Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sonia Pernas is active.

Publication


Featured researches published by Sonia Pernas.


Lancet Oncology | 2017

HER2-enriched subtype as a predictor of pathological complete response following trastuzumab and lapatinib without chemotherapy in early-stage HER2-positive breast cancer (PAMELA): an open-label, single-group, multicentre, phase 2 trial

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.


Stem cell reports | 2017

Resistance to Taxanes in Triple-Negative Breast Cancer Associates with the Dynamics of a CD49f+ Tumor-Initiating Population.

Jorge Gómez-Miragaya; Marta Palafox; Laia Paré; Guillermo Yoldi; Irene Ferrer; Sergi Vila; Patricia Galván; Pasquale Pellegrini; Héctor 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; Eva González-Suárez

Summary 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.


Oncotarget | 2015

Cancer stem cell-driven efficacy of trastuzumab (Herceptin): towards a reclassification of clinically HER2-positive breast carcinomas.

Begoña Martin-Castillo; Eugeni López-Bonet; Elisabet Cuyàs; Gemma Viñas; Sonia Pernas; Joan Dorca; Javier A. Menendez

Clinically HER2+ (cHER2+) breast cancer (BC) can no longer be considered a single BC disease entity in terms of trastuzumab responsiveness. Here we propose a framework for predicting the response of cHER2+ to trastuzumab that integrates the molecular distinctions of intrinsic BC subtypes with recent knowledge on cancer stem cell (CSC) biology. First, we consider that two interchangeable populations of epithelial-like, aldehyde dehydrogenase (ALDH)-expressing and mesenchymal-like, CD44+CD24−/low CSCs can be found in significantly different proportions across all intrinsic BC subtypes. Second, we overlap all the intrinsic subtypes across cHER2+ BC to obtain a continuum of mixed phenotypes in which one extreme exhibits a high identity with ALDH+ CSCs and the other extreme exhibits a high preponderance of CD44+CD24−/low CSCs. The differential enrichment of trastuzumab-responsive ALDH+ CSCs versus trastuzumab-refractory CD44+CD24−/low CSCs can explain both the clinical behavior and the primary efficacy of trastuzumab in each molecular subtype of cHER2+ (i.e., HER2-enriched/cHER2+, luminal A/cHER2+, luminal B/cHER2+, basal/cHER2+, and claudin-low/cHER2+). The intrinsic plasticity determining the epigenetic ability of cHER2+ tumors to switch between epithelial and mesenchymal CSC states will vary across the continuum of mixed phenotypes, thus dictating their intratumoral heterogeneity and, hence, their evolutionary response to trastuzumab. Because CD44+CD24−/low mesenchymal-like CSCs distinctively possess a highly endocytic activity, the otherwise irrelevant HER2 can open the door to a type of “Trojan horse” approach by employing antibody-drug conjugates such as T-DM1, which will allow a rapid and CSC-targeted delivery of cytotoxic drugs to therapeutically manage trastuzumab-unresponsive basal/cHER2+ BC. Contrary to the current dichotomous model used clinically, our model proposes that a reclassification of cHER2+ tumors based on the spectrum of molecular BC subtypes might inform on their CSC-determined sensitivity to trastuzumab, thus providing a better delineation of the predictive value of cHER2+ in BC by incorporating CSCs-driven intra-tumor heterogeneity into clinical decisions.


PLOS ONE | 2015

Lymphangioleiomyomatosis Biomarkers Linked to Lung Metastatic Potential and Cell Stemness.

Gorka Ruiz de Garibay; Carmen Herranz; Alicia Llorente; Jacopo Boni; Jordi Serra-Musach; Francesca Mateo; Helena Aguilar; Laia Gómez-Baldó; Anna Petit; August Vidal; Fina Climent; Javier Hernández-Losa; Alex Cordero; Eva González-Suárez; Jose V. Sanchez-Mut; Manel Esteller; Roger Llatjos; Mar Varela; José I. López; Nadia García; Ana I. Extremera; Anna Gumà; Raul Ortega; Maria Pla; Adela Fernandez; Sonia Pernas; Catalina Falo; Idoia Morilla; Miriam Campos; Miguel Gil

Lymphangioleiomyomatosis (LAM) is a rare lung-metastasizing neoplasm caused by the proliferation of smooth muscle-like cells that commonly carry loss-of-function mutations in either the tuberous sclerosis complex 1 or 2 (TSC1 or TSC2) genes. While allosteric inhibition of the mechanistic target of rapamycin (mTOR) has shown substantial clinical benefit, complementary therapies are required to improve response and/or to treat specific patients. However, there is a lack of LAM biomarkers that could potentially be used to monitor the disease and to develop other targeted therapies. We hypothesized that the mediators of cancer metastasis to lung, particularly in breast cancer, also play a relevant role in LAM. Analyses across independent breast cancer datasets revealed associations between low TSC1/2 expression, altered mTOR complex 1 (mTORC1) pathway signaling, and metastasis to lung. Subsequently, immunohistochemical analyses of 23 LAM lesions revealed positivity in all cases for the lung metastasis mediators fascin 1 (FSCN1) and inhibitor of DNA binding 1 (ID1). Moreover, assessment of breast cancer stem or luminal progenitor cell biomarkers showed positivity in most LAM tissue for the aldehyde dehydrogenase 1 (ALDH1), integrin-ß3 (ITGB3/CD61), and/or the sex-determining region Y-box 9 (SOX9) proteins. The immunohistochemical analyses also provided evidence of heterogeneity between and within LAM cases. The analysis of Tsc2-deficient cells revealed relative over-expression of FSCN1 and ID1; however, Tsc2-deficient cells did not show higher sensitivity to ID1-based cancer inhibitors. Collectively, the results of this study reveal novel LAM biomarkers linked to breast cancer metastasis to lung and to cell stemness, which in turn might guide the assessment of additional or complementary therapeutic opportunities for LAM.


Oncotarget | 2016

Gasdermin B expression predicts poor clinical outcome in HER2-positive breast cancer.

Marta Hergueta-Redondo; David Sarrió; Ángela Molina-Crespo; Rocio Vicario; Cristina Bernadó-Morales; Lidia Martínez; Alejandro Rojo-Sebastián; Jordi Serra-Musach; Alba Mota; Ángel Martínez-Ramírez; María Ángeles Castilla; Antonio Gonzalez-Martin; Sonia Pernas; Amparo Cano; Javier Cortes; Paolo Nuciforo; Vicente Peg; José Palacios; Miguel Angel Pujana; J. Arribas; Gema Moreno-Bueno

Around, 30–40% of HER2-positive breast cancers do not show substantial clinical benefit from the targeted therapy and, thus, the mechanisms underlying resistance remain partially unknown. Interestingly, ERBB2 is frequently co-amplified and co-expressed with neighbour genes that may play a relevant role in this cancer subtype. Here, using an in silico analysis of data from 2,096 breast tumours, we reveal a significant correlation between Gasdermin B (GSDMB) gene (located 175 kilo bases distal from ERBB2) expression and the pathological and clinical parameters of poor prognosis in HER2-positive breast cancer. Next, the analysis of three independent cohorts (totalizing 286 tumours) showed that approximately 65% of the HER2-positive cases have GSDMB gene amplification and protein over-expression. Moreover, GSDMB expression was also linked to poor therapeutic responses in terms of lower relapse free survival and pathologic complete response as well as positive lymph node status and the development of distant metastasis under neoadjuvant and adjuvant treatment settings, respectively. Importantly, GSDMB expression promotes survival to trastuzumab in different HER2-positive breast carcinoma cells, and is associated with trastuzumab resistance phenotype in vivo in Patient Derived Xenografts. In summary, our data identifies the ERBB2 co-amplified and co-expressed gene GSDMB as a critical determinant of poor prognosis and therapeutic response in HER2-positive breast cancer.


Lancet Oncology | 2018

Balixafortide plus eribulin in HER2-negative metastatic breast cancer: a phase 1, single-arm, dose-escalation trial

Sonia Pernas; Miguel Martin; Peter A. Kaufman; Marta Gil-Martin; Patricia Gomez Pardo; Sara López-Tarruella; Luis Manso; Eva Ciruelos; Jose Alejandro Perez-Fidalgo; Cristina Hernando; Foluso O. Ademuyiwa; Katherine N. Weilbaecher; Ingrid A. Mayer; Timothy J. Pluard; Maria Martinez Garcia; Linda T. Vahdat; Jose Perez-Garcia; Achim Wach; Debra Barker; Samson Fung; Barbara Romagnoli; Javier Cortes

BACKGROUND The C-X-C chemokine receptor type 4 (CXCR4)-stromal cell-derived factor-1α (SDF-1α) axis regulates function and trafficking of immune cells and the tumour microenvironment. CXCR4 antagonists have been shown to enhance the activity of different anticancer treatments in preclinical models. We assessed the safety, tolerability, pharmacokinetics, and preliminary phase 1 activity of the CXCR4 antagonist, balixafortide, in combination with eribulin chemotherapy in patients with heavily pretreated, relapsed metastatic breast cancer. METHODS This single-arm, dose-escalation, phase 1 trial enrolled patients at 11 sites in Spain and the USA. Eligible patients were women aged 18 years or older who had histologically confirmed HER2-negative metastatic breast cancer, evidence of tumour cell CXCR4 expression, an Eastern Cooperative Oncology Group performance status of 0 or 1, and who had previously received between one and three chemotherapy regimens for metastatic breast cancer, and at least one endocrine therapy if they had hormone receptor-positive disease, unless they were considered unsuitable for endocrine therapy. A standard 3+3 dose-escalation design was used, followed by an expanded cohort at the established maximum tolerated dose or highest dose if no dose-limiting toxicity was observed for the combination. After a treatment-related fatal adverse event in the first cohort who received 21-day cycles of treatment with eribulin and balixafortide, a protocol amendment modified the study design to be done in two parts. Patients enrolled to part 1 received an initial 28-day run-in cycle, with some cohorts receiving de-escalated doses of eribulin plus balixafortide to assess the safety and pharmacokinetics of the combination. The evaluation of part 1 did not confirm any dose-limiting toxicities or eribulin-balixafortide interactions, and therefore part 2 started enrolling patients to receive eribulin at the originally planned dose of 1·4 mg/m2 on days 2 and 9 of a 21-day cycle and balixafortide from a starting dose of 2 mg/kg with dose increments of 0·5 or 1 mg/kg on days 1-3 and 8-10 of the 21-day cycle. Both drugs were administered as intravenous infusions. All patients were to receive treatment until disease progression or unacceptable toxicity. The primary endpoints were dose-limiting toxicities and adverse events, and the establishment of a maximum tolerated dose or recommended phase 2 dose, and pharmacokinetic parameters. Safety analysis was done in all patients who received at least one dose of study treatment. Analysis of antitumour activity was done in all patients who received at least one full cycle of study treatment. The trial is registered at ClinicalTrials.gov, number NCT01837095, and is closed to accrual. FINDINGS Between Jan 28, 2014, and Oct 4, 2016, 56 patients were enrolled into the trial. No dose-limiting toxicities were confirmed and the maximum tolerated dose was not reached. The highest dose was established as eribulin 1·4 mg/m2 on days 2 and 9, and balixafortide 5·5 mg/kg on days 1-3 and 8-10 of the 21-day cycle. Objective responses (all partial responses) were observed in 16 (30%; 95% CI 18-44) of 54 patients who were evaluable for antitumour activity. The most common treatment-emergent adverse events of any grade were fatigue (44 [79%] of 56 patients), neutropenia (32 [57%]), infusion-related reactions (27 [48%]), alopecia (26 [46%]), constipation (26 [46%]), and nausea (25 [45%]). Serious adverse events occurred in 21 (38%) of 56 patients, including febrile neutropenia in five (9%) of 56 patients, neutrophil count decrease in two (4%) patients, constipation in two (4%) patients, pneumonia in two (4%) patients, and urinary tract infection in three (5%) patients. Two (4%) of 56 patients died while receiving study treatment; one from septic shock and one from pneumonia. INTERPRETATION The safety and tolerability of balixafortide plus eribulin seems to be similar to that of eribulin or balixafortide monotherapy, and the preliminary activity of the combination seems promising in patients with HER-negative metastatic breast cancer. The results suggest that balixafortide plus eribulin has potential to provide a new therapeutic option in heavily pretreated patients with metastatic breast cancer and warrants further investigation in randomised trials. FUNDING Polyphor.


Journal of The Peripheral Nervous System | 2016

Serum micronutrients and prealbumin during development and recovery of chemotherapy-induced peripheral neuropathy.

Roser Velasco; Cristina Santos; Gemma Soler; Miguel Gil-Gil; Sonia Pernas; Maica Galan; Ramon Palmero; Jordi Bruna

Chemotherapy‐induced peripheral neuropathy (CIPN) is a frequent adverse event. Nutritional status can become impaired in cancer patients, potentially contributing to neuropathys evolution. Our aim was to evaluate serum micronutrients and prealbumin in a cohort of 113 solid‐cancer patients receiving platinum and taxane compounds during the development and recovery of neuropathy, up to 1 year after finishing treatment. CIPN was graded according to Total Neuropathy Score© and NCI.CTCv3 at T0 (baseline), T1 (1–3 months), and T12 (12 months) after chemotherapy. CIPN was classified as asymptomatic (< grade 2) or symptomatic (≥2). CIPN recovery was defined as ≥1 grade improvement at T12. Symptomatic CIPN developed in 52% of patients. Symptomatic patients presented a higher increase in TNSc (p < 0.001), in TNSr© (p < 0.001), and decrease in sural (p < 0.001) and radial nerve conduction (p < 0.001). No significant differences with any of the micronutrients were observed along T0‐T1 period between severity or chemotherapy groups. By T12, symptomatic patients without recovery had a decrease in vitamin E levels (p = 0.019) and prealbumin (p = 0.062) compared with those symptomatic that improved. A correlation between the variation of vitamin E and prealbumin at T0‐T1 (r = 0.626, p = 0.001) and T1‐T12 (r = 0.411, p = 0.06) was observed. After chemotherapy treatment, the improvement of patients displaying symptomatic neuropathy is related to vitamin E and prealbumin serum levels. Our results suggest that nutritional status can play a role in CIPN recovery.


Clinical Breast Cancer | 2017

The Shift From Sentinel Lymph Node Biopsy Performed Either Before or After Neoadjuvant Systemic Therapy in the Clinical Negative Nodes of Breast Cancer Patients. Results, and the Advantages and Disadvantages of Both Procedures

Sergi Fernandez-Gonzalez; Catalina Falo; Maria Pla; Sonia Pernas; Maite Bajen; Teresa Soler; Raul Ortega; Cecilia Quetglas; Xavier Perez-Martin; Maria Eulalia Fernandez Montoli; Miriam Campos; Mar Varela-Rodriguez; Jordi Ponce; Amparo García-Tejedor

Micro‐Abstract The clinical consequence of performing sentinel lymph node biopsy (SLNB) before or after neoadjuvant therapy (NAT) in breast cancer patients remains under study. Therefore, we present a comparison between both approaches. The lymphadenectomy rate is reduced by threefold when the SLNB is assessed after NAT with no increase in early recurrences; consequently, it seems that this approach has more advantages for the patients than before NAT. Background: In patients with breast cancer who are candidates for neoadjuvant therapy (NAT), the timing of when to perform sentinel lymph node biopsy (SLNB) remains under discussion. The aim of this study was to compare the advantages and disadvantages of SLNB performed before and after NAT. Patients and Methods: One hundred seventy‐two patients, T1c to T3 and N0 (clinically and according to ultrasound) candidates for NAT were included. We compared the outcomes of 2 groups: (1) 122 patients of whom SLNB was performed before NAT (pre‐NAT) from December 2006 to April 2014; and (2) 50 patients with SLNB performed after NAT (post‐NAT) from May 2014 to July 2016. Results: Both groups were homogeneous in baseline patient characteristics. The SLNB was positive in 50 patients [41.7%] (33 macrometastases [66%] and 17 micrometastases [34%]) versus 6 patients [12%] (5 macrometastases [83.3%] and 1 micrometastases [16.7%]) in pre‐ and post‐NAT groups, respectively. The lymphadenectomy was performed in 34 patients [28.3%] versus 4 patients [8%], with an odds ratio of 3.48 (95% confidence interval, 1.3‐9.3). The recurrences in the pre‐NAT group after a median follow‐up of 62 months were 12 systemic, 2 local and systemic, and none axillary. In the post‐NAT group were no recurrences after a median follow‐up of 16 months. Finally, SLNB after NAT reduces the delay in starting NAT from 24 to 14 days (medians; P < .001) and the identification of the SLNB was in 122 patients [100%] versus 49 patients [98%]. Conclusion: SLNB performed after NAT significantly reduces the rate of lymphadenectomies without any increase in recurrences at early follow‐up. Furthermore, it allows systemic treatment to be started earlier without interfering in the SLNB identification rate.


Annals of Oncology | 2018

A predictive model of pathologic response based on tumor cellularity and tumor-infiltrating lymphocytes (CelTIL) in HER2-positive breast cancer treated with chemo-free dual HER2 blockade

Paolo Nuciforo; T Pascual; Javier Cortes; Antonio Llombart-Cussac; Roberta Fasani; Laia Paré; M.M. Oliveira; Patricia Galván; Noelia Martínez; Begoña Bermejo; Marcela A. Vidal; Sonia Pernas; Ramon Lopez; Montserrat Muñoz; I Garau; Luis Manso; J Alarcón; E Martínez; V Rodrik-Outmezguine; Jan C. Brase; P Villagrasa; Aleix Prat; E Holgado

Background The presence of stromal tumor-infiltrating lymphocytes (TILs) is associated with increased pathologic complete response (pCR) and improved outcomes in HER2-positive early-breast cancer (BC) treated with anti-HER2-based chemotherapy. In the absence of chemotherapy, the association of TILs with pCR following anti-HER2 therapy-only is largely unknown. Patients and methods The PAMELA neoadjuvant trial treated 151 women with HER2-positive BC with lapatinib and trastuzumab [and hormonal therapy if hormone receptor (HR)-positive] for 18 weeks. Percentage of TILs and tumor cellularity were determined at baseline (N = 148) and at day 15 (D15) of treatment (N = 134). Associations of TILs and tumor cellularity with pCR in the breast were evaluated. A combined score based on tumor cellularity and TILs (CelTIL) measured at D15 was derived in PAMELA, and validated in D15 samples from 65 patients with HER2-positive disease recruited in the LPT109096 neoadjuvant trial, where anti-HER2 therapy-only was administer for 2 weeks, then standard chemotherapy was added for 24 weeks. Results In PAMELA, baseline and D15 TILs were significantly associated with pCR in univariate analysis. In multivariable analysis, D15 TILs, but not baseline TILs, were significantly associated with pCR. At D15, TILs and tumor cellularity were found independently associated with pCR. A combined score (CelTIL) taking into account both variables was derived. CelTIL at D15 as a continuous variable was significantly associated with pCR, and patients with CelTIL-low and CelTIL-high scores had a pCR rate of 0% and 33%, respectively. In LPT109096, CelTIL at D15 was found associated with pCR both as a continuous variable and as group categories using a pre-defined cut-off (75.0% versus 33.3%). Conclusions On-treatment TILs, but not baseline TILs, are independently associated with response following anti-HER2 therapy-only. A combined score of TILs and tumor cellularity measured at D15 provides independent predictive information upon completion of neoadjuvant anti-HER2-based therapy. Clinical trial number NCT01973660.


Cancer Research | 2017

Abstract S3-03: PAM50 intrinsic subtype as a predictor of pathological complete response following neoadjuvant dual HER2 blockade without chemotherapy in HER2-positive breast cancer: First results of the PAMELA clinical trial

A Prat Aparicio; J. Cortes Castan; Laia Paré; Patricia Galván; Begoña Bermejo; Noelia Martínez; María N. Vidal; Sonia Pernas; Rafael López; Montserrat Muñoz; Paolo Nuciforo; Roberta Fasani; Serafin Morales; Mafalda Oliveira; L de la Pena; A Peláez; Antonio Llombart

Background: Prior neoadjuvant studies in HER2+ breast cancer have shown that dual HER2 blockade without chemotherapy achieves pathological complete responses (pCR) rates of 6-36% (TBCRC006/TBCRC023/NeoSphere). However, a major challenge today is how to select prospectively patients who will derive the maximum benefit from dual anti-HER2 therapies without chemotherapy. In this context, we and others have previously shown that HER2+ disease is biologically heterogeneous and composed of all the intrinsic molecular subtypes (Luminal A, Luminal B, HER2-enriched [HER2-E] and Basal-like). Among them, the HER2-E subtype shows the highest activation of the EGFR/HER2 pathway. Methods: PAMELA (NCT01973660) is a non-randomized, open-label, multicentric, prospective translational research study in stage I-IIIA HER2+ breast cancer designed to evaluate the ability of the PAM50 intrinsic subtypes to predict pCR in the breast (pCRB; in situ allowed) following 18 weeks of neoadjuvant lapatinib and trastuzumab). Patients with HR+ disease received letrozole (if postmenopausal) or tamoxifen (if pre-menopausal). The primary objective was to compare the pCRB rates of the HER2-E versus the non-HER2-E subtypes in the intent-to-treat population. The study was planned with a power of 95% at a significance level of 0.05 to detect an absolute relative difference in pCRB rates between the two groups of 27% (i.e. 35% in HER2E and 8% in non-HER2-E). Day-15 formalin-fixed, paraffin-embedded tissue samples were prospectively collected and gene expression profiled using the nCounter platform. The intrinsic subtypes were identified using the research-based PAM50 predictor (Parker JCO 2009). Results: A total of 151 patients were recruited (n=77 HR+ and n=74 HR-). Patient characteristics were: mean age (55 years), mean tumor size (2.84 cm), negative axilla (63.5%) and postmenopausal (60.2%). At baseline, intrinsic subtype distribution was: HER2-E (n=101, 66.9%), Luminal A (n=22; 14.6%), Luminal B (n=16; 10.6%), Basal-like (n=9; 6%) and Normal-Like (n=3; 2%). The overall pCRB was 30.5% (46/151), 18.2% in HR+ disease and 43.2% in HR- disease. Five patients (3.3%) presented progressive disease. Rates of pCRB in HER2-E and non-HER2-E subtypes were 40.6% and 10.0% (p Conclusions: The PAMELA trial met its primary endpoint. PAM50 HER2-E subtype identifies patients with HER2+ disease likely to derive a large benefit from dual anti-HER2 therapies +/- endocrine therapy, especially in HER2+/HR+ disease. In addition, early changes in gene expression indicative of a reduction of tumor cellularity are predictive of pathological complete response at surgery. Citation Format: Prat Aparicio A, Cortes Castan J, Pare L, Galvan P, Bermejo B, Martinez N, Vidal M, Pernas S, Lopez R, Munoz M, Nuciforo P, Fasani R, Morales S, Oliveira M, de La Pena L, Pelaez A, Llombart A. PAM50 intrinsic subtype as a predictor of pathological complete response following neoadjuvant dual HER2 blockade without chemotherapy in HER2-positive breast cancer: First results of the PAMELA clinical trial [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr S3-03.

Collaboration


Dive into the Sonia Pernas's collaboration.

Top Co-Authors

Avatar

Anna Petit

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Aleix Prat

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Pla

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Noelia Martínez

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Antonio Llombart

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eugeni López-Bonet

Autonomous University of Barcelona

View shared research outputs
Researchain Logo
Decentralizing Knowledge