126 results on '"Ander Urruticoechea"'
Search Results
2. A prospective study on tumour response assessment methods after neoadjuvant endocrine therapy in early oestrogen receptor-positive breast cancer
- Author
-
Joanna I. López-Velazco, Sara Manzano, María Otaño, Kepa Elorriaga, Núria Bultó, Julio Herrero, Ainhara Lahuerta, Virginia Segur, Isabel Álvarez-López, Maria M. Caffarel, and Ander Urruticoechea
- Subjects
Neoadjuvant endocrine therapy ,Aromatase inhibitors ,Tumour cellularity size ,Oestrogen receptor (ER)-positive breast cancer ,Pathological and radiological tumour response ,Preoperative endocrine prognostic index (PEPI) score ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Neoadjuvant endocrine therapy (NET) in oestrogen receptor-positive (ER+) /HER2-negative (HER2-) breast cancer allows real-time evaluation of drug efficacy as well as investigation of the biological and molecular changes that occur after estrogenic deprivation. Clinical and pathological evaluation after NET may be used to obtain prognostic and predictive information of tumour response to decide adjuvant treatment. In this setting, clinical scales developed to evaluate response after neoadjuvant chemotherapy are not useful and there are not validated biomarkers to assess response to NET beyond Ki67 levels and preoperative endocrine prognostic index score (mPEPI). Methods In this prospective study, we extensively analysed radiological (by ultrasound scan (USS) and magnetic resonance imaging (MRI)) and pathological tumour response of 104 postmenopausal patients with ER+ /HER2- resectable breast cancer, treated with NET for a mean of 7 months prior to surgery. We defined a new score, tumour cellularity size (TCS), calculated as the product of the residual tumour cellularity in the surgical specimen and the tumour pathological size. Results Our results show that radiological evaluation of response to NET by both USS and MRI underestimates pathological tumour size (path-TS). Tumour size [mean (range); mm] was: path-TS 20 (0–80); radiological-TS by USS 9 (0–31); by MRI: 12 (0–60). Nevertheless, they support the use of MRI over USS to clinically assess radiological tumour response (rad-TR) due to the statistically significant association of rad-TR by MRI, but not USS, with Ki67 decrease (p = 0.002 and p = 0.3, respectively) and mPEPI score (p = 0.002 and p = 0.6, respectively). In addition, we propose that TCS could become a new tool to standardize response assessment to NET given its simplicity, reproducibility and its good correlation with existing biomarkers (such as ΔKi67, p = 0.001) and potential added value. Conclusion Our findings shed light on the dynamics of tumour response to NET, challenge the paradigm of the ability of NET to decrease surgical volume and point to the utility of the TCS to quantify the scattered tumour response usually produced by endocrine therapy. In the future, these results should be validated in independent cohorts with associated survival data.
- Published
- 2024
- Full Text
- View/download PDF
3. Preventing alpelisib-related hyperglycaemia in HR+/HER2−/PIK3CA-mutated advanced breast cancer using metformin (METALLICA): a multicentre, open-label, single-arm, phase 2 trialResearch in context
- Author
-
Antonio Llombart-Cussac, José Manuel Pérez-Garcia, Manuel Ruiz Borrego, Pablo Tolosa, Salvador Blanch, Adela Fernández-Ortega, Ander Urruticoechea, Isabel Blancas, Cristina Saura, Beatriz Rojas, Begoña Bermejo, José Ponce Lorenzo, María Gion, Patricia Cortez-Castedo, Elisenda Llabres, Elena Galve, Juan Fernando Cueva, Ana López, José Luis Alonso-Romero, Santiago González-Santiago, Eduardo Martínez de Dueñas, Eva Ciruelos, Griselda Martrat, Petra Gener, Daniel Alcalá-López, Miguel Sampayo-Cordero, Fernando Gómez-Peralta, and Javier Cortés
- Subjects
Alpelisib ,Hyperglycaemia ,Prophylactic metformin ,HR+/HER2−/PIK3CA-mutated advanced breast cancer ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Hyperglycaemia is an early and frequent adverse event during alpelisib treatment. METALLICA aimed to evaluate prophylactic metformin to prevent or reduce hyperglycaemia occurrence in patients with HR+/HER2−/PIK3CA-mutated advanced breast cancer (ABC). Methods: Between August 13th, 2020 and March 23rd, 2022, this 2-cohort, phase 2, multicentre, single-arm trial (NCT04300790) enrolled patients with HR+/HER2−/PIK3CA-mutated ABC: cohort A, normal glycaemia (fasting plasma glucose
- Published
- 2024
- Full Text
- View/download PDF
4. Design of a multinational randomized controlled trial to assess the effects of structured and individualized exercise in patients with metastatic breast cancer on fatigue and quality of life: the EFFECT study
- Author
-
Anouk E. Hiensch, Evelyn M. Monninkhof, Martina E. Schmidt, Eva M. Zopf, Kate A. Bolam, Neil K. Aaronson, Jon Belloso, Wilhelm Bloch, Dorothea Clauss, Johanna Depenbusch, Milena Lachowicz, Mireia Pelaez, Helene Rundqvist, Elzbieta Senkus, Martijn M. Stuiver, Mark Trevaskis, Ander Urruticoechea, Friederike Rosenberger, Elsken van der Wall, G. Ardine de Wit, Philipp Zimmer, Yvonne Wengström, Karen Steindorf, and Anne M. May
- Subjects
Exercise ,Fatigue ,Metastatic breast cancer ,Quality of life ,Randomized controlled trial ,Medicine (General) ,R5-920 - Abstract
Abstract Background Many patients with metastatic breast cancer experience cancer- and treatment-related side effects that impair activities of daily living and negatively affect the quality of life. There is a need for interventions that improve quality of life by alleviating fatigue and other side effects during palliative cancer treatment. Beneficial effects of exercise have been observed in the curative setting, but, to date, comparable evidence in patients with metastatic breast cancer is lacking. The aim of this study is to assess the effects of a structured and individualized 9-month exercise intervention in patients with metastatic breast cancer on quality of life, fatigue, and other cancer- and treatment-related side effects. Methods The EFFECT study is a multinational, randomized controlled trial including 350 patients with metastatic breast cancer. Participants are randomly allocated (1:1) to an exercise or control group. The exercise group participates in a 9-month multimodal exercise program, starting with a 6-month period where participants exercise twice a week under the supervision of an exercise professional. After completing this 6-month period, one supervised session is replaced by one unsupervised session for 3 months. In addition, participants are instructed to be physically active for ≥30 min/day on all remaining days of the week, while being supported by an activity tracker and exercise app. Participants allocated to the control group receive standard medical care, general written physical activity advice, and an activity tracker, but no structured exercise program. The primary outcomes are quality of life (EORTC QLQ-C30, summary score) and fatigue (EORTC QLQ-FA12), assessed at baseline, 3, 6 (primary endpoint), and 9 months post-baseline. Secondary outcomes include physical fitness, physical performance, physical activity, anxiety, depression, pain, sleep problems, anthropometric data, body composition, and blood markers. Exploratory outcomes include quality of working life, muscle thickness, urinary incontinence, disease progression, and survival. Additionally, the cost-effectiveness of the exercise program is assessed. Adherence and safety are monitored throughout the intervention period. Discussion This large randomized controlled trial will provide evidence regarding the (cost-) effectiveness of exercise during treatment of metastatic breast cancer. If proven (cost-)effective, exercise should be offered to patients with metastatic breast cancer as part of standard care. Trial registration ClinicalTrials.gov NCT04120298 . Registered on October 9, 2019.
- Published
- 2022
- Full Text
- View/download PDF
5. Stromal oncostatin M cytokine promotes breast cancer progression by reprogramming the tumor microenvironment
- Author
-
Angela M. Araujo, Andrea Abaurrea, Peio Azcoaga, Joanna I. López-Velazco, Sara Manzano, Javier Rodriguez, Ricardo Rezola, Leire Egia-Mendikute, Fátima Valdés-Mora, Juana M. Flores, Liam Jenkins, Laura Pulido, Iñaki Osorio-Querejeta, Patricia Fernández-Nogueira, Nicola Ferrari, Cristina Viera, Natalia Martín-Martín, Alexandar Tzankov, Serenella Eppenberger-Castori, Isabel Alvarez-Lopez, Ander Urruticoechea, Paloma Bragado, Nicholas Coleman, Asís Palazón, Arkaitz Carracedo, David Gallego-Ortega, Fernando Calvo, Clare M. Isacke, María M. Caffarel, and Charles H. Lawrie
- Subjects
Medicine - Published
- 2022
- Full Text
- View/download PDF
6. Resistance to Taxanes in Triple-Negative Breast Cancer Associates with the Dynamics of a CD49f+ Tumor-Initiating Population
- Author
-
Jorge Gómez-Miragaya, Marta Palafox, Laia Paré, Guillermo Yoldi, Irene Ferrer, Sergi Vila, Patricia Galván, Pasquale Pellegrini, Hector Pérez-Montoyo, Ana Igea, Purificación Muñoz, Manel Esteller, Angel R. Nebreda, Ander Urruticoechea, Idoia Morilla, Sonia Pernas, Fina Climent, María Teresa Soler-Monso, Ana Petit, Violeta Serra, Aleix Prat, and Eva González-Suárez
- Subjects
TNBC ,triple-negative breast cancer ,PDX ,patient-derived orthoxenografts ,chemoresistance ,docetaxel ,CD49f tumor-initiating cells ,drug holidays ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Taxanes are a mainstay of treatment for breast cancer, but resistance often develops followed by metastatic disease and mortality. Aiming to reveal the mechanisms underlying taxane resistance, we used breast cancer patient-derived orthoxenografts (PDX). Mimicking clinical behavior, triple-negative breast tumors (TNBCs) from PDX models were more sensitive to docetaxel than luminal tumors, but they progressively acquired resistance upon continuous drug administration. Mechanistically, we found that a CD49f+ chemoresistant population with tumor-initiating ability is present in sensitive tumors and expands during the acquisition of drug resistance. In the absence of the drug, the resistant CD49f+ population shrinks and taxane sensitivity is restored. We describe a transcriptional signature of resistance, predictive of recurrent disease after chemotherapy in TNBC. Together, these findings identify a CD49f+ population enriched in tumor-initiating ability and chemoresistance properties and evidence a drug holiday effect on the acquired resistance to docetaxel in triple-negative breast cancer.
- Published
- 2017
- Full Text
- View/download PDF
7. A Pathology-Based Combined Model to Identify PAM50 Non-luminal Intrinsic Disease in Hormone Receptor-Positive HER2-Negative Breast Cancer
- Author
-
Tomás Pascual, Miguel Martin, Aranzazu Fernández-Martínez, Laia Paré, Emilio Alba, Álvaro Rodríguez-Lescure, Giuseppe Perrone, Javier Cortés, Serafín Morales, Ana Lluch, Ander Urruticoechea, Blanca González-Farré, Patricia Galván, Pedro Jares, Adela Rodriguez, Nuria Chic, Daniela Righi, Juan Miguel Cejalvo, Giuseppe Tonini, Barbara Adamo, Maria Vidal, Patricia Villagrasa, Montserrat Muñoz, and Aleix Prat
- Subjects
intrinsic subtype ,non-luminal ,PAM50 ,breast cancer ,gene expression ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: In hormone receptor-positive (HR+)/HER2-negative breast cancer, the HER2-enriched and Basal-like intrinsic subtypes are associated with poor outcome, low response to anti-estrogen therapy and high response to chemotherapy. To date, no validated biomarker exists to identify both molecular entities other than gene expression.Methods: PAM50 subtyping and immunohistochemical data were obtained from 8 independent studies of 1,416 HR+/HER2-negative early breast tumors. A non-luminal disease score (NOLUS) from 0 to 100, based on percentage of estrogen receptor (ER), progesterone receptor (PR) and Ki67 tumor cells, was derived in a combined cohort of 5 studies (training dataset) and tested in a combined cohort of 3 studies. The performance of NOLUS was estimated using Area Under the ROC Curve (AUC).Results: In the training dataset (n = 903) and compared to luminal disease, non-luminal disease had lower percentage of ER-positive cells (median 65.2 vs. 86.2%, p < 0.01) and PR-positive cells (33.2 vs. 56.4%, p < 0.01) and higher percentage of Ki67-positive cells (18.2 vs. 13.1%, p = 0.01). A NOLUS formula was derived: −0.45*ER −0.28*PR +0.27*Ki67 + 73.02. The proportion of non-luminal tumors in NOLUS-positive (≥51.38) and NOLUS-negative (
- Published
- 2019
- Full Text
- View/download PDF
8. Altered Levels of Desaturation and ω-6 Fatty Acids in Breast Cancer Patients’ Red Blood Cell Membranes
- Author
-
Javier Amézaga, Gurutze Ugartemendia, Aitziber Larraioz, Nerea Bretaña, Aizpea Iruretagoyena, Joana Camba, Ander Urruticoechea, Carla Ferreri, and Itziar Tueros
- Subjects
arachidonic acid ,breast cancer ,linoleic acid ,membrane lipidome ,omega 6 ,red blood cell ,Microbiology ,QR1-502 - Abstract
Red blood cell (RBC) membrane can reflect fatty acid (FA) contribution from diet and biosynthesis. In cancer, membrane FAs are involved in tumorigenesis and invasiveness, and are indicated as biomarkers to monitor the disease evolution as well as potential targets for therapies and nutritional strategies. The present study provides RBC membrane FA profiles in recently diagnosed breast cancer patients before starting chemotherapy treatment. Patients and controls were recruited, and their dietary habits were collected. FA lipidomic analysis of mature erythrocyte membrane phospholipids in blood samples was performed. Data were adjusted to correct for the effects of diet, body mass index (BMI), and age, revealing that patients showed lower levels of saturated fatty acids (SFA) and higher levels of monounsaturated fatty acid, cis-vaccenic (25%) than controls, with consequent differences in desaturase enzymatic index (∆9 desaturase, –13.1%). In the case of polyunsaturated fatty acids (PUFA), patients had higher values of ω-6 FA (C18:2 (+11.1%); C20:4 (+7.4%)). RBC membrane lipidomic analysis in breast cancer revealed that ω-6 pathways are favored. These results suggest new potential targets for treatments and better nutritional guidelines.
- Published
- 2020
- Full Text
- View/download PDF
9. Spatial intratumoural heterogeneity in the expression of GIT1 is associated with poor prognostic outcome in oestrogen receptor positive breast cancer patients with synchronous lymph node metastases [version 2; referees: 2 approved]
- Author
-
Ibai Goicoechea, Ricardo Rezola, María Arestin, María M. Caffarel, Ana Rosa Cortazar, Lorea Manterola, Marta Fernandez-Mercado, María Armesto, Carla Sole, Erika Larrea, Angela M. Araujo, Nerea Ancizar, Arrate Plazaola, Ander Urruticoechea, Arkaitz Carracedo, Irune Ruiz, Isabel Alvarez Lopez, and Charles H. Lawrie
- Subjects
Breast Diseases: Benign & Malignant ,Medicine ,Science - Abstract
Background: The outcome for oestrogen receptor positive (ER+) breast cancer patients has improved greatly in recent years largely due to targeted therapy. However, the presence of involved multiple synchronous lymph nodes remains associated with a poor outcome. Consequently, these patients would benefit from the identification of new prognostic biomarkers and therapeutic targets. The expression of G-protein-coupled receptor kinase-interacting protein 1 (GIT1) has recently been shown to be an indicator of advanced stage breast cancer. Therefore, we investigated its expression and prognostic value of GIT1 in a cohort of 140 ER+ breast cancer with synchronous lymph node involvement. Methods: Immunohistochemistry was employed to assess GIT1 expression in a tissue microarray (TMA) containing duplicate non-adjacent cores with matched primary tumour and lymph node tissue (n=140). GIT1 expression in tumour cells was scored and statistical correlation analyses were carried out. Results: The results revealed a sub-group of patients that displayed discordant expression of GIT1 between the primary tumour and the lymph nodes (i.e. spatial intratumoural heterogeneity). We observed that loss of GIT1 expression in the tumour cells of the metastasis was associated with a shorter time to recurrence, poorer overall survival, and a shorter median survival time. Moreover, multivariate analysis demonstrated that GIT1 expression was an independent prognostic indicator. Conclusions: GIT1 expression enabled the identification of a sub-class of ER+ patients with lymph node metastasis that have a particularly poor prognostic outcome. We propose that this biomarker could be used to further stratify ER+ breast cancer patients with synchronous lymph node involvement and therefore facilitate adjuvant therapy decision making.
- Published
- 2018
- Full Text
- View/download PDF
10. Predictive and Prognostic Brain Metastases Assessment in Luminal Breast Cancer Patients: FN14 and GRP94 from Diagnosis to Prophylaxis
- Author
-
Antonio Martínez-Aranda, Vanessa Hernández, Ferran Moreno, Núria Baixeras, Daniel Cuadras, Ander Urruticoechea, Miguel Gil-Gil, Noemí Vidal, Xavier Andreu, Miquel A. Seguí, Rosa Ballester, Eva Castella, and Angels Sierra
- Subjects
biomarkers ,brain metastasis ,breast cancer ,FN14 ,GRP94 ,prediction ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
FN14 has been implicated in many intracellular signaling pathways, and GRP94 is a well-known endoplasmic reticulum protein regulated by glucose. Recently, both have been associated with metastasis progression in breast cancer patients. We studied the usefulness of FN14 and GRP94 expression to stratify breast cancer patients according their risk of brain metastasis (BrM) progression. We analyzed FN14 and GRP94 by immunohistochemistry in a retrospective multicenter study using tissue microarrays from 208 patients with breast carcinomas, of whom 52 had developed BrM. Clinical and pathological characteristics and biomarkers expression in Luminal and non-Luminal patients were analyzed using a multivariate logistic regression model adjusted for covariates, and brain metastasis-free survival (BrMFS) was estimated using the Kaplan–Meier method and the Cox proportional hazards model. FN14 expression was associated with BrM progression mainly in Luminal breast cancer patients with a sensitivity (53.85%) and specificity (89.60%) similar to Her2 expression (46.15 and 89.84%, respectively). Moreover, the likelihood to develop BrM in FN14-positive Luminal carcinomas increased 36.70-fold (3.65–368.25, p = 0.002). Furthermore, the worst prognostic factor for BrMFS in patients with Luminal carcinomas was FN14 overexpression (HR = 8.25; 95% CI: 2.77–24.61; p = 0.00015). In these patients, GRP94 overexpression also increased the risk of BrM (HR = 3.58; 95% CI: 0.98–13.11; p = 0.054—Wald test). Therefore, FN14 expression in Luminal breast carcinomas is a predictive/prognostic biomarker of BrM, which combined with GRP94 predicts BrM progression in non-Luminal tumors 4.04-fold (1.19–8.22, p = 0.025), suggesting that both biomarkers are useful to stratify BrM risk at early diagnosis. We propose a new follow-up protocol for the early prevention of clinical BrM of breast cancer patients with BrM risk.
- Published
- 2017
- Full Text
- View/download PDF
11. Interplay between BRCA1 and RHAMM regulates epithelial apicobasal polarization and may influence risk of breast cancer.
- Author
-
Christopher A Maxwell, Javier Benítez, Laia Gómez-Baldó, Ana Osorio, Núria Bonifaci, Ricardo Fernández-Ramires, Sylvain V Costes, Elisabet Guinó, Helen Chen, Gareth J R Evans, Pooja Mohan, Isabel Català, Anna Petit, Helena Aguilar, Alberto Villanueva, Alvaro Aytes, Jordi Serra-Musach, Gad Rennert, Flavio Lejbkowicz, Paolo Peterlongo, Siranoush Manoukian, Bernard Peissel, Carla B Ripamonti, Bernardo Bonanni, Alessandra Viel, Anna Allavena, Loris Bernard, Paolo Radice, Eitan Friedman, Bella Kaufman, Yael Laitman, Maya Dubrovsky, Roni Milgrom, Anna Jakubowska, Cezary Cybulski, Bohdan Gorski, Katarzyna Jaworska, Katarzyna Durda, Grzegorz Sukiennicki, Jan Lubiński, Yin Yao Shugart, Susan M Domchek, Richard Letrero, Barbara L Weber, Frans B L Hogervorst, Matti A Rookus, J Margriet Collee, Peter Devilee, Marjolijn J Ligtenberg, Rob B van der Luijt, Cora M Aalfs, Quinten Waisfisz, Juul Wijnen, Cornelis E P van Roozendaal, HEBON, EMBRACE, Douglas F Easton, Susan Peock, Margaret Cook, Clare Oliver, Debra Frost, Patricia Harrington, D Gareth Evans, Fiona Lalloo, Rosalind Eeles, Louise Izatt, Carol Chu, Diana Eccles, Fiona Douglas, Carole Brewer, Heli Nevanlinna, Tuomas Heikkinen, Fergus J Couch, Noralane M Lindor, Xianshu Wang, Andrew K Godwin, Maria A Caligo, Grazia Lombardi, Niklas Loman, Per Karlsson, Hans Ehrencrona, Anna von Wachenfeldt, SWE-BRCA, Rosa Bjork Barkardottir, Ute Hamann, Muhammad U Rashid, Adriana Lasa, Trinidad Caldés, Raquel Andrés, Michael Schmitt, Volker Assmann, Kristen Stevens, Kenneth Offit, João Curado, Hagen Tilgner, Roderic Guigó, Gemma Aiza, Joan Brunet, Joan Castellsagué, Griselda Martrat, Ander Urruticoechea, Ignacio Blanco, Laima Tihomirova, David E Goldgar, Saundra Buys, Esther M John, Alexander Miron, Melissa Southey, Mary B Daly, BCFR, Rita K Schmutzler, Barbara Wappenschmidt, Alfons Meindl, Norbert Arnold, Helmut Deissler, Raymonda Varon-Mateeva, Christian Sutter, Dieter Niederacher, Evgeny Imyamitov, Olga M Sinilnikova, Dominique Stoppa-Lyonne, Sylvie Mazoyer, Carole Verny-Pierre, Laurent Castera, Antoine de Pauw, Yves-Jean Bignon, Nancy Uhrhammer, Jean-Philippe Peyrat, Philippe Vennin, Sandra Fert Ferrer, Marie-Agnès Collonge-Rame, Isabelle Mortemousque, GEMO Study Collaborators, Amanda B Spurdle, Jonathan Beesley, Xiaoqing Chen, Sue Healey, kConFab, Mary Helen Barcellos-Hoff, Marc Vidal, Stephen B Gruber, Conxi Lázaro, Gabriel Capellá, Lesley McGuffog, Katherine L Nathanson, Antonis C Antoniou, Georgia Chenevix-Trench, Markus C Fleisch, Víctor Moreno, and Miguel Angel Pujana
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Differentiated mammary epithelium shows apicobasal polarity, and loss of tissue organization is an early hallmark of breast carcinogenesis. In BRCA1 mutation carriers, accumulation of stem and progenitor cells in normal breast tissue and increased risk of developing tumors of basal-like type suggest that BRCA1 regulates stem/progenitor cell proliferation and differentiation. However, the function of BRCA1 in this process and its link to carcinogenesis remain unknown. Here we depict a molecular mechanism involving BRCA1 and RHAMM that regulates apicobasal polarity and, when perturbed, may increase risk of breast cancer. Starting from complementary genetic analyses across families and populations, we identified common genetic variation at the low-penetrance susceptibility HMMR locus (encoding for RHAMM) that modifies breast cancer risk among BRCA1, but probably not BRCA2, mutation carriers: n = 7,584, weighted hazard ratio ((w)HR) = 1.09 (95% CI 1.02-1.16), p(trend) = 0.017; and n = 3,965, (w)HR = 1.04 (95% CI 0.94-1.16), p(trend) = 0.43; respectively. Subsequently, studies of MCF10A apicobasal polarization revealed a central role for BRCA1 and RHAMM, together with AURKA and TPX2, in essential reorganization of microtubules. Mechanistically, reorganization is facilitated by BRCA1 and impaired by AURKA, which is regulated by negative feedback involving RHAMM and TPX2. Taken together, our data provide fundamental insight into apicobasal polarization through BRCA1 function, which may explain the expanded cell subsets and characteristic tumor type accompanying BRCA1 mutation, while also linking this process to sporadic breast cancer through perturbation of HMMR/RHAMM.
- Published
- 2011
- Full Text
- View/download PDF
12. Biological convergence of cancer signatures.
- Author
-
Xavier Solé, Núria Bonifaci, Núria López-Bigas, Antoni Berenguer, Pilar Hernández, Oscar Reina, Christopher A Maxwell, Helena Aguilar, Ander Urruticoechea, Silvia de Sanjosé, Francesc Comellas, Gabriel Capellá, Víctor Moreno, and Miguel Angel Pujana
- Subjects
Medicine ,Science - Abstract
Gene expression profiling has identified cancer prognostic and predictive signatures with superior performance to conventional histopathological or clinical parameters. Consequently, signatures are being incorporated into clinical practice and will soon influence everyday decisions in oncology. However, the slight overlap in the gene identity between signatures for the same cancer type or condition raises questions about their biological and clinical implications. To clarify these issues, better understanding of the molecular properties and possible interactions underlying apparently dissimilar signatures is needed. Here, we evaluated whether the signatures of 24 independent studies are related at the genome, transcriptome or proteome levels. Significant associations were consistently observed across these molecular layers, which suggest the existence of a common cancer cell phenotype. Convergence on cell proliferation and death supports the pivotal involvement of these processes in prognosis, metastasis and treatment response. In addition, functional and molecular associations were identified with the immune response in different cancer types and conditions that complement the contribution of cell proliferation and death. Examination of additional, independent, cancer datasets corroborated our observations. This study proposes a comprehensive strategy for interpreting cancer signatures that reveals common design principles and systems-level properties.
- Published
- 2009
- Full Text
- View/download PDF
13. Preliminary Qualitative and Quantitative Evaluation of DESIREE, a Decision Support Platform for the Management of Primary Breast Cancer Patients.
- Author
-
Sylvia Pelayo, Jacques Bouaud, Claudia Blancafort, Jean-Baptiste Lamy, Booma Devi Sekar, Nekane Larburu, Naiara Muro, Ander Urruticoechea, Jon Belloso, Guillermo Valderas, Sara Guardiola, Charlotte Ngo, Luis Teixeira, Gilles Guézennec, and Brigitte Séroussi
- Published
- 2020
14. Early Detection of Brain Metastases in a Supervised Exercise Program for Patients with Advanced Breast Cancer: A Case Report
- Author
-
Mireia Pelaez, Martijn M. Stuiver, Marike Broekman, Kathryn H. Schmitz, Eva M. Zopf, Dorothea Clauss, Yvonne Wengström, Friederike Rosenberger, Karen Steindorf, Ander Urruticoechea, and Anne M. May
- Subjects
Physical Therapy, Sports Therapy and Rehabilitation ,Orthopedics and Sports Medicine - Published
- 2023
15. Supplementary Table 4 from Phase Ib Study of Buparlisib plus Trastuzumab in Patients with HER2-Positive Advanced or Metastatic Breast Cancer That Has Progressed on Trastuzumab-Based Therapy
- Author
-
Jose Baselga, Luc Dirix, Samit Hirawat, Cristian Massacesi, David W. Sternberg, Emmanuelle Di Tomaso, Joseph T. Beck, Ander Urruticoechea, Ana Bosch, Sophie Ruquet, David Mills, Stefan De Buck, Qinhua Ru, Shaun Su, Guy Jerusalem, Johanna Bendell, and Cristina Saura
- Abstract
PDF file - 45K, Summary of adverse events suspected to be study-drug related by grade (safety set).
- Published
- 2023
16. Supplementary Figure 1 from Phase Ib Study of Buparlisib plus Trastuzumab in Patients with HER2-Positive Advanced or Metastatic Breast Cancer That Has Progressed on Trastuzumab-Based Therapy
- Author
-
Jose Baselga, Luc Dirix, Samit Hirawat, Cristian Massacesi, David W. Sternberg, Emmanuelle Di Tomaso, Joseph T. Beck, Ander Urruticoechea, Ana Bosch, Sophie Ruquet, David Mills, Stefan De Buck, Qinhua Ru, Shaun Su, Guy Jerusalem, Johanna Bendell, and Cristina Saura
- Abstract
PDF file - 67K, Bayesian logistic regression model: dose-response curve and model inference results at the time of determination of the recommended Phase II dose.
- Published
- 2023
17. Supplementary Data from Novel Inhibitors of Fatty Acid Synthase with Anticancer Activity
- Author
-
Ramon Colomer, María Luz López-Rodríguez, Diego Haro, Ander Urruticoechea, Pedro F. Marrero, Gemma Casals, Silvia Ortega-Gutiérrez, Joana Relat, Helena Aguilar, Bellinda Benhamú, Carlos Turrado, and Teresa Puig
- Abstract
Supplementary Data from Novel Inhibitors of Fatty Acid Synthase with Anticancer Activity
- Published
- 2023
18. Supplementary Table 1 from Phase Ib Study of Buparlisib plus Trastuzumab in Patients with HER2-Positive Advanced or Metastatic Breast Cancer That Has Progressed on Trastuzumab-Based Therapy
- Author
-
Jose Baselga, Luc Dirix, Samit Hirawat, Cristian Massacesi, David W. Sternberg, Emmanuelle Di Tomaso, Joseph T. Beck, Ander Urruticoechea, Ana Bosch, Sophie Ruquet, David Mills, Stefan De Buck, Qinhua Ru, Shaun Su, Guy Jerusalem, Johanna Bendell, and Cristina Saura
- Abstract
PDF file - 65K, Criteria for defining dose-limiting toxicities.
- Published
- 2023
19. Data from Phase Ib Study of Buparlisib plus Trastuzumab in Patients with HER2-Positive Advanced or Metastatic Breast Cancer That Has Progressed on Trastuzumab-Based Therapy
- Author
-
Jose Baselga, Luc Dirix, Samit Hirawat, Cristian Massacesi, David W. Sternberg, Emmanuelle Di Tomaso, Joseph T. Beck, Ander Urruticoechea, Ana Bosch, Sophie Ruquet, David Mills, Stefan De Buck, Qinhua Ru, Shaun Su, Guy Jerusalem, Johanna Bendell, and Cristina Saura
- Abstract
Purpose: Phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway activation in patients with HER2-positive (HER2+) breast cancer has been implicated in de novo and acquired trastuzumab resistance. The purpose of this study was to determine the clinical activity of the PI3K inhibitor buparlisib (BKM120) in patients with HER2+ advanced/metastatic breast cancer resistant to trastuzumab-based therapy.Experimental Design: In the dose-escalation portion of this phase I/II study, patients with trastuzumab-resistant locally advanced or metastatic HER2+ breast cancer were treated with daily oral doses of buparlisib and weekly intravenous trastuzumab (2 mg/kg). Dose escalation was guided by a Bayesian logistic regression model with overdose control.Results: Of 18 enrolled patients, 17 received buparlisib. One dose-limiting toxicity of grade 3 general weakness was reported at the 100-mg/day dose level (the single-agent maximum tolerated dose) and this dose level was declared the recommended phase II dose (RP2D) of buparlisib in combination with trastuzumab. Common (>25%) adverse events included rash (39%), hyperglycemia (33%), and diarrhea (28%). The pharmacokinetic profile of buparlisib was not affected by its combination with trastuzumab. At the RP2D, there were two (17%) partial responses, 7 (58%) patients had stable disease (≥6 weeks), and the disease control rate was 75%. Pharmacodynamic studies showed inhibition of the PI3K/AKT/mTOR and RAS/MEK/ERK pathways.Conclusions: In this patient population, the combination of buparlisib and trastuzumab was well tolerated, and preliminary signs of clinical activity were observed. The phase II portion of this study will further explore the safety and efficacy of this combination at the RP2D. Clin Cancer Res; 20(7); 1935–45. ©2014 AACR.
- Published
- 2023
20. Data from Novel Inhibitors of Fatty Acid Synthase with Anticancer Activity
- Author
-
Ramon Colomer, María Luz López-Rodríguez, Diego Haro, Ander Urruticoechea, Pedro F. Marrero, Gemma Casals, Silvia Ortega-Gutiérrez, Joana Relat, Helena Aguilar, Bellinda Benhamú, Carlos Turrado, and Teresa Puig
- Abstract
Purpose: Fatty acid synthase (FASN) is overexpressed in human breast carcinoma. The natural polyphenol (−)-epigallocatechin-3-gallate blocks in vitro FASN activity and leads to apoptosis in breast cancer cells without any effects on carnitine palmitoyltransferase-1 (CPT-1) activity, and in vivo, does not decrease body weight. We synthesized a panel of new polyphenolic compounds and tested their effects on breast cancer models.Experimental Design: We evaluated the in vitro effects of the compounds on breast cancer cell growth (SK-Br3, MCF-7, and MDA-MB-231), apoptosis [as assessed by cleavage of poly(ADP-ribose) polymerase], cell signaling (HER2, ERK1/2, and AKT), and fatty acid metabolism enzymes (FASN and CPT-1). In vivo, we have evaluated their antitumor activity and their effect on body weight in a mice model of BT474 breast cancer cells.Results: Two compounds potently inhibited FASN activity and showed high cytotoxicity. Moreover, the compounds induced apoptosis and caused a marked decrease in the active forms of HER2, AKT, and ERK1/2 proteins. Interestingly, the compounds did not stimulate CPT-1 activity in vitro. We show evidence that one of the FASN inhibitors blocked the growth of BT474 breast cancer xenografts and did not induce weight loss in vivo.Conclusions: The synthesized polyphenolic compounds represent a novel class of FASN inhibitors, with in vitro and in vivo anticancer activity, that do not exhibit cross-activation of β-oxidation and do not induce weight loss in animals. One of the compounds blocked the growth of breast cancer xenografts. These FASN inhibitors may represent new agents for breast cancer treatment. (Clin Cancer Res 2009;15(24):7608–15)
- Published
- 2023
21. Supplementary Table 2 from Phase Ib Study of Buparlisib plus Trastuzumab in Patients with HER2-Positive Advanced or Metastatic Breast Cancer That Has Progressed on Trastuzumab-Based Therapy
- Author
-
Jose Baselga, Luc Dirix, Samit Hirawat, Cristian Massacesi, David W. Sternberg, Emmanuelle Di Tomaso, Joseph T. Beck, Ander Urruticoechea, Ana Bosch, Sophie Ruquet, David Mills, Stefan De Buck, Qinhua Ru, Shaun Su, Guy Jerusalem, Johanna Bendell, and Cristina Saura
- Abstract
PDF file - 38K, Patient disposition (full analysis set).
- Published
- 2023
22. Supplementary Table 3 from Phase Ib Study of Buparlisib plus Trastuzumab in Patients with HER2-Positive Advanced or Metastatic Breast Cancer That Has Progressed on Trastuzumab-Based Therapy
- Author
-
Jose Baselga, Luc Dirix, Samit Hirawat, Cristian Massacesi, David W. Sternberg, Emmanuelle Di Tomaso, Joseph T. Beck, Ander Urruticoechea, Ana Bosch, Sophie Ruquet, David Mills, Stefan De Buck, Qinhua Ru, Shaun Su, Guy Jerusalem, Johanna Bendell, and Cristina Saura
- Abstract
PDF file - 48K, Exposure to buparlisib and trastuzumab (safety set).
- Published
- 2023
23. Supplementary Table 6 from Phase Ib Study of Buparlisib plus Trastuzumab in Patients with HER2-Positive Advanced or Metastatic Breast Cancer That Has Progressed on Trastuzumab-Based Therapy
- Author
-
Jose Baselga, Luc Dirix, Samit Hirawat, Cristian Massacesi, David W. Sternberg, Emmanuelle Di Tomaso, Joseph T. Beck, Ander Urruticoechea, Ana Bosch, Sophie Ruquet, David Mills, Stefan De Buck, Qinhua Ru, Shaun Su, Guy Jerusalem, Johanna Bendell, and Cristina Saura
- Abstract
PDF file - 62K, Summary of buparlisib pharmacokinetic parameters.
- Published
- 2023
24. Supplementary Figure 3 from RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis
- Author
-
Eva González-Suárez, William C. Dougall, Dan Branstetter, Mark Tometsko, Francesc Viñals, Purificación Muñoz, Maria Teresa Soler, Fina Climent, Ander Urruticoechea, Sara Hernandez-Ortega, Sergi Vila, Pasquale Pellegrini, Irene Ferrer, and Marta Palafox
- Abstract
PDF file - 72K, RANK overexpression induces mammary stem markers
- Published
- 2023
25. Supplementary Figure 6 from RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis
- Author
-
Eva González-Suárez, William C. Dougall, Dan Branstetter, Mark Tometsko, Francesc Viñals, Purificación Muñoz, Maria Teresa Soler, Fina Climent, Ander Urruticoechea, Sara Hernandez-Ortega, Sergi Vila, Pasquale Pellegrini, Irene Ferrer, and Marta Palafox
- Abstract
PDF file - 172K, RANK overexpression promotes growth, invasive changes and metastasis in cells with non-functional BRCA1
- Published
- 2023
26. Supplementary Figure 4 from RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis
- Author
-
Eva González-Suárez, William C. Dougall, Dan Branstetter, Mark Tometsko, Francesc Viñals, Purificación Muñoz, Maria Teresa Soler, Fina Climent, Ander Urruticoechea, Sara Hernandez-Ortega, Sergi Vila, Pasquale Pellegrini, Irene Ferrer, and Marta Palafox
- Abstract
PDF file - 114K, FL-RANK cells grow in the absence of EGF and do not form tumors in vivo.
- Published
- 2023
27. Supplementary Figure 1 from RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis
- Author
-
Eva González-Suárez, William C. Dougall, Dan Branstetter, Mark Tometsko, Francesc Viñals, Purificación Muñoz, Maria Teresa Soler, Fina Climent, Ander Urruticoechea, Sara Hernandez-Ortega, Sergi Vila, Pasquale Pellegrini, Irene Ferrer, and Marta Palafox
- Abstract
PDF file - 91K, RANK is expressed and active in MCF10A cells
- Published
- 2023
28. Supplementary Figure 7 from RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis
- Author
-
Eva González-Suárez, William C. Dougall, Dan Branstetter, Mark Tometsko, Francesc Viñals, Purificación Muñoz, Maria Teresa Soler, Fina Climent, Ander Urruticoechea, Sara Hernandez-Ortega, Sergi Vila, Pasquale Pellegrini, Irene Ferrer, and Marta Palafox
- Abstract
PDF file - 89K, RANK/RANKL expression significantly associates with tumor subtype and lymph node metastasis
- Published
- 2023
29. Supplementary Figure 2 from RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis
- Author
-
Eva González-Suárez, William C. Dougall, Dan Branstetter, Mark Tometsko, Francesc Viñals, Purificación Muñoz, Maria Teresa Soler, Fina Climent, Ander Urruticoechea, Sara Hernandez-Ortega, Sergi Vila, Pasquale Pellegrini, Irene Ferrer, and Marta Palafox
- Abstract
PDF file - 113K, RANK expression induces EMT in MCF10A and in HMECS immortalized with telomerase
- Published
- 2023
30. Supplementary Methods, Figure Legends 1-7 from RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis
- Author
-
Eva González-Suárez, William C. Dougall, Dan Branstetter, Mark Tometsko, Francesc Viñals, Purificación Muñoz, Maria Teresa Soler, Fina Climent, Ander Urruticoechea, Sara Hernandez-Ortega, Sergi Vila, Pasquale Pellegrini, Irene Ferrer, and Marta Palafox
- Abstract
PDF file - 167K
- Published
- 2023
31. Abstract P5-16-12: Neoadjuvant letrozole plus palbociclib in patients (pts) with hormone receptor (HR)-positive/HER2-negative early breast cancer (EBC) with baseline Ki67 ≥20% and an Oncotype DX Breast Recurrence Score® result (RS) ≥18: DxCARTES
- Author
-
Antonio Llombart-Cussac, Ángel Guerrero-Zotano, Manuel Ruiz, Begoña Bermejo, Miguel Gil-Gil, Juan de la Haba, Emilio Alba, Vanesa Quiroga, Vicente Carañana, Ander Urruticoechea, Serafín Morales, Meritxell Bellet, Antonio Antón, José Manuel Pérez-García, María Fernández-Abad, Sonia Servitja, Pedro Sánchez-Rovira, Sofia Braga, Miguel Sampayo-Cordero, Andrea Malfettone, and Javier Cortes
- Subjects
Cancer Research ,Oncology - Abstract
Background: Cyclin-dependent kinases 4 and 6 inhibitors in combination with endocrine therapy frequently lead to a complete cell-cycle arrest (CCCA) in luminal EBC. However, the rates of pathological complete response (pCR) or Residual Cancer Burden (RCB) 0-I are modest. The effect of this treatment in terms of molecular downstaging as assessed by a genomic signature more refined than Ki67 remains undetermined. We aimed to assess the biological and clinical activity of letrozole plus palbociclib as neoadjuvant treatment in HR-positive/HER2-negative EBC pts with an Oncotype DX RS ≥18. Methods: DxCARTES was a multicenter, open-label, non-comparative, phase 2 trial across 16 hospitals in Spain. Participants were pre- and post-menopausal women aged ≥18 years with centrally confirmed HR-positive/HER2-negative, Ki67≥20%, stage II-IIIB EBC with a RS ≥18. Eligible pts with baseline RS 18-25 (cohort A) and RS 26-100 (cohort B) received six 28-days cycles of letrozole (2.5 mg QD), ± goserelin if premenopausal, plus palbociclib (125 mg QD, 3/1 schedule) before surgery. Tumor samples were prospectively collected at baseline and surgery for Ki67 and RS assessments. The coprimary endpoint was the proportion of pts in either cohort who achieved RS ≤25 or a pCR (ypT0/is ypN0) at surgery. Secondary endpoints included analysis of RCB, preoperative endocrine prognostic index (PEPI), CCCA (Ki67 Citation Format: Antonio Llombart-Cussac, Ángel Guerrero-Zotano, Manuel Ruiz, Begoña Bermejo, Miguel Gil-Gil, Juan de la Haba, Emilio Alba, Vanesa Quiroga, Vicente Carañana, Ander Urruticoechea, Serafín Morales, Meritxell Bellet, Antonio Antón, José Manuel Pérez-García, María Fernández-Abad, Sonia Servitja, Pedro Sánchez-Rovira, Sofia Braga, Miguel Sampayo-Cordero, Andrea Malfettone, Javier Cortes. Neoadjuvant letrozole plus palbociclib in patients (pts) with hormone receptor (HR)-positive/HER2-negative early breast cancer (EBC) with baseline Ki67 ≥20% and an Oncotype DX Breast Recurrence Score® result (RS) ≥18: DxCARTES [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P5-16-12.
- Published
- 2022
32. Abstract P2-15-01: Conversion from luminal to normal intrinsic subtype by PAM50 after neoadjuvant endocrine therapy is associate with biomarkers of good prognosis in luminal breast cancer
- Author
-
Joanna Ines Lopez Velazco, María Otaño, Inazio Lacambra, Kepa Elorriaga, Ainhara Lahuerta, Ana Martínez, Virginia Segur, Sara Manzano, Aleix Prat, María Caffarel, and Ander Urruticoechea
- Subjects
Cancer Research ,Oncology - Abstract
Background. Neoadjuvant endocrine therapy (NET) in luminal breast cancer (LBC) is the perfect scenario for real-time evaluation of the biological and molecular changes that occur after estrogenic deprivation. This evaluation is mainly performed in surgical specimens after NET and has been used as a research tool to obtain prognostic and predictive information using tumor response to decide adjuvant treatment. Nevertheless, there are not many validated biomarkers in this setting to predict response beyond Ki67 levels and modified Preoperative Prognostic Index (mPEPI score) after treatment. Hence, the aim of this study is to determine if changes in mRNA-based PAM50 analysis (intrinsic subtype) after NET in LBC correlate with such biomarkers. Methods. We collected a series of postmenopausal ER+/HER2- breast cancer patients (n=58) treated for at least 4 months with NET before surgery. Next, we performed gene expression analysis by PAM50 accompanied by pathological characterization of surgical tumor specimens and immunohistochemistry characterization of ER and Ki67 levels, both at diagnosis and in surgery specimen. Finally, we studied the association of our results with clinical and histopathological features and with validated biomarkers for endocrine response, such as mPEPI score and changes in Ki67 levels. Results. The distribution of changes in intrinsic subtype determined by PAM50 after NET is presented in Table 1. We observed that tumors that changed from luminal to a normal intrinsic subtype showed larger changes in Ki67 levels after NET and reduced percentage of Ki67 positive cells at surgery, compared to those that presented a luminal persistent status: both analysis P Table 1.- Intrinsic subtypes determined by PAM50 in samples pre and post NETPAM50 PREPAM50 POSTNO.%PERSISTANT LUMINAL STATUSCHANGE FROM LUMINAL TO A NOLMAL INTRINSIC SUBTYPELumALumA2136YESLumANormal1526YESLumALumB35YESLumBLumA610YESLumBNormal35YESLumBLumB59YESHER2HER223NANAHER2LumA12NANALumAHER212NANANormalNormal12NANATOTAL58100Lum=LuminalNA= Not applicable Citation Format: Joanna Ines Lopez Velazco, María Otaño, Inazio Lacambra, Kepa Elorriaga, Ainhara Lahuerta, Ana Martínez, Virginia Segur, Sara Manzano, Aleix Prat, María Caffarel, Ander Urruticoechea. Conversion from luminal to normal intrinsic subtype by PAM50 after neoadjuvant endocrine therapy is associate with biomarkers of good prognosis in luminal breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-15-01.
- Published
- 2022
33. A prospective study on tumour response assessments methods after neoadjuvant endocrine therapy in early oestrogen receptor positive breast cancer
- Author
-
Joanna I. López-Velazco, Sara Manzano, María Otaño, Kepa Elorriaga, Núria Bultó, Julio Herrero, Ainhara Lahuerta, Virginia Segur, Isabel Álvarez-López, Maria M. Caffarel, and Ander Urruticoechea
- Abstract
Neoadjuvant endocrine therapy in oestrogen receptor (ER) positive HER2 negative breast cancer allows real-time evaluation of drug efficacy as well as investigation of the biological and molecular changes that occur after estrogenic deprivation. Clinical and pathological evaluation after neoadjuvant endocrine therapy may be used to obtain prognostic and predictive information of tumour response to decide adjuvant treatment. In this setting, clinical scales developed to evaluate response after neoadjuvant chemotherapy are not useful and there are not many validated biomarkers to assess response to neoadjuvant endocrine therapy beyond Ki67 expression and preoperative prognostic index (PEPI) score. In this prospective study, we extensively analysed radiological (by ultrasound and magnetic resonance imaging) and pathological tumour response of 104 postmenopausal patients with ER+/HER2-resectable breast cancer, treated with neoadjuvant endocrine treatment for a mean of 7 months prior to surgery. Our results show that radiological evaluation by both USS and MRI underestimate pathological tumour size, although they support the use of MRI over USS to clinically assess tumour response. In addition, we propose that the tumour cellularity size, calculated as the product of the percentage of residual tumour cellularity in the surgical specimen and the tumour pathological size, could become a new tool to standardize response assessment to NET given its good correlation with and potential added value to existing biomarkers. Our findings shed light on the dynamics of tumour response to neoadjuvant endocrine therapy, challenge the paradigm of the ability of NET to decrease surgical volume and point to the utility of the tumour cellularity size to quantify the scattered tumour response usually produced by endocrine therapy.
- Published
- 2023
34. Spatial intratumoural heterogeneity in the expression of GIT1 is associated with poor prognostic outcome in oestrogen receptor positive breast cancer patients with synchronous lymph node metastases [version 1; referees: 2 approved with reservations]
- Author
-
Ibai Goicoechea, Ricardo Rezola, María Arestin, María M. Caffarel, Ana Rosa Cortazar, Lorea Manterola, Marta Fernandez-Mercado, María Armesto, Carla Sole, Erika Larrea, Angela M. Araujo, Nerea Ancizar, Arrate Plazaola, Ander Urruticoechea, Arkaitz Carracedo, Irune Ruiz, Isabel Alvarez Lopez, and Charles H. Lawrie
- Subjects
Research Article ,Articles ,Breast Diseases: Benign & Malignant ,GIT1 ,breast cancer ,immunohistochemistry ,biomarker ,lymph node - Abstract
Background: The outcome for oestrogen receptor positive (ER+) breast cancer patients has improved greatly in recent years largely due to targeted therapy. However, the presence of involved multiple synchronous lymph nodes remains associated with a poor outcome. Consequently, these patients would benefit from the identification of new prognostic biomarkers and therapeutic targets. The expression of G-protein-coupled receptor kinase-interacting protein 1 (GIT1) has recently been shown to be an indicator of advanced stage breast cancer. Therefore, we investigated its expression and prognostic value of GIT1 in a cohort of 140 ER+ breast cancer with synchronous lymph node involvement. Methods: Immunohistochemistry was employed to assess GIT1 expression in a tissue microarray (TMA) containing duplicate non-adjacent cores with matched primary tumour and lymph node tissue (n=140). GIT1 expression in tumour cells was scored and statistical correlation analyses were carried out. Results: The results revealed a sub-group of patients that displayed discordant expression of GIT1 between the primary tumour and the lymph nodes (i.e. spatial intratumoural heterogeneity). We observed that loss of GIT1 expression in the metastasis was associated with a shorter time to recurrence, poorer overall survival, and a shorter median survival time. Moreover, multivariate analysis demonstrated that GIT1 expression was an independent prognostic indicator. Conclusions: GIT1 expression enabled the identification of a sub-class of ER+ patients with lymph node metastasis that have a particularly poor prognostic outcome. We propose that this biomarker could be used to further stratify ER+ breast cancer patients with synchronous lymph node involvement and therefore facilitate adjuvant therapy decision making.
- Published
- 2017
- Full Text
- View/download PDF
35. Abstract PS5-39: Tumour cellularity size as a biomarker to predict response after neoadjuvant endocrine therapy: Correlation analysis between Ki67 expression and PEPI score
- Author
-
Ainhara Lahuerta, María Otaño, Virginia Segur, Lide Larburu, María M. Caffarel, Joanna Ines Lopez Velazco, Juan Carlos Irizabal, Kepa Elorriaga, Lourdes Jáuregui, Ana Martínez, and Ander Urruticoechea
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,Correlation analysis ,medicine ,Endocrine therapy ,Biomarker (medicine) ,business - Abstract
Background: Neoadjuvant endocrine therapy (NET) is an approach that allows real-time evaluation of drug efficacy as well as investigation of the biological and molecular changes that occur after estrogenic deprivation. This evaluation is mainly performed in surgical specimens after NET and it has been used as a research tool to obtain prognostic and predictive information using tumour response to decide adjuvant treatment. In this setting, there are not many validated biomarkers to predict response beyond Ki67 expression and Preoperative Prognostic Index (PEPI score) in NET. The aim of this study is to determine if the tumour cellularity size (TCS) in surgical specimen after NET correlates with PEPI score and Ki67 expression. Methods: Retrospective study of postmenopausal patients with estrogen receptor (ER) positive/HER2 negative resectable breast cancer, treated with an aromatase inhibitor for at least 4 months prior to surgery. Pathological characterization of tumour specimens: Evaluation of the percentage of residual tumour cellularity of formalin fixed paraffin embedded surgical specimens and immunohistochemistry characterization of ER and Ki67. Tumour cellularity size: calculated by combining the percentage of residual tumour cellularity and tumour pathological size. Results: N=104. Tumour characteristics at surgery and breakdown for the calculated PEPI score: table 1. Correlation between the percentage of Ki67 positive cells at surgery and TCS: (r=0.2503) p=0.04 (95% CI, 0.0014 to 0.4700). Correlation between TCS and PEPI score: (r=0.2582) p=0.05 (95% CI, -0.0131 to 0.4940). Conclusions: Tumour cellularity size is a promising biomarker to determine response and prognosis after NET. There is a need to find other biomarkers to predict response after NET. Table 1Pathology/Biomarker statusPEPI score RFS pointsNo. of patientspTNot available (NA)1pT1/T20102pT3/T431pNNA7Negative075Positive322KI67 levelNA10%-2.7%039>2.7%-7.3%131>7.3%-19.7%119>19.7%-53.1%213>53.1%31ER-status Allred scoreNA120-2303-8092PEPI scoreNA190281342533495561PEPI groupNot calculated19I (0 score)28II (1-3 score)42III (≥4 score)15 Citation Format: Joanna Ines Lopez Velazco, María Otaño, Lide Larburu, Ainhara Lahuerta, Kepa Elorriaga, Virginia Segur, Juan Carlos Irizabal, Ana Martínez, Lourdes Jáuregui, Maria M. Caffarel, Ander Urruticoechea. Tumour cellularity size as a biomarker to predict response after neoadjuvant endocrine therapy: Correlation analysis between Ki67 expression and PEPI score [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS5-39.
- Published
- 2021
36. Evaluating the clinical effectiveness and safety of various HER2-targeted regimens after prior taxane/trastuzumab in patients with previously treated, unresectable, or metastatic HER2-positive breast cancer: a systematic review and network meta-analysis
- Author
-
Xavier Pivot, Ander Urruticoechea, Ian E. Krop, Noman Paracha, Adriana Reyes, and Véronique Diéras
- Subjects
Bridged-Ring Compounds ,Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,Network Meta-Analysis ,Neratinib ,Breast Neoplasms ,Review ,Lapatinib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Trastuzumab ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Adjuvant therapy ,Humans ,Molecular Targeted Therapy ,030212 general & internal medicine ,Trastuzumab emtansine ,Capecitabine ,Pertuzumab ,business.industry ,medicine.disease ,Treatment Outcome ,Tolerability ,chemistry ,030220 oncology & carcinogenesis ,Female ,Taxoids ,Locally advanced ,business ,medicine.drug - Abstract
Purpose In the absence of head-to-head trial data, network meta-analysis (NMA) was used to compare trastuzumab emtansine (T-DM1) with other approved treatments for previously treated patients with unresectable or metastatic HER2-positive breast cancer (BC). Methods Systematic reviews were conducted of published controlled trials of treatments for unresectable or metastatic HER2-positive BC with early relapse (≤ 6 months) following adjuvant therapy or progression after trastuzumab (Tras) + taxane published from January 1998 to January 2018. Random-effects NMA was conducted for overall survival (OS), progression-free survival (PFS), overall response rate (ORR), and safety endpoints. Results The NMA included regimens from seven randomized controlled trials: T-DM1 and combinations of Tras, capecitabine (Cap), lapatinib (Lap), neratinib, or pertuzumab (Per; unapproved). OS results favored T-DM1 over approved comparators: hazard ratio (HR) (95% credible interval [95% CrI]) vs Cap 0.68 (0.39, 1.10), LapCap 0.76 (0.51, 1.07), TrasCap 0.78 (0.44, 1.19). PFS trends favored T-DM1 over all other treatments: HR (95% CrI) vs Cap 0.38 (0.19, 0.74), LapCap 0.65 (0.40, 1.10), TrasCap 0.62 (0.34, 1.18); ORR with T-DM1 was more favorable than with all approved treatments. In surface under cumulative ranking curve (SUCRA) analysis T-DM1 ranked highest for all efficacy outcomes. Discontinuation due to adverse events was less likely with T-DM1 than with all comparators except neratinib. In general, gastrointestinal side effects were less likely and elevated liver transaminases and thrombocytopenia more likely with T-DM1 than with comparators. Conclusions The efficacy and tolerability profiles of T-DM1 are generally favorable compared with other treatments for unresectable or metastatic HER2-positive BC.
- Published
- 2020
37. Abstract P5-11-01: Phamacodynamic and circulating tumor DNA evaluation in a phase I study of GDC-0927, a selective estrogen receptor antagonist/ degrader (SERD)
- Author
-
Erika Hamilton, Maura N. Dickler, Jennifer O. Lauchle, Mary Gates, EP Winer, Ciara Metcalfe, J.A. Perez Fidalgo, Ander Urruticoechea, Jill M. Spoerke, Eric W. Humke, Xuehai Wang, Rui Li, A Daemen, Jill Fredrickson, Ingrid A. Mayer, M. Martin, Aditya Bardia, Valentina Boni, LS Friedman, A. González Martín, Sravanthi Cheeti, Lars Mueller, S Milan, Iris T. Chan, Jennifer M. Giltnane, Luna Liu, J. Cortes, Meritxell Bellet, C-W Chang, and Lackner
- Subjects
Cancer Research ,biology ,medicine.drug_class ,business.industry ,Wild type ,Estrogen receptor ,medicine.disease ,Molecular biology ,Metastatic breast cancer ,CDH1 ,Breast cancer ,Oncology ,Estrogen ,medicine ,biology.protein ,PTEN ,business ,Estrogen receptor alpha - Abstract
Background: Modulation of estrogen activity and/or synthesis is the mainstay therapeutic strategy in the treatment of ER positive breast cancer. However, despite the effectiveness of available endocrine therapies, many patients ultimately relapse or develop resistance to these agents via estrogen-dependent and estrogen-independent mechanisms, including mutations in ESR1 affecting the ER ligand binding domain that drive ER-dependent transcription and proliferation in the absence of estrogen. Based on preclinical and clinical data, SERDs are expected be effective in patients harboring ESR1 mutations. Biomarker analysis was performed on plasma and tumor samples from the Phase I study of GDC-0927 in metastatic breast cancer (Dickler et al, SABCS 2017) with the goal of evaluating activity in both ESR1 mutant and wildtype tumors, and to assess ER pathway modulation. Methods: Hotspot mutations in ESR1, PIK3CA, and AKT1 were analyzed in baseline, on-treatment and end of treatment plasma derived circulating tumor DNA (ctDNA) using the BEAMing assay in patients treated at multiple dose levels of GDC-0927. A subset of samples was analyzed with Foundation Medicine's next generation sequencing ctDNA assay (FACT), which covers genomic alterations in 62 commonly altered genes. Paired pre- and on-treatment biopsies were collected to assess ER pathway modulation. ER, PR, and Ki67 protein levels were analyzed by immunohistochemistry. Gene expression analysis was performed using Illumina's RNA Access library preparation kit followed by paired-end (2x50b, 50M reads) sequencing on the HiSeq. Results: Baseline and on-treatment plasma samples were available for 40 patients. ESR1 and PIK3CA mutations were observed in 52% and 33% of patient baseline samples, respectively (BEAMing method). Mutant allele frequencies (MAF) generally declined in the first on-treatment samples collected for both ESR1 (16 out of 21 samples) and PIK3CA (7 out of 12 samples). The majority of the reductions were greater than 95% relative to baseline. Increases in ESR1 MAFs were observed in later time-points and were not associated with any particular ESR1 mutation. There were six instances for which an ESR1 mutation was detected in an on-treatment sample that was not detected in the baseline sample, three at L536P and one each at D538G, L536H, and S463P, and four out of six with MAFs close to the limit of detection. The FACT assay also detected alterations in CDH1, NF1, PTEN, and TP53 in baseline samples. The relationship between MAF changes and clinical benefit to GDC-0927 will be presented. A predefined, experimentally-derived set of ER target genes were evaluated in pre- and on-treatment tumor biopsy pairs from six patients. Four of the six patients showed evidence of suppression in ER pathway activity, one patient treated at the 1000 mg dose level and three at the 1400 mg dose. The degree of pathway suppression was associated with pre-treatment pathway levels and decreases of ER and Ki67 protein levels. Conclusions: We report here evidence of consistent reduction of ESR1 and PIK3CA ctDNA in patients treated with GDC-0927. ER pathway suppression was observed at both the transcript and protein level confirming pharmacodynamic activity of the SERD. Citation Format: Spoerke JM, Daemen A, Chang C-W, Giltnane J, Metcalfe C, Dickler MN, Bardia A, Perez Fidalgo JA, Mayer IA, Boni V, Winer EP, Hamilton EP, Bellet M, Urruticoechea A, Gonzalez Martin A, Cortes J, Martin M, Gates M, Cheeti S, Fredrickson J, Wang X, Friedman LS, Liu L, Li R, Chan IT, Mueller L, Milan S, Lauchle J, Humke EW, Lackner MR. Phamacodynamic and circulating tumor DNA evaluation in a phase I study of GDC-0927, a selective estrogen receptor antagonist/ degrader (SERD) [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P5-11-01.
- Published
- 2019
38. Abstract P2-06-16: New targets in triple negative breast cancer: Role of Oncostatin M receptor pathway
- Author
-
Ander Urruticoechea, R Rezola, Angela M Araujo, Isabel Alvarez, María M. Caffarel, A. Abaurrea, and Charles H. Lawrie
- Subjects
0301 basic medicine ,Cancer Research ,Oncogene ,biology ,business.industry ,Angiogenesis ,Oncostatin M ,Cancer ,Oncostatin M receptor ,medicine.disease ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Medicine ,business ,Triple-negative breast cancer - Abstract
BACKGROUND:Triple negative breast cancer (TNBC) has poor prognosis, lack of targeted therapies and are often refractory to conventional chemotherapy treatments. Therefore, finding new therapeutic targets for those tumours is an unmet need with high clinical impact. In this context, Oncostatin M receptor (OSMR) is a promising therapeutic target as it is over-expressed in this tumour subtype and its activation promotes invasiveness (Guo L, et al. 2013 Oncogene; West NR, et al.2014 Oncogene). We previously showed that OSMR is frequently copy-number gained and over-expressed in squamous cell carcinoma, where it induces migration, invasion and metastasis (Caffarel MM, et al 2013 Journal of Pathology; Caffarel MM, et al 2014 Journal of Pathology; Kucia-Tran JA, et al. 2016 Brit J Cancer; Kucia-Tran JA, et al 2018 Journal of Pathology). We now investigate the role of OSMR in breast cancer progression. METHODS: To address this issue we use a wide array of tools including in vitro cell cultures and in vivo models. The expression of OSMR pathway was analysed in FFPE samples and large datasets of publicly available breast cancer samples (METABRIC, n=1462; and TCGA, n=547). RESULTS: OSMR and its ligand Oncostatin M (OSM) are over-expressed in basal tumours, where they associate with shorter overall survival (p=0.015). While OSMR is expressed by breast cancer cells and cancer associated fibroblasts, the main source of OSM seems to be primarily macrophages. OSM treatment of breast cancer cells induces the expression of important mediators of angiogenesis and invasion. Importantly, OSMR activation accelerates tumour onset, tumour growth and metastasis in orthotopic xenografts in nude mice. CONCLUSIONS: Our results support that OSMR pathway may have an important role in the initiation and progression of breast cancer and that it could be a promising candidate for therapeutic targeting in TNBC. OSMR could be blocked by antibody based inhibition, strategy that has had a major impact on breast cancer. Citation Format: Alvarez I, Araujo A, Abaurrea A, Rezola R, Urruticoechea A, Lawrie C, Caffarel MM. New targets in triple negative breast cancer: Role of Oncostatin M receptor pathway [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P2-06-16.
- Published
- 2019
39. Discovery and Proof-of-Concept Study of Nuclease Activity as a Novel Biomarker for Breast Cancer Tumors
- Author
-
Luiza I. Hernandez, Marcos J. Araúzo-Bravo, Daniela Gerovska, Ricardo Rezola Solaun, Isabel Machado, Alien Balian, Juliana Botero, Tania Jiménez, Olaia Zuriarrain Bergara, Lide Larburu Gurruchaga, Ander Urruticoechea, Frank J. Hernandez
- Published
- 2021
- Full Text
- View/download PDF
40. Stromal Oncostatin M axis promotes breast cancer progression
- Author
-
A. Abaurrea, Arkaitz Carracedo, Azcoaga P, Alexandar Tzankov, Rezola R, López-Velazco Ji, Charles H. Lawrie, Juana M. Flores, María M. Caffarel, Alvarez-Lopez I, Liam Jenkins, David Gallego-Ortega, Paloma Bragado, Iñaki Osorio-Querejeta, Eppenberger-Castori S, Fatima Valdes-Mora, Clare M. Isacke, Ander Urruticoechea, Nicholas Coleman, Fernando Calvo, Nicola Ferrari, Natalia Martín-Martín, Angela M Araujo, and Patricia Fernández-Nogueira
- Subjects
Stromal cell ,biology ,medicine.medical_treatment ,fungi ,Oncostatin M ,Oncostatin M receptor ,Cancer ,medicine.disease ,Cytokine ,Stroma ,Cancer cell ,Cancer research ,biology.protein ,medicine ,CXCL16 - Abstract
Cancer cells are constantly communicating with the surrounding tumour microenvironment (TME) and they hijack physiological cell interactions to overcome immune system surveillance and promote cancer progression1,2. However, the contribution of stromal cells to the reprogramming of the TME is not well understood. In this study we provide unprecedented evidence of the role of the cytokine Oncostatin M (OSM) as central node for multicellular interactions between immune and non-immune stroma and the epithelial compartment. We show that stromal expression of the OSM:Oncostatin M Receptor (OSMR) axis plays a key role in breast cancer progression. OSMR deletion in a multistage breast cancer model delays tumour onset, tumour growth and reduces metastatic burden. We ascribed causality to the stromal function of OSM axis by demonstrating reduced tumour burden of syngeneic tumours implanted in mice. Single-cell and bioinformatic analysis of murine and human breast tumours revealed that the expression of OSM signalling components is compartmentalized in the tumour stroma. OSM expression is restricted to myeloid cells, whereas OSMR expression is detected predominantly in fibroblasts and, to a lower extent, cancer cells. Myeloid-derived OSM reprograms fibroblasts to a more contractile and pro-tumorigenic phenotype, elicits the secretion of VEGF and pro-inflammatory chemokines (e.g. CXCL1 and CXCL16), leading to increased neutrophil and macrophage recruitment. In summary, our work sheds light on the mechanism of immune regulation by the tumour microenvironment, and supports that targeting OSM:OSMR interactions is a potential therapeutic strategy to inhibit tumour-promoting inflammation and breast cancer progression.
- Published
- 2020
41. Altered Levels of Desaturation and ω-6 Fatty Acids in Breast Cancer Patients' Red Blood Cell Membranes
- Author
-
Joana Camba, Gurutze Ugartemendia, Nerea Bretaña, Carla Ferreri, Javier Amézaga, Ander Urruticoechea, Aizpea Iruretagoyena, Aitziber Larraioz, and Itziar Tueros
- Subjects
0301 basic medicine ,linoleic acid ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Linoleic acid ,lcsh:QR1-502 ,membrane lipidome ,red blood cell ,medicine.disease_cause ,Biochemistry ,lcsh:Microbiology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,breast cancer ,Internal medicine ,medicine ,arachidonic acid ,Molecular Biology ,chemistry.chemical_classification ,business.industry ,Cancer ,Fatty acid ,omega 6 ,medicine.disease ,Red blood cell ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Arachidonic acid ,Carcinogenesis ,business ,SCD1 ,Polyunsaturated fatty acid - Abstract
Red blood cell (RBC) membrane can reflect fatty acid (FA) contribution from diet and biosynthesis. In cancer, membrane FAs are involved in tumorigenesis and invasiveness, and are indicated as biomarkers to monitor the disease evolution as well as potential targets for therapies and nutritional strategies. The present study provides RBC membrane FA profiles in recently diagnosed breast cancer patients before starting chemotherapy treatment. Patients and controls were recruited, and their dietary habits were collected. FA lipidomic analysis of mature erythrocyte membrane phospholipids in blood samples was performed. Data were adjusted to correct for the effects of diet, body mass index (BMI), and age, revealing that patients showed lower levels of saturated fatty acids (SFA) and higher levels of monounsaturated fatty acid, cis-vaccenic (25%) than controls, with consequent differences in desaturase enzymatic index (∆9 desaturase, &ndash, 13.1%). In the case of polyunsaturated fatty acids (PUFA), patients had higher values of &omega, 6 FA (C18:2 (+11.1%), C20:4 (+7.4%)). RBC membrane lipidomic analysis in breast cancer revealed that &omega, 6 pathways are favored. These results suggest new potential targets for treatments and better nutritional guidelines.
- Published
- 2020
42. A phase Ib study of sonidegib (LDE225), an oral small molecule inhibitor of smoothened or Hedgehog pathway, in combination with docetaxel in triple negative advanced breast cancer patients: GEICAM/2012–12 (EDALINE) study
- Author
-
Sara Benito, Jesus Corral, Susana Bezares, Helena Colom, Eva Carrasco, Ander Urruticoechea, Miguel Martin, Jose Manuel Trigo, Yolanda Jerez, José A. García-Sáenz, Federico Rojo, Silvia Antolín, Rosalia Caballero, Begoña Jiménez, Nuria Gonzalo, Carmen Muñoz, and Manuel Ruiz-Borrego
- Subjects
Adult ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Maximum Tolerated Dose ,Pyridines ,Triple Negative Breast Neoplasms ,Docetaxel ,Neutropenia ,Sonidegib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Tissue Distribution ,Pharmacology (medical) ,Adverse effect ,Triple-negative breast cancer ,Aged ,Pharmacology ,Leukopenia ,business.industry ,Biphenyl Compounds ,Carcinoma, Ductal, Breast ,Cancer ,Middle Aged ,Prognosis ,medicine.disease ,Smoothened Receptor ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Smoothened ,business ,medicine.drug - Abstract
Up-regulation of the Hedgehog (Hh) pathway is implicated in the genesis of a wide range of tumors including triple negative breast cancer (TNBC). Sonidegib is a potent and selective oral inhibitor of Smo, a key component of the Hh signaling pathway. We designed a phase I clinical study to explore the combination of sonidegib plus docetaxel (fixed dose at 75 mg/m2) in advanced TNBC patients. The primary objective was to ascertain the combination’s maximum tolerated dose and the recommended phase II dose (RP2D), based on dose limiting toxicities (DLTs) in the first 2 cycles. A standard “3 + 3” design was followed including three dose levels (DL) of sonidegib: 400 mg (DL1), 600 mg (DL2), and 800 mg (DL3). Twelve patients were included. Sonidegib 800 mg orally q.d. plus docetaxel 75 mg/m2 given intravenously on day 1 of 21-day cycles was established as the RP2D. No DLTs were observed at any DL. The median number of administered cycles at DL3 was 8 (range: 6 to 9). Grade 3 adverse events (AEs) at DL3 were neutropenia (66.7%), CPK increase (33.3%), leukopenia (33.3%), and paresthesia (33.3%), grade 4 AEs were not reported at this DL. At the RP2D, the combination showed antitumor activity in three out of 10 patients with measurable disease. Median time to progression for the overall study was 42.5 days (95% Confidence Interval: 29–155), and 188 days at DL3. No drug-to-drug interactions between sonidegib and docetaxel were found in the PK assessment. Trial Registration: EudraCT study number: 2013–001750-96. Study GEICAM/2012–12. TRIAL REGISTRATION: EudraCT study number: 2013-001750-96. Study GEICAM/2012-12. ClinicalTrials.gov: NCT02027376
- Published
- 2018
43. Abstract PD5-10: A first-in-human phase I study to evaluate the oral selective estrogen receptor degrader (SERD), GDC-0927, in postmenopausal women with estrogen receptor positive (ER+) HER2-negative metastatic breast cancer (BC)
- Author
-
J.A. Perez Fidalgo, Luna Liu, Antonio González-Martín, U McCurry, J. Cortés, Margaret A. Gates, Jill Fredrickson, Lars Mueller, Xiaojing Wang, Jennifer O. Lauchle, Ingrid A. Mayer, EP Winer, Roland Morley, Jennifer M. Giltnane, Erika Hamilton, Sravanthi Cheeti, R Villanueva, Lori Friedman, Ander Urruticoechea, Maura N. Dickler, Eric W. Humke, Iris T. Chan, S Milan, Meritxell Bellet, Jill M. Spoerke, M. Martin, Aditya Bardia, Valentina Boni, Ciara Metcalfe, and Robert Jinze Li
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,business.industry ,Estrogen receptor ,medicine.disease ,Metastatic breast cancer ,03 medical and health sciences ,030104 developmental biology ,Breast cancer ,Pharmacokinetics ,Estrogen ,Internal medicine ,Pharmacodynamics ,medicine ,Adverse effect ,business ,Estrogen receptor alpha - Abstract
Background: Modulation of estrogen activity and/or synthesis is the mainstay therapeutic strategy in the treatment of ER+ BC. However, despite the effectiveness of available endocrine therapies, many patients ultimately relapse or develop resistance to these agents via estrogen-dependent and estrogen-independent mechanisms, including mutations in ESR1 affecting the ER ligand binding domain that drive ER-dependent transcription and proliferation in the absence of estrogen. ER antagonists that are efficacious against ligand-dependent and ligand-independent, constitutively active ESR1 mutant tumors may be of substantial therapeutic benefit. GDC-0927 (formerly known as SRN-927) is a novel, potent, non-steroidal, orally bioavailable, selective ER antagonist/ER degrader (SERD) that induces tumor regression in ER+ BC patient-derived xenograft models. Methods: A phase I dose escalation study with 3+3 design was conductedin postmenopausal women with ER+ (HER2-) metastatic BC (progressing ≥ 6 months on endocrine therapy and with ≤ 2 prior chemotherapies in the advanced or metastatic setting) to determine the safety, pharmacokinetics (PK) and the recommended Phase 2 dose (RP2D) of GDC-0927. Pharmacodynamic (PD) activity was assessed with [18F]-fluoroestradiol (FES)-PET scans. Plasma PK samples (after single dose and at steady state), CT scans, and when feasible, pre and on-study tumor biopsies were obtained Results: From March 16, 2015 to March 17, 2017 patients (pts) with a median age of 53 years (range 44-69) and a median number of prior therapies for MBC 4 (range 1-7) were enrolled at 3 total daily dose levels (600, 1000, 1400 mg) once daily (QD) given orally with fasting (n = 12). Increases in GDC-0927 exposure were approximately dose proportional. Treatment related adverse events (AEs) were all grade 1 or 2. The most common treatment-related AEs were nausea (54%, n = 7), diarrhea (46%, n = 6), elevated aspartate aminotransferase (39%, n = 5) and anemia, constipation, (each 31%, n = 4). Treatment interruption was required for 2 pts due to nausea and vomiting. Of those pts with FES-PET avid disease at baseline (9 of 12), all post-therapy scans showed complete or near complete (> 90%) suppression of FES uptake to background levels, including pts with ESR1 mutations. Evidence of reduced ER levels and Ki67 staining was observed in on-treatment biopsies. Five of 12 pts (1 at 600 mg and 4 at 1400 mg) were on study ≥ 24 weeks (CBR = 41.6 %) with the best overall response of stable disease with 1 patient (ESR1 mt+ D538G) on study for over 490 days. There were no dose limiting toxicities and no SAEs related to study drug. R2PD was 1400 mg and was selected for single arm dose-expansion which is now complete with last patient enrolled on March 17, 2017. Updated results from dose-escalation and dose-expansion will be presented at the meeting (N = 43). Conclusions: GDC-0927 appears well-tolerated to date with PK exposure supporting QD dosing, evidence of robust PD target engagement, and encouraging anti-tumor activity in heavily pretreated pts with advanced or metastatic ER+ BC, including pts with ESR1 mutations. Citation Format: Dickler MN, Villanueva R, Perez Fidalgo JA, Mayer IA, Boni V, Winer EP, Hamilton EP, Bellet M, Urruticoechea A, Gonzalez-Martin A, Cortes J, Martin M, Giltnane J, Gates M, Cheeti S, Fredrickson J, Wang X, Friedman LS, Spoerke JM, Metcalfe C, Liu L, Li R, Morley R, McCurry U, Chan IT, Mueller L, Milan S, Lauchle J, Humke EW, Bardia A. A first-in-human phase I study to evaluate the oral selective estrogen receptor degrader (SERD), GDC-0927, in postmenopausal women with estrogen receptor positive (ER+) HER2-negative metastatic breast cancer (BC) [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr PD5-10.
- Published
- 2018
44. Phase II study of buparlisib (BKM120) and trastuzumab in patients with HER2+ locally advanced or metastatic breast cancer resistant to trastuzumab-based therapy
- Author
-
Patrick Urban, Barbara Pistilli, Emmanuelle di Tomaso, D. Farci, Cristian Massacesi, Ander Urruticoechea, Cristina Saura, Anthony Kong, H. S. Han, Steve Chan, Guy Jerusalem, S. L. Mouhaer, Thomas Bachelot, Douglas Robinson, and T. Pluard
- Subjects
Adult ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Receptor, ErbB-2 ,Morpholines ,Buparlisib ,Aminopyridines ,Phases of clinical research ,Breast Neoplasms ,Article ,Capecitabine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Trastuzumab ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Humans ,skin and connective tissue diseases ,Response Evaluation Criteria in Solid Tumors ,Aged ,Brain Neoplasms ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,Metastatic breast cancer ,030104 developmental biology ,Tolerability ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,business ,medicine.drug - Abstract
PURPOSE: A Phase Ib study in patients with trastuzumab-resistant, human epidermal growth factor receptor-2- (HER2)-positive advanced breast cancer defined the recommended Phase II dose of buparlisib as 100 mg/day in combination with 2 mg/kg weekly trastuzumab, and reported preliminary signs of clinical activity. Here we present results from the Phase II portion. METHODS: Patients with trastuzumab-resistant, HER2-positive advanced breast cancer received buparlisib plus trastuzumab. Study endpoints included safety/tolerability and antitumour activity. The study was extended to include a Phase Ib dose-escalation phase, in which patients with progressive brain metastases also received capecitabine. RESULTS: In the Phase II portion, of 50 patients treated with buparlisib and trastuzumab, the most common (≥ 30%) all-grade adverse events (AEs) were diarrhoea (54%), nausea (48%), decreased appetite, increased alanine aminotransferase (36% each), increased aspartate aminotransferase (34%), fatigue, rash (32% each), cough and hyperglycemia (30% each). One (2%) patient achieved complete response and four (8%) patients had confirmed partial responses [PR; including two patients with phosphatidylinositol 3-kinase (PI3 K) pathway-activated tumours]. Overall response rate (ORR) was 10%: the primary endpoint (ORR ≥ 25%) was therefore not met. In the Phase Ib portion, all patients with measurable brain lesions at baseline showed tumour shrinkage to some degree; due to low enrollment, maximum tolerated dose of buparlisib in combination with trastuzumab and capecitabine was not determined. CONCLUSION: Buparlisib plus trastuzumab, as a chemotherapy-free regimen, demonstrated an acceptable safety profile but limited efficacy in patients with heavily pretreated, trastuzumab-resistant HER2-positive breast cancer, and in patients with progressive brain metastases also receiving capecitabine.
- Published
- 2017
45. Effects of Esomeprazole on the Pharmacokinetics of Lapatinib in Breast Cancer Patients
- Author
-
Koch, Kevin M., Im, Young-Hyuck, Kim, Sung-Bae, Ribate, Ander Urruticoechea, Stephenson, Joe, Botbyl, Jeffrey, Cartee, Leanne, Holshouser, Jane, and Ridgway, Derry
- Published
- 2013
- Full Text
- View/download PDF
46. Randomized Phase III Trial of Trastuzumab Plus Capecitabine With or Without Pertuzumab in Patients With Human Epidermal Growth Factor Receptor 2–Positive Metastatic Breast Cancer Who Experienced Disease Progression During or After Trastuzumab-Based Therapy
- Author
-
Ander Urruticoechea, Seock-Ah Im, Antonio Carlos Sánchez Ruiz, M. Rizwanullah, J Eng-Wong, István Láng, Montserrat Muñoz, Hannah Douthwaite, Gianluca Tomasello, Tanja Badovinac Crnjevic, and Sarah Heeson
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,Breast Neoplasms ,Neutropenia ,Antibodies, Monoclonal, Humanized ,Drug Administration Schedule ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,Trastuzumab ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,Humans ,Medicine ,Neoplasm Metastasis ,Taxane ,business.industry ,Hazard ratio ,medicine.disease ,Metastatic breast cancer ,030104 developmental biology ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,Pertuzumab ,business ,medicine.drug - Abstract
Purpose To assess the efficacy and safety of trastuzumab plus capecitabine with or without pertuzumab in patients with human epidermal growth factor receptor 2–positive metastatic breast cancer who experienced disease progression during or after trastuzumab-based therapy and received a prior taxane. Patients and Methods Patients were randomly assigned to arm A: trastuzumab 8 mg/kg → 6 mg/kg once every 3 weeks plus capecitabine 1,250 mg/m2 twice a day (2 weeks on, 1 week off, every 3 weeks); or arm B: pertuzumab 840 mg → 420 mg once every 3 weeks plus trastuzumab at the same dose and schedule as arm A plus capecitabine 1,000 mg/m2 on the same schedule as arm A. The primary end point was independent review facility–assessed progression-free survival (IRF PFS). Secondary end points included overall survival (OS) and safety. Hierarchical testing procedures were used to control type I error for statistical testing of IRF PFS, OS, and objective response rate. Results Randomly assigned (intent-to-treat) populations were 224 and 228 patients in arms A and B, respectively. Median IRF PFS at 28.6 and 25.3 months’ median follow-up was 9.0 v 11.1 months (hazard ratio, 0.82; 95% CI, 0.65 to 1.02; P = .0731) and interim OS was 28.1 v 36.1 months (hazard ratio, 0.68; 95% CI, 0.51 to 0.90). The most common adverse events (all grades; incidence of ≥ 10% in either arm and ≥ 5% difference between arms) were hand-foot syndrome, nausea, and neutropenia in arm A, and diarrhea, rash, and nasopharyngitis in arm B. Conclusion The addition of pertuzumab to trastuzumab and capecitabine did not significantly improve IRF PFS. An 8-month increase in median OS to 36.1 months with pertuzumab was observed. Statistical significance for OS cannot be claimed because of the hierarchical testing of OS after the primary PFS end point; however, the magnitude of OS difference is in keeping with prior experience of pertuzumab in metastatic breast cancer. No new safety signals were identified.
- Published
- 2017
47. Efficacy and safety of dasatinib with trastuzumab and paclitaxel in first line HER2-positive metastatic breast cancer: results from the phase II GEICAM/2010-04 study
- Author
-
Nuria Ribelles, Sara Benito, Sonia Pernas, Ander Urruticoechea, Rosalia Caballero, Juan Carlos Montero, Silvia Antolín, Javier Orlando, Atanasio Pandiella, Eva Carrasco, Alejandro Falcon, M. J. Escudero, Alberto Ocaña, Federico Rojo, Alvaro Montaño, María Atienza, M. Gil-Martin, Manuel Ruiz-Borrego, and Bristol Myers Squibb Foundation
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Receptor, ErbB-2 ,Dasatinib ,chemistry.chemical_compound ,0302 clinical medicine ,Trastuzumab ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Neoplasm Metastasis ,Phosphorylation ,skin and connective tissue diseases ,Trastuzumab resistance ,Extracellular Signal-Regulated MAP Kinases ,Aged, 80 and over ,Middle Aged ,Metastatic breast cancer ,Phase II ,Treatment Outcome ,Paclitaxel ,030220 oncology & carcinogenesis ,Female ,SRC kinase inhibitor ,medicine.drug ,Adult ,medicine.medical_specialty ,Proto-Oncogene Proteins pp60(c-src) ,Breast Neoplasms ,Neutropenia ,Drug Administration Schedule ,03 medical and health sciences ,Breast cancer ,Internal medicine ,medicine ,Humans ,Adverse effect ,HER2-positive breast cancer ,neoplasms ,Aged ,business.industry ,medicine.disease ,Survival Analysis ,030104 developmental biology ,chemistry ,Pharmacodynamics ,business ,Proto-Oncogene Proteins c-akt - Abstract
[Background]: An important proportion of HER2-positive metastatic breast cancer patients do not respond to trastuzumab. The combination of dasatinib and trastuzumab has shown to be synergistic in preclinical models., [Methods]: We conducted a phase II trial combining dasatinib 100 mg once daily with trastuzumab 2 mg/kg and paclitaxel 80 mg/m2 weekly. Primary objective was objective response rate (ORR) and secondary included safety, other efficacy parameters and pharmacodynamics in tumour tissue, blood samples and skin biopsies., [Results]: From June 2013 to December 2015, 29 patients were included. Median number of cycles was 12 (1–49). Only 6 patients discontinued due to adverse events. ORR was 79.3% (95% CI 60.3–92), clinical benefit rate 82.8% (95% CI 64.2–94.2). Median time to progression 23.9 months (95% CI 14.9–not reached [NR]), median progression-free survival 23.9 months (95% CI 10.3–NR). No grade 4 toxicity was seen. Grade 3 toxicities included: ejection fraction decrease, neutropenia, hyponatremia, fatigue and sensory neuropathy and one left ventricular systolic dysfunction. Phosphorylated (p)-SRC was reduced in peripheral blood mononuclear cells. Phosphorylated SRC, ERK and AKT were also reduced in epidermal keratinocytes., [Conclusions]: Dasatinib can be safely combined with trastuzumab and paclitaxel. The combination is active with an ORR of almost 80%. Trial registration: NCT01306942, EudraCT 2010-023304-27., The study was financially supported by Bristol-Myers Squibb which also supplied the dasatinib.
- Published
- 2018
48. 129TiP Metformin (MF) in the prevention of hyperglycemia (HG) in patients (pts) with PIK3CA-mutated, hormone receptor (HR)[+]/HER2[–] advanced breast cancer (ABC) treated with alpelisib (ALP) plus fulvestrant (F): METALLICA
- Author
-
Marta Capelán, B. Bermejo De Las Heras, E. Martínez, A. Fernádez, E. Galve, Jesús Alonso, M. Ruiz Borrego, Ander Urruticoechea, Salvador Blanch, F. Gómez-Peralta, Beatriz Rojas, M. Gion Cortes, Jose Perez-Garcia, J.F. Cueva Banuelos, Asunción Camino López, T. Martos, J. Ponce, A. Llombart Cussac, J. Cortés, and P. Tolosa
- Subjects
Oncology ,medicine.medical_specialty ,Fulvestrant ,business.industry ,Advanced breast ,Cancer ,Hematology ,medicine.disease ,Metformin ,Hormone receptor ,Internal medicine ,Medicine ,In patient ,business ,medicine.drug - Published
- 2021
49. 14P Immunomodulatory effect of denosumab in early breast cancer: Preliminary results of a randomized window-opportunity clinical trial D-Biomark
- Author
-
A. Vethencourt, Amparo Garcia-Tejedor, A. Guma Martinez, Teresa Soler, Agostina Stradella, Catalina Falo, C. Capó, Eva M. Trinidad, S. Recalde Penabad, M. Pla, Eva González-Suárez, C. Gómez Aleza, S. Pernas Simon, A. Fernádez, A. Iserte, Anna Petit, Sandra Vázquez, M. Cejuela, M. Gil Gil, and Ander Urruticoechea
- Subjects
Clinical trial ,Oncology ,medicine.medical_specialty ,Denosumab ,business.industry ,Internal medicine ,medicine ,Window (computing) ,Hematology ,business ,medicine.drug ,Early breast cancer - Published
- 2021
50. Measurement of total and free docetaxel concentration in human plasma by ultra-performance liquid chromatography–tandem mass spectrometry
- Author
-
Helena Colom, Catalina Falo, Núria Gonzalo-Diego, Ander Urruticoechea, Raül Rigo-Bonnin, Carmen Muñoz-Sánchez, Sara Cobo-Sacristán, and Pedro Alía
- Subjects
Electrospray ionization ,Clinical Biochemistry ,Ultrafiltration ,Pharmaceutical Science ,Antineoplastic Agents ,Ion suppression in liquid chromatography–mass spectrometry ,Docetaxel ,Tandem mass spectrometry ,Mass spectrometry ,030226 pharmacology & pharmacy ,01 natural sciences ,High-performance liquid chromatography ,Analytical Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,Liquid chromatography–mass spectrometry ,Drug Discovery ,medicine ,Humans ,Chromatography, High Pressure Liquid ,Spectroscopy ,Chromatography ,Chemistry ,010401 analytical chemistry ,Selected reaction monitoring ,0104 chemical sciences ,Taxoids ,medicine.drug - Abstract
Docetaxel is a semi-synthetic taxane with cytotoxic anti-neoplastic activity and, currently used as anticancer agent in several types of cancer. Docetaxel is highly bound to plasma proteins, and this significantly determines its clearance and activity. Therefore, measurement of free docetaxel in plasma is pharmacologically important when pharmacokinetics is investigated. We developed and validated chromatographic methods by ultra-performance liquid chromatography-tandem mass spectrometry to measure total and free docetaxel concentration in human plasma. The final validated methods involved liquid-liquid extraction followed by dryness under nitrogen evaporation. To measure free docetaxel concentration, sample preparation was preceded by ultrafiltration. Chromatographic separation was achieved using an Acquity(®) UPLC(®) BEH™ (2.1×100 mm id, 1.7 μm) reverse-phase C18 column at a flow rate of 0.4 mL/min, using isocratic elution mode containing ammonium acetate/formic acid in water/methanol (30:70 v/v) as mobile phase. Docetaxel and its internal standard (paclitaxel) were detected by electrospray ionization mass spectrometry in positive ion multiple reaction monitoring mode using mass-to-charge (m/z) transitions of 808.3→527.0 (quantifier) and 808.3→509.0 (qualifier); and 854.3→569.0 (quantifier) and 854,3→509,0 (qualifier), respectively. The run time per sample was 3.5 min. The limits of quantification were 1,95 and 0.42 μg/L and linearity was observed between 1.95 and 1000 and 0.42-100 μg/L for total and free docetaxel, respectively. Coefficients of variation and absolute relative biases were less than 13.8% and 10.0%. Recovery values were greater than 79.4%. Evaluation of the matrix effect showed ion suppression and no carry-over was observed. The validated methods could be useful for both therapeutic drug monitoring and pharmacokinetic studies. They could be applied to daily clinical laboratory practice to measure the concentration of total and free docetaxel in plasma.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.