Rafael López
University of Santiago de Compostela
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Rafael López.
Journal of Clinical Oncology | 2005
Jaime Feliu; P. Escudero; Ferrán Llosa; Matilde Bolaños; Jose-Manuel Vicent; Alfonso Yubero; Jose-Javier Sanz-Lacalle; Rafael López; Luis Lopez-Gomez; Enrique Casado; María-José Gómez-Reina; Manuel González-Barón
PURPOSE To determine the tolerability of capecitabine in elderly patients with advanced colorectal cancer (CRC). PATIENTS AND METHODS Fifty-one patients with advanced CRC who were >/= 70 years and considered ineligible for combination chemotherapy received oral capecitabine 1,250 mg/m(2) twice daily on days 1 to 14 every 3 weeks. Patients with a creatinine clearance of 30 to 50 mL/min received a dose of 950 mg/m(2) twice daily. RESULTS A total of 248 cycles of capecitabine were administered (median, five cycles; range, one to eight cycles). The overall response rate was 24% (95% CI, 15% to 41%), including two complete responses (CR; 4%) and 10 partial responses (PR; 20%). Disease control (CR + PR + stable disease) was achieved in 67% of patients. The median times to disease progression and overall survival were 7 months (95% CI, 6.4 to 9.5 months) and 11 months (95% CI, 8.6 to 13.3 months), respectively. Of the 35 patients evaluated for clinical benefit response, 14 (40%; 95% CI, 24% to 58%) showed clinical benefit. Capecitabine was well tolerated. Treatment-related grade 3 and 4 adverse events were observed in only six patients (12%), and the most common events were diarrhea, hand-foot syndrome, and thrombocytopenia. One patient (2%) had an episode of angina, but no treatment-related deaths were reported. CONCLUSION Our findings suggest that capecitabine is effective and well tolerated in elderly patients with advanced CRC who are considered ineligible for combination chemotherapy.
Cancers | 2014
Laura Muinelo-Romay; María Vieito; Alicia Abalo; Marta Alonso Nocelo; Francisco Baron; Urbano Anido; Elena Brozos; Francisca Vázquez; Santiago Aguín; Miguel Abal; Rafael López
In the present study we investigated the prognostic value of Circulating Tumour Cells (CTC) and their utility for therapy monitoring in non-small cell lung cancer (NSCLC). A total of 43 patients newly diagnosed with NSCLC were prospectively enrolled. Blood samples were obtained before the 1st, 2nd and 5th cycles of chemotherapy and analyzed using CellSearch technology. Both CTC and CTC-related objects (not morphological standard or broken epithelial cells) were counted. At baseline 18 (41.9%) patients were positive for intact CTC count and 10 (23.2%) of them had ≥5 CTC, while CK positive events were found in 79.1% of patients. The group of patients with CTC ≥5 at baseline presented worse PFS and OS than those with <5 CTC (p = 0.034 and p = 0.008, respectively). Additionally, high levels of total CK positive events were associated with poor prognosis in the group of patients with <5 CTC. Regarding therapy monitoring, patients presenting increased levels of CTC during the treatment demonstrated lower OS and PFS rates. All these data supported the value of CTC as a prognostic biomarker and as a surrogate indicator of chemotherapy effectiveness in advanced NSCLC patients, with the additional value of analyzing other “objects” such as apoptotic CTC or CK fragments to guide the clinical management of these patients.
Lancet Oncology | 2017
Antonio Llombart-Cussac; Javier Cortes; Laia Paré; Patricia Galván; Begoña Bermejo; Noelia Martínez; Maria Vidal; Sonia Pernas; Rafael López; Montserrat Muñoz; Paolo Nuciforo; Serafin Morales; Mafalda Oliveira; Lorena de la Peña; Alexandra Peláez; Aleix Prat
BACKGROUND HER2-positive breast cancer consists of four intrinsic molecular subtypes-luminal A, luminal B, HER2-enriched, and basal-like-and a normal-like subtype, with the HER2-enriched subtype having the highest activation of the EGFR-HER2 pathway. We aimed to test the hypothesis that patients with the HER2-enriched subtype benefit the most from dual HER2 blockade. METHODS PAMELA is an open-label, single-group, phase 2 trial done in 19 hospitals in Spain. We recruited female patients aged at least 18 years with previously untreated, centrally confirmed HER2-positive, stage I-IIIA invasive breast cancer regardless of hormone receptor status. Patients were given lapatinib (1000 mg per day orally) and trastuzumab (loading dose of 8 mg/kg, followed by 6 mg/kg every 3 weeks intravenously) for 18 weeks; hormone receptor-positive patients were additionally given letrozole (2·5 mg per day orally; if menopausal) or tamoxifen (20 mg per day orally; if premenopausal). Surgery was done 1-3 weeks after the last dose of study treatment. Intrinsic molecular subtypes of tumour biopsy samples taken at baseline (day 0) and day 14 were determined with the PAM50 predictor. The primary outcome was the ability of the HER2-enriched subtype to predict pathological complete response at the time of surgery. The primary outcome was assessed in the evaluable population (ie, all patients who had initial tumour biopsy samples available and who underwent definitive surgery) and safety was assessed in all patients who received at least one part of study treatment. This study is registered with ClinicalTrials.gov, number NCT01973660, and is completed. FINDINGS Between Oct 28, 2013, and Nov 26, 2015, we recruited 151 patients, of whom 14 (9%) discontinued treatment and 137 (91%) completed treatment as planned. At baseline, most patients had the HER2-enriched subtype (101 [67%]), followed by luminal A (22 [15%]), luminal B (16 [11%]), basal-like (nine [6%]), and normal-like (three [2%]) subtypes. At the time of surgery, 46 (30%, 95% CI 23-39) of 151 patients had pathological complete response in the breast. 41 (41%, 31-51) of 101 patients with the HER2-enriched subtype and five (10%, 4-23) of 50 patients with non-HER2-enriched subtypes achieved pathological complete response at the time of surgery (odds ratio 6·2, 95% CI 2·3-16·8; p=0·0004). INTERPRETATION The HER2-enriched subtype can identify patients with HER2-positive breast cancer who are likely to benefit from dual HER2 blockade therapies. FUNDING GlaxoSmithKline, Susan Komen Foundation, CERCA Programme-Generalitat de Catalunya, Banco Bilbao Vizcaya Argentaria Foundation, Pas a Pas, and the Breast Cancer Research Foundation.
Annals of Oncology | 2017
J Vidal; L Muinelo; Alba Dalmases; Frederick S. Jones; Daniel L. Edelstein; Mar Iglesias; M Orrillo; Alicia Abalo; Cecilia Lourdes Díaz Rodríguez; E Brozos; Y Vidal; Sonia Candamio; F Vázquez; Julio Chico Ruiz; M Guix; L Visa; Sikri; Joan Albanell; Beatriz Bellosillo; Rafael López; Clara Montagut
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.
Expert Review of Pharmacoeconomics & Outcomes Research | 2013
Emilio Alba; Eva Ciruelos; Rafael López; José Manuel López-Vega; Ana Lluch; Miguel Martín; Montserrat Muñoz; Pedro Sánchez-Rovira; Miguel Ángel Seguí; Marta Rubio Liria; Ferran Pérez-Alcántara
Aim: The COSTABRAX study evaluated the cost–effectiveness of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) versus polyethylated castor oil-based standard paclitaxel (sb-paclitaxel) in the treatment of patients with previously treated metastatic breast cancer in Spain. Materials & methods: Efficacy data were obtained from the CA012 trial (nab-paclitaxel administered every 3 weeks [q3w] and sb-paclitaxel q3w) and indirect comparison (sb-paclitaxel administered weekly), and were modeled to a time horizon of 5 years using a Markov model. The analysis was performed from the National Health Service perspective. Use of resources and key assumptions of the model were validated by a panel of 22 local oncologists. Results: Compared with sb-paclitaxel q3w, nab-paclitaxel q3w was cost effective, with a cost per life year gained of €11,088 and a cost per quality-adjusted life year of €17,808. Compared with sb-paclitaxel administered weekly, it showed savings of €711 per patient. Conclusion: The COSTABRAX study showed that nab-paclitaxel q3w is a cost-effective alternative compared with sb-paclitaxel q3w and a cost-saving alternative to sb-paclitaxel administered weekly in Spain.
Oncotarget | 2016
Anthony Kong; Daniel Rea; Samreen Ahmed; J. Thaddeus Beck; Rafael López; Laura Biganzoli; Anne C Armstrong; Massimo Aglietta; Emilio Alba; Mario Campone; Shu Fang Hsu Schmitz; Caroline Lefebvre; Mikhail Akimov; Soo-Chin Lee
This open-label, multicenter, phase 1B/2 trial assessed AUY922 plus trastuzumab in patients with locally advanced or metastatic HER2-positive breast cancer previously treated with chemotherapy and anti-HER2 therapy. This study was composed of a dose-escalation part with AUY922 administered weekly at escalating doses with trastuzumab 2 mg/kg/week (phase 1B), followed by a phase 2 part using the same regimen at recommended phase 2 dose (RP2D). The primary objectives were to determine the maximum tolerated dose (MTD) and/or RP2D (phase 1B), and to evaluate preliminary antitumor activity (phase 2) of AUY922 plus trastuzumab at MTD/RP2D. Forty-five patients were treated with AUY922 plus trastuzumab (4 in phase 1B with AUY922 at 55 mg/m2 and 41 in phase 1B/2 with AUY922 at 70 mg/m2 [7 in phase 1B and 34 in phase 2]). One patient in phase 1B (70 mg/m2) experienced a dose-limiting toxicity (grade 3 diarrhea); the RP2D was weekly AUY922 70 mg/m2 plus trastuzumab. Of the 41 patients in the 70 mg/m2 cohort, the overall response rate (complete or partial responses) was 22.0% and 48.8% patients had stable disease. Study treatment-related adverse events occurred in 97.8% of patients; of these, 31.1% were grade 3 or 4. Forty-one patients (91.1%) reported ocular events (82.3% had grade 1 or 2 events). Two patients (4.4%) had ocular events leading to the permanent discontinuation of study treatment. AUY922 at 70 mg/m2 plus trastuzumab standard therapy is well tolerated and active in patients with HER2-positive metastatic breast cancer who progressed on trastuzumab-based therapy.
Oncotarget | 2016
María Roel; Juan A. Rubiolo; Jorge Guerra-Varela; S. Silva; O. Thomas; Pablo Cabezas-Sainz; Laura Sánchez; Rafael López; Luis M. Botana
The marine environment constitutes an extraordinary resource for the discovery of new therapeutic agents. In the present manuscript we studied the effect of 3 different sponge derived guanidine alkaloids, crambescidine-816, -830, and -800. We show that these compounds strongly inhibit tumor cell proliferation by down-regulating cyclin-dependent kinases 2/6 and cyclins D/A expression while up-regulating the cell cyclin-dependent kinase inhibitors -2A, -2D and -1A. We also show that these guanidine compounds disrupt tumor cell adhesion and cytoskeletal integrity promoting the activation of the intrinsic apoptotic signaling, resulting in loss of mitochondrial membrane potential and concomitant caspase-3 cleavage and activation. The crambescidin 816 anti-tumor effect was fnally assayed in a zebrafish xenotransplantation model confirming its potent antitumor activity against colorectal carcinoma in vivo. Considering these results crambescidins could represent promising natural anticancer agents and therapeutic tools.
Journal of Clinical Oncology | 2004
Jaime Feliu; P. Escudero; F. Losa; M. Bolaños; J. M. Vicent; A. Yubero; J. Sanz-Lacalle; Rafael López; J. Dorta; Manuel González-Barón
3567 Background: X has superior efficacy and improved safety compared to bolus 5-FU/LV in 1st line MCRC and preserves its safety advantage in pts >65 in the adjuvant setting. We investigated its use in an elderly pt population. METHODS Objectives were to determine efficacy and safety of twice daily oral X as 1st line therapy in elderly pts with histologically confirmed MCRC. Pts were not candidates for combination therapy, aged >70 years with no prior chemotherapy (except adjuvant), measurable disease (RECIST), ECOG PS ≤2 and adequate bone marrow, renal and hepatic function. They received X 1250 mg/m2 twice daily D1-14 every 3 weeks (1000 mg/m2 for moderate renal impairment). RESULTS 51 pts were enrolled (M/F 31/20), median age 75 (71-90), ECOG PS 0-1 86%, PS 2 14%. No comorbidity in 82%, 78% mild dependence on help (Barthel Index) and most (M/F 61%/44%) were autonomous (Lawton Index). Tumor sites were colon (n=26), rectum (n=24) or both (n=1). Median number of metastatic sites was 1 (1 site 59%, ≥ 2 sites 35%): 67% in liver and 33% lung. Previous treatment included surgery (75%), chemotherapy (24%) and radiotherapy (16%). A total of 254 cycles (median 5, range 1-8) were administered with a median relative dose intensity of 89%. All pts were evaluated for toxicity. No grade 3/4 toxicity exceeded 6% pts: thrombocytopenia 4%, neutropenia 2%, diarrhea and hand-foot syndrome 6%, dyspnea, nausea, vomiting, epigastric pain, liver, renal, cardiac, anorexia and abdominal pain all 2% pts. There was no significant difference in grade 3/4 toxicities between the overall population and those >80 years old. Intent-to-treat efficacy analysis: 2 pts achieved CR, 12 PR, 19 SD, 10 PD and 8 NE: response rate 27% (95% CI 14-40). Median TTP and OS were 8 (95% CI 5-11) and 11 (95% CI 9-13) months, respectively. CONCLUSIONS This trial confirms that published data of 1st line X in the MCRC general population also apply to elderly pts. X is an effective and well tolerated 1st line treatment. No significant financial relationships to disclose.
Expert Review of Pharmacoeconomics & Outcomes Research | 2015
Alfredo Carrato; Pilar García; Rafael López; Teresa Macarulla; Fernando Rivera; J. Sastre; Joana Gostkorzewicz; Patricia Benedit; Ferran Pérez-Alcántara
The PANCOSTABRAX study evaluated the cost–effectiveness of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) in combination with gemcitabine (GEM) versus GEM alone in the treatment of patients with metastatic pancreatic cancer in Spain. Efficacy data were obtained from the MPACT study and were modeled to a lifetime horizon using a Markov model. The analysis was performed from the payer’s perspective. Use of resources and key assumptions of the analysis were validated by a panel of oncologists. The addition of nab-paclitaxel to GEM showed higher effectiveness results (0.156 additional quality adjusted life years) at a higher cost (€6,477), resulting in a cost per quality-adjusted life years gained of €41,519. The combination of nab-paclitaxel and GEM has been shown to be an effective and well-tolerated option for the treatment of metastatic pancreatic cancer and, in addition to becoming the new standard of care, could also be considered a cost-effective option.
Oncotarget | 2017
Luis León-Mateos; Helena Casas; Alicia Abalo; María Vieito; Manuel Abreu; Urbano Anido; Antonio Gómez-Tato; Rafael López; Miguel Abal; Laura Muinelo-Romay
Introduction There is a critical need of new surrogate markers for improving the therapeutic selection and monitoring of metastatic prostate cancer patients. Nowadays clinical management of these patients is been driven by biochemical and clinical parameters without enough accuracy to allow a real personalized medicine. The present study was conducted to go insight the molecular profile of circulating tumor cells (CTCs) isolated from advanced metastatic castration-resistant prostate cancer (mCRPC) with the aim of identifying prognostic marker with potential utility for therapy selection and monitoring. Materials and Methods CTCs isolation was carried out in peripheral blood samples from 29 mCRPC patients that undergo systemic chemotherapy based on taxanes (docetaxel/cabazitaxel) and 19 healthy controls using in parallel CellSearch and an alternative EpCAM-based immunoisolation followed by RT-qPCR analysis to characterize the CTC population. A panel of 17 genes related with prostate biology, hormone regulation, stem properties, tumor aggressiveness and taxanes responsiveness was analysed to identify an expression signature characterizing the CTCs. Results Patients with ≥ 5 CTCs/7.5ml of peripheral blood at baseline and during the treatment showed lower progression free survival (PFS) and overall survival (OS). Changes of CTCs levels during the treatment were also associated with the patients outcome. These results confirmed previous data obtained using CellSearch in mCRPC. In addition, we found a CTC profile mainly characterized by the expression of relevant genes for the hormone dependent regulation of PCa such as AR and CYP19 together with genes strongly implicated in PCa progression and resistance development such as BIRC5, TUB1A, GDF15, RAB7 and SPINK1. Our gene-expression profiling also permitted the identification of valuable prognostic biomarkers. Thus, high levels of AR, CYP19 and GDF15 were associated with poor PFS rates while AR, GDF15 and BIRC5 were also found as reliable predictors of OS. Besides, a logistic model using KLK3 and BIRC5 showed a high specificity and sensitivity compared to CellSearch to discriminate patients with a more aggressive evolution. Conclusions The molecular characterization of CTCs from advanced mCRPC patients provided with a panel of specific biomarkers, including genes related to taxanes resistance, with a promising applicability as “liquid biopsy” for the management of these patients.