodies, Monoclonal, Humanized chemistry, Antibodies, Monoclonal, Humanized pharmacokinetics, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacokinetics, Breast Neoplasms pathology, Clinical Trials, Phase III as Topic, Disease Progression, Female, Humans, Maytansine chemistry, Maytansine pharmacokinetics, Maytansine therapeutic use, Neoplasm Metastasis, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local metabolism, Trastuzumab, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Maytansine analogs & derivatives, Receptor, ErbB-2 metabolism
- Abstract
Background: Trastuzumab emtansine (T-DM1), a new agent developed for the treatment of HER2-positive breast cancer, is an antibody-drug conjugate with a complex compound obtained by the conjugation of trastuzumab, a stable thioether linker, and the potent cytotoxic drug maytansine-derivate(DM1), which inhibits cell division and induces cell death., Field of Study: PubMed database, ESMO, ASCO, San Antonio Breast Cancer Symposium Meeting abstracts and clinicaltrials.gov were searched using the terms "Anti-HER2 treatment breast cancer and trastuzumab emtansine (T-DM1) "; papers considered relevant for the aim of this review were selected., Findings/results: The phase I trials have determined the safe dosing range of T-DM1, established at 3.6 mg/kg every 3 weeks. The phase III randomized EMILIA and TR3RESA trials have shown that T-DM1 provides objective tumor responses and significantly improves progression free survival and overall survival in HER2-positive metastatic breast cancer patients previously treated with anti-HER2-based regimens. The ongoing phase III trials KAITLIN and KATHERINE will give us further information about the place T-DM1 should occupy in the treatment of patients with early stage HER2-positive breast cancer. In this review we analyze the most relevant clinical trials conducted with T-DM1 and the role of this compound in the management of advanced breast cancer., Conclusion: T-DM1 has shown clinically relevant activity in the treatment of HER2-positive breast cancer patients after progression on trastuzumab and taxane based therapy, both in the second line treatment setting and after early relapse on adjuvant trastuzumab therapy. This is accompanied by a favorable safety and tolerability profile., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
138. Angiopoietin-2 is a negative prognostic marker in small cell lung cancer.
- Author
-
Cañadas I, Taus Á, Villanueva X, Arpí O, Pijuan L, Rodríguez Y, Menéndez S, Mojal S, Rojo F, Albanell J, Rovira A, and Arriola E
- Subjects
- Aged, Disease Progression, Female, Humans, Male, Middle Aged, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Prognosis, Vascular Endothelial Growth Factor A metabolism, Angiopoietin-2 metabolism, Lung Neoplasms metabolism, Lung Neoplasms pathology, Small Cell Lung Carcinoma metabolism, Small Cell Lung Carcinoma pathology
- Abstract
Background: Small cell lung cancer (SCLC) is a highly lethal disease due to its chemorefractory nature after initial treatment. Angiogenesis plays an important role in tumor growth, metastasis and chemoresistance. We hypothesized that angiogenesis could predict chemoresistance in SCLC patients and be potentially a therapeutic target in this disease., Methods: Serum samples from forty-three SCLC patients were prospectively obtained at diagnosis, response evaluation and progression. Angiogenesis-related cytokines (Angiopoietin-2, VEGF-A, C and D) were simultaneously quantified by Luminex Technology. Clinical data were prospectively recorder., Results: Significantly higher concentration of angiogenesis-related cytokines were found in SCLC patients at diagnosis compared to healthy volunteers. High baseline serum concentration of Angiopoietin-2 (sAngiopoietin-2) were associated with a worse overall survival (p=0.006) and remained independently associated with survival in the multivariate analysis (p=0.008). In addition, sAngiopoietin-2 significantly increased at progression when compared to baseline., Conclusion: These data provide novel evidence on a role of sAngiopoietin-2 in the adverse clinical behavior of SCLC and could be a potential therapeutic target in this disease., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
139. Enhanced MAF Oncogene Expression and Breast Cancer Bone Metastasis.
- Author
-
Pavlovic M, Arnal-Estapé A, Rojo F, Bellmunt A, Tarragona M, Guiu M, Planet E, Garcia-Albéniz X, Morales M, Urosevic J, Gawrzak S, Rovira A, Prat A, Nonell L, Lluch A, Jean-Mairet J, Coleman R, Albanell J, and Gomis RR
- Subjects
- Animals, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Heterografts, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Incidence, Mice, Mice, Inbred BALB C, Odds Ratio, Predictive Value of Tests, Prognosis, Proportional Hazards Models, Up-Regulation, Biomarkers, Tumor metabolism, Bone Neoplasms metabolism, Bone Neoplasms secondary, Breast Neoplasms metabolism, Breast Neoplasms pathology, DNA Copy Number Variations, Proto-Oncogene Proteins c-maf metabolism
- Abstract
Background: There are currently no biomarkers for early breast cancer patient populations at risk of bone metastasis. Identification of mediators of bone metastasis could be of clinical interest., Methods: A de novo unbiased screening approach based on selection of highly bone metastatic breast cancer cells in vivo was used to determine copy number aberrations (CNAs) associated with bone metastasis. The CNAs associated with bone metastasis were examined in independent primary breast cancer datasets with annotated clinical follow-up. The MAF gene encoded within the CNA associated with bone metastasis was subjected to gain and loss of function validation in breast cancer cells (MCF7, T47D, ZR-75, and 4T1), its downstream mechanism validated, and tested in clinical samples. A multivariable Cox cause-specific hazard model with competing events (death) was used to test the association between 16q23 or MAF and bone metastasis. All statistical tests were two-sided., Results: 16q23 gain CNA encoding the transcription factor MAF mediates breast cancer bone metastasis through the control of PTHrP. 16q23 gain (hazard ratio (HR) for bone metastasis = 14.5, 95% confidence interval (CI) = 6.4 to 32.9, P < .001) as well as MAF overexpression (HR for bone metastasis = 2.5, 95% CI = 1.7 to 3.8, P < .001) in primary breast tumors were specifically associated with risk of metastasis to bone but not to other organs., Conclusions: These results suggest that MAF is a mediator of breast cancer bone metastasis. 16q23 gain or MAF protein overexpression in tumors may help to select patients at risk of bone relapse., (© The Author 2015. Published by Oxford University Press.)
- Published
- 2015
- Full Text
- View/download PDF
140. Enhancing tumor-targeting monoclonal antibodies therapy by PARP inhibitors.
- Author
-
Yélamos J, Galindo M, Navarro J, Albanell J, Rovira A, Rojo F, and Oliver J
- Abstract
Monoclonal antibodies (mAbs) have become a successful therapeutic approach in cancer. However, some patients do not achieve long-term clinical benefit and most mAbs only exert modest effects as monotherapies. Therefore, combinations with chemotherapy are currently being investigated. Emerging studies have shown a synergistic therapeutic effect of PARP inhibitors and mAbs in cancer. PARP enzymes catalytically cleave β-NAD
+ and transfer the ADP-ribose moiety to acceptor proteins, modifying their function. In here, we update recent data about the therapeutic effect of the combination of PARP inhibitors with mAbs in cancer treatment and discuss the molecular mechanisms involved in this synergy.- Published
- 2015
- Full Text
- View/download PDF
141. MET expression and copy number heterogeneity in nonsquamous non-small cell lung cancer (nsNSCLC).
- Author
-
Casadevall D, Gimeno J, Clavé S, Taus Á, Pijuan L, Arumí M, Lorenzo M, Menéndez S, Cañadas I, Albanell J, Serrano S, Espinet B, Salido M, and Arriola E
- Subjects
- Adenocarcinoma mortality, Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung pathology, Female, Follow-Up Studies, Humans, Immunoenzyme Techniques, In Situ Hybridization, Fluorescence, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Grading, Neoplasm Staging, Prognosis, Survival Rate, Tissue Array Analysis, Adenocarcinoma genetics, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung genetics, DNA Copy Number Variations genetics, Lung Neoplasms genetics, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-met metabolism
- Abstract
Objective: We aimed to assess MET intratumoral heterogeneity and its potential impact on biomarker-based patient selection as well as potential surrogate biomarkers of MET activation., Methods: Our study included 120 patients with non-squamous Non-small-cell Lung Cancer (nsNSCLC), of which 47 were incorporated in tissue microarrays (TMA). Four morphologically distinct tumor areas were selected to assess MET heterogeneity. MET positivity by immunohistochemistry (IHC) was defined as an above-median H-score and by +2/+3 staining intensity in >50% of tumor cells (Metmab criteria). MET FISH positivity was defined by MET/CEP7 ratio ≥ 2.0 and/or MET ≥ 5.0. MET staining pattern (cytoplasmic vs. membranous) and mesenchymal markers were investigated as surrogates of MET activation., Results: Median MET H-score was 140 (range 0-400) and 47.8% of patients were MET positive by Metmab criteria. Eight cases (6.8%) were MET FISH positive and showed higher H-scores (p = 0.021). MET positivity by IHC changed in up to 40% of cases among different tumor areas, and MET amplification in 25-50%. Cytoplasmic MET staining and positivity for vimentin predicted poor survival (p = 0.042 and 0.047, respectively)., Conclusions: MET status is highly heterogeneous among different nsNSCLC tumor areas, hindering adequate patient selection for MET-targeted therapies. MET cytoplasmic staining and vimentin might represent surrogate markers for MET activation.
- Published
- 2015
- Full Text
- View/download PDF
142. Potential clinical relevant drug-drug interactions: comparison between different compendia, do we have a validated method?
- Author
-
Conde-Estévez D, Echeverría-Esnal D, Tusquets I, and Albanell J
- Subjects
- Female, Humans, Male, Antineoplastic Agents adverse effects, Medication Therapy Management, Neoplasms drug therapy, Pharmacy Service, Hospital, Polypharmacy
- Published
- 2015
- Full Text
- View/download PDF
143. Prospective study of the impact of the Prosigna assay on adjuvant clinical decision-making in unselected patients with estrogen receptor positive, human epidermal growth factor receptor negative, node negative early-stage breast cancer.
- Author
-
Martín M, González-Rivera M, Morales S, de la Haba-Rodriguez J, González-Cortijo L, Manso L, Albanell J, González-Martín A, González S, Arcusa A, de la Cruz-Merino L, Rojo F, Vidal M, Galván P, Aguirre E, Morales C, Ferree S, Pompilio K, Casas M, Caballero R, Goicoechea U, Carrasco E, Michalopoulos S, Hornberger J, and Prat A
- Subjects
- Breast Neoplasms pathology, Chemotherapy, Adjuvant methods, ErbB Receptors, Female, Humans, Middle Aged, Neoplasm Recurrence, Local, Prospective Studies, Spain, Breast Neoplasms drug therapy, Clinical Decision-Making, Receptors, Estrogen metabolism
- Abstract
Purpose: Improved understanding of risk of recurrence (ROR) is needed to reduce cases of recurrence and more effectively treat breast cancer patients. The purpose of this study was to examine how a gene-expression profile (GEP), identified by Prosigna, influences physician adjuvant treatment selection for early breast cancer (EBC) and the effects of this influence on optimizing adjuvant treatment recommendations in clinical practice., Methods: A prospective, observational, multicenter study was carried out in 15 hospitals across Spain. Participating medical oncologists completed pre-assessment, post-assessment, and follow-up questionnaires recording their treatment recommendations and confidence in these recommendations, before and after knowing the patient's ROR. Patients completed questionnaires on decision-making, anxiety, and health status., Results: Between June 2013 and January 2014, 217 patients enrolled and a final 200 were included in the study. Patients were postmenopausal, estrogen receptor positive, human epidermal growth hormone factor negative, and node negative with either stage 1 or stage 2 tumors. After receiving the GEP results, treatment recommendations were changed for 40 patients (20%). The confidence of medical oncologists in their treatment recommendations increased in 41.6% and decreased in 6.5% of total cases. Patients reported lower anxiety after physicians made treatment recommendations based on the GEP results (p < 0.05)., Conclusions: Though this study does not include evaluation of the impact of GEP on long-term outcomes, it was found that GEP results influenced the treatment decisions of medical oncologists and their confidence in adjuvant therapy selection. Patients' anxiety about the selected adjuvant therapy decreased with use of the GEP.
- Published
- 2015
- Full Text
- View/download PDF
144. Emergence of Multiple EGFR Extracellular Mutations during Cetuximab Treatment in Colorectal Cancer.
- Author
-
Arena S, Bellosillo B, Siravegna G, Martínez A, Cañadas I, Lazzari L, Ferruz N, Russo M, Misale S, González I, Iglesias M, Gavilan E, Corti G, Hobor S, Crisafulli G, Salido M, Sánchez J, Dalmases A, Bellmunt J, De Fabritiis G, Rovira A, Di Nicolantonio F, Albanell J, Bardelli A, and Montagut C
- Subjects
- Antineoplastic Agents therapeutic use, Blotting, Western, Cell Line, Tumor, Cetuximab therapeutic use, Colorectal Neoplasms drug therapy, DNA Mutational Analysis, Extracellular Space genetics, Flow Cytometry, Humans, Real-Time Polymerase Chain Reaction, Colorectal Neoplasms genetics, Drug Resistance, Neoplasm genetics, Genes, erbB-1 genetics, Mutation
- Abstract
Purpose: Patients with colorectal cancer who respond to the anti-EGFR antibody cetuximab often develop resistance within several months of initiating therapy. To design new lines of treatment, the molecular landscape of resistant tumors must be ascertained. We investigated the role of mutations in the EGFR signaling axis on the acquisition of resistance to cetuximab in patients and cellular models., Experimental Design: Tissue samples were obtained from 37 patients with colorectal cancer who became refractory to cetuximab. Colorectal cancer cells sensitive to cetuximab were treated until resistant derivatives emerged. Mutational profiling of biopsies and cell lines was performed. Structural modeling and functional analyses were performed to causally associate the alleles to resistance., Results: The genetic profile of tumor specimens obtained after cetuximab treatment revealed the emergence of a complex pattern of mutations in EGFR, KRAS, NRAS, BRAF, and PIK3CA genes, including two novel EGFR ectodomain mutations (R451C and K467T). Mutational profiling of cetuximab-resistant cells recapitulated the molecular landscape observed in clinical samples and revealed three additional EGFR alleles: S464L, G465R, and I491M. Structurally, these mutations are located in the cetuximab-binding region, except for the R451C mutant. Functionally, EGFR ectodomain mutations prevent binding to cetuximab but a subset is permissive for interaction with panitumumab., Conclusions: Colorectal tumors evade EGFR blockade by constitutive activation of downstream signaling effectors and through mutations affecting receptor-antibody binding. Both mechanisms of resistance may occur concomitantly. Our data have implications for designing additional lines of therapy for patients with colorectal cancer who relapse upon treatment with anti-EGFR antibodies., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
145. PP2A inhibition determines poor outcome and doxorubicin resistance in early breast cancer and its activation shows promising therapeutic effects.
- Author
-
Rincón R, Cristóbal I, Zazo S, Arpí O, Menéndez S, Manso R, Lluch A, Eroles P, Rovira A, Albanell J, García-Foncillas J, Madoz-Gúrpide J, and Rojo F
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antibiotics, Antineoplastic pharmacology, Blotting, Western, Breast Neoplasms drug therapy, Breast Neoplasms mortality, Doxorubicin pharmacology, Enzyme Activation physiology, Female, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Mice, Mice, Inbred BALB C, Middle Aged, Prognosis, Real-Time Polymerase Chain Reaction, Treatment Outcome, Biomarkers, Tumor analysis, Breast Neoplasms pathology, Drug Resistance, Neoplasm physiology, Protein Phosphatase 2 metabolism
- Abstract
The protein phosphatase 2A (PP2A) is a key tumor suppressor which has emerged as a novel molecular target in some human cancers. Here, we show that PP2A inhibition is a common event in breast cancer and identified PP2A phosphorylation and deregulation SET and CIP2A as molecular contributing mechanisms to inactivate PP2A. Interestingly, restoration of PP2A activity after FTY720 treatment reduced cell growth, induced apoptosis and decreased AKT and ERK activation. Moreover, FTY720 led to PP2A activation then enhancing doxorubicin-induced antitumor effects both in vitro and in vivo. PP2A inhibition (CPscore: PP2A phosphorylation and/or CIP2A overexpression) was detected in 27% of cases (62/230), and associated with grade (p = 0.017), relapse (p < 0.001), negative estrogen (p < 0.001) and progesterone receptor expression (p < 0.001), HER2-positive tumors (p = 0.049), Ki-67 expression (p < 0.001), and higher AKT (p < 0.001) and ERK (p < 0.001) phosphorylation. Moreover, PP2A inhibition determined shorter overall (p = 0.006) and event-free survival (p = 0.003), and multivariate analysis confirmed its independent prognostic impact. Altogether, our results indicate that PP2A is frequently inactivated in breast cancer and determines worse outcome, and its restoration using PP2A activators represents an alternative therapeutic strategy in this disease.
- Published
- 2015
- Full Text
- View/download PDF
146. Snail1-expressing fibroblasts in the tumor microenvironment display mechanical properties that support metastasis.
- Author
-
Stanisavljevic J, Loubat-Casanovas J, Herrera M, Luque T, Peña R, Lluch A, Albanell J, Bonilla F, Rovira A, Peña C, Navajas D, Rojo F, García de Herreros A, and Baulida J
- Subjects
- Animals, Humans, Mice, Mice, Knockout, Neoplasm Metastasis, Snail Family Transcription Factors, Tumor Microenvironment, rho GTP-Binding Proteins metabolism, rhoA GTP-Binding Protein metabolism, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Fibroblasts metabolism, Fibroblasts pathology, Transcription Factors biosynthesis
- Abstract
Crosstalk between tumor and stromal cells in the tumor microenvironment alter its properties in ways that facilitate the invasive behavior of tumor cells. Here, we demonstrate that cancer-associated fibroblasts (CAF) increase the stiffness of the extracellular matrix (ECM) and promote anisotropic fiber orientation, two mechanical signals generated through a Snail1/RhoA/αSMA-dependent mechanism that sustains oriented tumor cell migration and invasiveness. Snail1-depleted CAF failed to acquire myofibroblastic traits in response to TGFβ, including RhoA activation, αSMA-positive stress fibers, increased fibronectin fibrillogenesis, and production of a stiff ECM with oriented fibers. Snail1 expression in human tumor-derived CAF was associated with an ability to organize the ECM. In coculture, a relatively smaller number of Snail1-expressing CAF were capable of imposing an anisotropic ECM architecture, compared with nonactivated fibroblasts. Pathologically, human breast cancers with Snail1(+) CAF tended to exhibit desmoplastic areas with anisotropic fibers, lymph node involvement, and poorer outcomes. Snail1 involvement in driving an ordered ECM was further confirmed in wound-healing experiments in mice, with Snail1 depletion preventing the anisotropic organization of granulation tissue and delaying wound healing. Overall, our results showed that inhibiting Snail1 function in CAF could prevent tumor-driven ECM reorganization and cancer invasion., (©2014 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
147. Assessment of ALK status by FISH on 1000 Spanish non-small cell lung cancer patients.
- Author
-
Vidal J, Clavé S, de Muga S, González I, Pijuan L, Gimeno J, Remón J, Reguart N, Viñolas N, Gironés R, Bernet L, Majem M, Bosch-Barrera J, Porta R, Alonso N, Palmero R, Taus A, Albanell J, Espinet B, Salido M, and Arriola E
- Subjects
- Anaplastic Lymphoma Kinase, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Female, Gene Rearrangement, Humans, In Situ Hybridization, Fluorescence methods, Lung Neoplasms genetics, Lung Neoplasms pathology, Male, Middle Aged, Retrospective Studies, Carcinoma, Non-Small-Cell Lung enzymology, Lung Neoplasms enzymology, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Introduction: Patients with non-small cell lung cancer (NSCLC) harboring anaplastic lymphoma kinase (ALK) rearrangement selectively respond to ALK inhibitors. Thus, identification of ALK rearrangements has become a standard diagnostic test in advanced NSCLC patients. Our institution has been a referral center in Spain for ALK determination by Fluorescent in situ hybridization (FISH). The aim of our study was to assess the feasibility and the FISH patterns of the ALK gene and to evaluate the clinical and pathological features of patients with ALK alterations., Methods: Between 2010 and 2014, 1092 samples were evaluated for ALK using FISH technique (927 histological samples, 165 cytological samples). Correlation with available clinical-pathological information was assessed., Results: ALK rearrangement was found in 35 patients (3.2%). Cytological samples (using either direct smears or cell blocks), were more frequently non-assessable than histological samples (69% versus 89%, respectively) (p < 0.001). Within the ALK-rearranged cases the majority were female, non-smokers, and stage IV., Conclusions: Although assessable in cytological samples, biopsies are preferred when available for ALK evaluation by FISH. The ALK translocation prevalence and the associated clinico-pathological features in Spanish NSCLC patients are similar to those previously reported.
- Published
- 2014
- Full Text
- View/download PDF
148. Methylation status of IGFBP-3 as a useful clinical tool for deciding on a concomitant radiotherapy.
- Author
-
Pernía O, Belda-Iniesta C, Pulido V, Cortes-Sempere M, Rodriguez C, Vera O, Soto J, Jiménez J, Taus A, Rojo F, Arriola E, Rovira A, Albanell J, Macías MT, de Castro J, Perona R, and Ibañez de Caceres I
- Subjects
- Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung surgery, Cell Line, Tumor radiation effects, Cell Survival genetics, Cell Survival radiation effects, Combined Modality Therapy, DNA Methylation, Female, Humans, Insulin-Like Growth Factor Binding Protein 3 genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms mortality, Lung Neoplasms surgery, Male, Middle Aged, Phosphorylation drug effects, Prognosis, Proto-Oncogene Proteins c-akt metabolism, Survival Analysis, Treatment Outcome, Carcinoma, Non-Small-Cell Lung radiotherapy, Insulin-Like Growth Factor Binding Protein 3 metabolism, Lung Neoplasms radiotherapy, Promoter Regions, Genetic
- Abstract
The methylation status of the IGFBP-3 gene is strongly associated with cisplatin sensitivity in patients with non-small cell lung cancer (NSCLC). In this study, we found in vitro evidence that linked the presence of an unmethylated promoter with poor response to radiation. Our data also indicate that radiation might sensitize chemotherapy-resistant cells by reactivating IGFBP-3-expression through promoter demethylation, inactivating the PI3K/AKT pathway. We also explored the IGFBP-3 methylation effect on overall survival (OS) in a population of 40 NSCLC patients who received adjuvant therapy after R0 surgery. Our results indicate that patients harboring an unmethylated promoter could benefit more from a chemotherapy schedule alone than from a multimodality therapy involving radiotherapy and platinum-based treatments, increasing their OS by 2.5 y (p = .03). Our findings discard this epi-marker as a prognostic factor in a patient population without adjuvant therapy, indicating that radiotherapy does not improve survival for patients harboring an unmethylated IGFBP-3 promoter.
- Published
- 2014
- Full Text
- View/download PDF
149. Predicting response and survival in chemotherapy-treated triple-negative breast cancer.
- Author
-
Prat A, Lluch A, Albanell J, Barry WT, Fan C, Chacón JI, Parker JS, Calvo L, Plazaola A, Arcusa A, Seguí-Palmer MA, Burgues O, Ribelles N, Rodriguez-Lescure A, Guerrero A, Ruiz-Borrego M, Munarriz B, López JA, Adamo B, Cheang MC, Li Y, Hu Z, Gulley ML, Vidal MJ, Pitcher BN, Liu MC, Citron ML, Ellis MJ, Mardis E, Vickery T, Hudis CA, Winer EP, Carey LA, Caballero R, Carrasco E, Martín M, Perou CM, and Alba E
- Subjects
- Cohort Studies, Female, Humans, Middle Aged, Treatment Outcome, Triple Negative Breast Neoplasms physiopathology, Antineoplastic Agents therapeutic use, Survival Analysis, Triple Negative Breast Neoplasms drug therapy
- Abstract
Background: In this study, we evaluated the ability of gene expression profiles to predict chemotherapy response and survival in triple-negative breast cancer (TNBC)., Methods: Gene expression and clinical-pathological data were evaluated in five independent cohorts, including three randomised clinical trials for a total of 1055 patients with TNBC, basal-like disease (BLBC) or both. Previously defined intrinsic molecular subtype and a proliferation signature were determined and tested. Each signature was tested using multivariable logistic regression models (for pCR (pathological complete response)) and Cox models (for survival). Within TNBC, interactions between each signature and the basal-like subtype (vs other subtypes) for predicting either pCR or survival were investigated., Results: Within TNBC, all intrinsic subtypes were identified but BLBC predominated (55-81%). Significant associations between genomic signatures and response and survival after chemotherapy were only identified within BLBC and not within TNBC as a whole. In particular, high expression of a previously identified proliferation signature, or low expression of the luminal A signature, was found independently associated with pCR and improved survival following chemotherapy across different cohorts. Significant interaction tests were only obtained between each signature and the BLBC subtype for prediction of chemotherapy response or survival., Conclusions: The proliferation signature predicts response and improved survival after chemotherapy, but only within BLBC. This highlights the clinical implications of TNBC heterogeneity, and suggests that future clinical trials focused on this phenotypic subtype should consider stratifying patients as having BLBC or not.
- Published
- 2014
- Full Text
- View/download PDF
150. Subcutaneous trastuzumab: drug development and current position.
- Author
-
Martín Martorell P, Bermejo de Las Heras B, Pérez-Fidalgo JA, Huerta Alvaro M, Martín M, Albanell J, and Lluch Hernández A
- Subjects
- Breast Neoplasms metabolism, Drug Discovery, Female, Humans, Injections, Subcutaneous, Receptor, ErbB-2 metabolism, Trastuzumab, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Agents administration & dosage, Breast Neoplasms drug therapy
- Abstract
HER2-positive breast cancer, accounting for 15 % of the total breast cancer patient population, carries in itself a bad prognosis, which has now become much better after the advent of anti-HER2 drugs. HER2-targeted therapy has significantly improved disease free- and overall survival in HER2-positive breast cancer, and has rendered better disease control both in the early and advanced disease setting. Trastuzumab treatment duration is often prolonged and poses significant time and resource challenges both on the treatment institutions and on the patient. The recent development of a subcutaneous formulation has meant a significant advance in this respect. We review the drug development of the compound and the current evidence on its use.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.