Network


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

Hotspot


Dive into the research topics where Marta Palafox is active.

Publication


Featured researches published by Marta Palafox.


Cancer Research | 2012

RANK Induces Epithelial–Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis

Marta Palafox; Irene Ferrer; Pasquale Pellegrini; Sergi Vila; Sara Hernandez-Ortega; Ander Urruticoechea; Fina Climent; María Teresa Soler; Purificación Muñoz; Francesc Viñals; Mark E. Tometsko; Dan Branstetter; William C. Dougall; Eva González-Suárez

Paracrine signaling through receptor activator of NF-κB (RANK) pathway mediates the expansion of mammary epithelia that occurs during pregnancy, and activation of RANK pathway promotes mammary tumorigenesis in mice. In this study we extend these previous data to human cells and show that the RANK pathway promotes the development of mammary stem cells and breast cancer. Overexpression of RANK (FL-RANK) in a panel of tumoral and normal human mammary cells induces the expression of breast cancer stem and basal/stem cell markers. High levels of RANK in untransformed MCF10A cells induce changes associated with both stemness and transformation, including mammary gland reconstitution, epithelial-mesenchymal transition (EMT), increased migration, and anchorage-independent growth. In addition, spheroids of RANK overexpressing MCF10A cells display disrupted acinar formation, impair growth arrest and polarization, and luminal filling. RANK overexpression in tumor cells with nonfunctional BRCA1 enhances invasiveness in acinar cultures and increases tumorigenesis and metastasis in immunodeficient mice. High levels of RANK were found in human primary breast adenocarcinomas that lack expression of the hormone receptors, estrogen and progesterone, and in tumors with high pathologic grade and proliferation index; high RANK/RANKL expression was significantly associated with metastatic tumors. Together, our findings show that RANK promotes tumor initiation, progression, and metastasis in human mammary epithelial cells by increasing the population of CD44(+)CD24(-) cells, inducing stemness and EMT. These results suggest that RANK expression in primary breast cancer associates with poor prognosis.


Cancer Research | 2016

Early Adaptation and Acquired Resistance to CDK4/6 Inhibition in Estrogen Receptor-Positive Breast Cancer

Maria Teresa Herrera-Abreu; Marta Palafox; Uzma Asghar; Martín A. Rivas; Rosalind J. Cutts; Isaac Garcia-Murillas; Alex Pearson; Marta Guzman; Olga Rodriguez; Judit Grueso; Meritxell Bellet; Javier Cortes; Richard Elliott; Sunil Pancholi; José Baselga; Mitch Dowsett; Lesley-Ann Martin; Nicholas C. Turner; Violeta Serra

Small-molecule inhibitors of the CDK4/6 cell-cycle kinases have shown clinical efficacy in estrogen receptor (ER)-positive metastatic breast cancer, although their cytostatic effects are limited by primary and acquired resistance. Here we report that ER-positive breast cancer cells can adapt quickly to CDK4/6 inhibition and evade cytostasis, in part, via noncanonical cyclin D1-CDK2-mediated S-phase entry. This adaptation was prevented by cotreatment with hormone therapies or PI3K inhibitors, which reduced the levels of cyclin D1 (CCND1) and other G1-S cyclins, abolished pRb phosphorylation, and inhibited activation of S-phase transcriptional programs. Combined targeting of both CDK4/6 and PI3K triggered cancer cell apoptosis in vitro and in patient-derived tumor xenograft (PDX) models, resulting in tumor regression and improved disease control. Furthermore, a triple combination of endocrine therapy, CDK4/6, and PI3K inhibition was more effective than paired combinations, provoking rapid tumor regressions in a PDX model. Mechanistic investigations showed that acquired resistance to CDK4/6 inhibition resulted from bypass of cyclin D1-CDK4/6 dependency through selection of CCNE1 amplification or RB1 loss. Notably, although PI3K inhibitors could prevent resistance to CDK4/6 inhibitors, they failed to resensitize cells once resistance had been acquired. However, we found that cells acquiring resistance to CDK4/6 inhibitors due to CCNE1 amplification could be resensitized by targeting CDK2. Overall, our results illustrate convergent mechanisms of early adaptation and acquired resistance to CDK4/6 inhibitors that enable alternate means of S-phase entry, highlighting strategies to prevent the acquisition of therapeutic resistance to these agents. Cancer Res; 76(8); 2301-13. ©2016 AACR.


PLOS ONE | 2015

Dual Fatty Acid Synthase and HER2 Signaling Blockade Shows Marked Antitumor Activity against Breast Cancer Models Resistant to Anti-HER2 Drugs

Adriana Blancafort; Ariadna Giró-Perafita; Glòria Oliveras; Sònia Palomeras; Carlos Turrado; Oscar Campuzano; Dolors Carrión-Salip; Anna Massaguer; Ramon Brugada; Marta Palafox; Jorge Gómez-Miragaya; Eva González-Suárez; Teresa Puig

Blocking the enzyme Fatty Acid Synthase (FASN) leads to apoptosis of HER2-positive breast carcinoma cells. The hypothesis is that blocking FASN, in combination with anti-HER2 signaling agents, would be an effective antitumor strategy in preclinical HER2+ breast cancer models of trastuzumab and lapatinib resistance. We developed and molecularly characterized in vitro HER2+ models of resistance to trastuzumab (SKTR), lapatinib (SKLR) and both (SKLTR). The cellular interactions of combining anti-FASN polyphenolic compounds (EGCG and the synthetic G28UCM) with anti-HER2 signaling drugs (trastuzumab plus pertuzumab and temsirolimus) were analyzed. Tumor growth inhibition after treatment with EGCG, pertuzumab, temsirolimus or the combination was evaluated in two in vivo orthoxenopatients: one derived from a HER2+ patient and another from a patient who relapsed on trastuzumab and lapatinib-based therapy. SKTR, SKLR and SKLTR showed hyperactivation of EGFR and p-ERK1/2 and PI3KCA mutations. Dual-resistant cells (SKLTR) also showed hyperactivation of HER4 and recovered levels of p-AKT compared with mono-resistant cells. mTOR, p-mTOR and FASN expression remained stable in SKTR, SKLR and SKLTR. In vitro, anti-FASN compounds plus pertuzumab showed synergistic interactions in lapatinib- and dual- resistant cells and improved the results of pertuzumab plus trastuzumab co-treatment. FASN inhibitors combined with temsirolimus displayed the strongest synergistic interactions in resistant cells. In vivo, both orthoxenopatients showed strong response to the antitumor activity of the combination of EGCG with pertuzumab or temsirolimus, without signs of toxicity. We showed that the simultaneous blockade of FASN and HER2 pathways is effective in cells and in breast cancer models refractory to anti-HER2 therapies.


Nature Cell Biology | 2018

MSK1 regulates luminal cell differentiation and metastatic dormancy in ER + breast cancer

Sylwia Gawrzak; Lorenzo Rinaldi; Sara Gregorio; Enrique J. Arenas; Fernando Salvador; Jelena Urosevic; Cristina Figueras-Puig; Federico Rojo; Ivan del Barco Barrantes; Juan Miguel Cejalvo; Marta Palafox; Marc Guiu; Antonio Berenguer-Llergo; Aikaterini Symeonidi; Anna Bellmunt; Daniela Kalafatovic; Anna Arnal-Estapé; Esther Fernández; Barbara Müllauer; Rianne Groeneveld; Konstantin Slobodnyuk; Camille Stephan-Otto Attolini; Cristina Saura; J. Arribas; Javier Cortes; Ana Rovira; Montse Muñoz; Ana Lluch; Violeta Serra; Joan Albanell

For many patients with breast cancer, symptomatic bone metastases appear after years of latency. How micrometastatic lesions remain dormant and undetectable before initiating colonization is unclear. Here, we describe a mechanism involved in bone metastatic latency of oestrogen receptor-positive (ER+) breast cancer. Using an in vivo genome-wide short hairpin RNA screening, we identified the kinase MSK1 as an important regulator of metastatic dormancy in breast cancer. In patients with ER+ breast cancer, low MSK1 expression associates with early metastasis. We show that MSK1 downregulation impairs the differentiation of breast cancer cells, increasing their bone homing and growth capacities. MSK1 controls the expression of genes required for luminal cell differentiation, including the GATA3 and FOXA1 transcription factors, by modulating their promoter chromatin status. Our results indicate that MSK1 prevents metastatic progression of ER+ breast cancer, suggesting that stratifying patients with breast cancer as high or low risk for early relapse based on MSK1 expression could improve prognosis.Gawrzak et al. show that MSK1 regulates bone metastatic dormancy of ER+ breast cancer. MSK1 affects histone modifications at luminal transcription factor promoters to prevent cell differentiation and bone homing.


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.


Journal of Experimental Medicine | 2018

Loss of USP28-mediated BRAF degradation drives resistance to RAF cancer therapies

Azad Saei; Marta Palafox; Touati Benoukraf; Nishi Kumari; Patrick Jaynes; Prasanna Vasudevan Iyengar; Eva Muñoz-Couselo; Paolo Nuciforo; Javier Cortes; Christopher Nötzel; Nesaretnam Barr Kumarakulasinghe; John Lalith Charles Richard; Zul Fazreen Bin Adam Isa; Brendan Pang; Marta Guzman; Zhou Siqin; Henry Yang; Wai Leong Tam; Violeta Serra; Pieter J.A. Eichhorn

RAF kinase inhibitors are clinically active in patients with BRAF (V600E) mutant melanoma. However, rarely do tumors regress completely, with the majority of responses being short-lived. This is partially mediated through the loss of negative feedback loops after MAPK inhibition and reactivation of upstream signaling. Here, we demonstrate that the deubiquitinating enzyme USP28 functions through a feedback loop to destabilize RAF family members. Loss of USP28 stabilizes BRAF enhancing downstream MAPK activation and promotes resistance to RAF inhibitor therapy in culture and in vivo models. Importantly, we demonstrate that USP28 is deleted in a proportion of melanoma patients and may act as a biomarker for response to BRAF inhibitor therapy in patients. Furthermore, we identify Rigosertib as a possible therapeutic strategy for USP28-depleted tumors. Our results show that loss of USP28 enhances MAPK activity through the stabilization of RAF family members and is a key factor in BRAF inhibitor resistance.


Nature Communications | 2017

Modulation of telomere protection by the PI3K/AKT pathway

Marinela Méndez-Pertuz; Paula Martínez; Carmen Blanco-Aparicio; Elena Gomez-Casero; Ana Belén García; Jorge Luis Martínez-Torrecuadrada; Marta Palafox; Javier Cortes; Violeta Serra; Joaquín Pastor; Maria A. Blasco

Telomeres and the insulin/PI3K pathway are considered hallmarks of aging and cancer. Here, we describe a role for PI3K/AKT in the regulation of TRF1, an essential component of the shelterin complex. PI3K and AKT chemical inhibitors reduce TRF1 telomeric foci and lead to increased telomeric DNA damage and fragility. We identify the PI3Kα isoform as responsible for this TRF1 inhibition. TRF1 is phosphorylated at different residues by AKT and these modifications regulate TRF1 protein stability and TRF1 binding to telomeric DNA in vitro and are important for in vivo TRF1 telomere location and cell viability. Patient-derived breast cancer PDX mouse models that effectively respond to a PI3Kα specific inhibitor, BYL719, show decreased TRF1 levels and increased DNA damage. These findings functionally connect two of the major pathways for cancer and aging, telomeres and the PI3K pathway, and pinpoint PI3K and AKT as novel targets for chemical modulation of telomere protection.Regulation of telomeres and the insulin/PI3K pathway both have roles in aging and cancer development but have not been functionally linked. Here the authors demonstrate that PI3K, via downstream targets, regulates TRF1 via phosphorylation.


Nature Cell Biology | 2018

Publisher Correction: MSK1 regulates luminal cell differentiation and metastatic dormancy in ER + breast cancer

Sylwia Gawrzak; Lorenzo Rinaldi; Sara Gregorio; Enrique J. Arenas; Fernando Salvador; Jelena Urosevic; Cristina Figueras-Puig; Federico Rojo; Ivan del Barco Barrantes; Juan Miguel Cejalvo; Marta Palafox; Marc Guiu; Antonio Berenguer-Llergo; Aikaterini Symeonidi; Anna Bellmunt; Daniela Kalafatovic; Anna Arnal-Estapé; Esther Fernández; Barbara Müllauer; Rianne Groeneveld; Konstantin Slobodnyuk; Camille Stephan-Otto Attolini; Cristina Saura; Joaquín Arribas; Javier Cortes; Ana Rovira; Montse Muñoz; Ana Lluch; Violeta Serra; Joan Albanell

In the version of this Article originally published, the boxes framing the two plots in Fig. 1g were misaligned from the axes due to a technical error. This has now been corrected in all versions of the Article.


Cancer Research | 2016

Abstract 2825: Identification of CDK4/6-response biomarkers using estrogen receptor-positive breast cancer patient-derived xenografts (PDX)

Violeta Serra; Marta Palafox; Maria-Teresa Herrera; Martín A. Rivas; Marta Guzman; Olga Rodriguez; Judit Grueso; Meritxell Bellet; Mafalda Oliveira; Cristina Saura; Emmanuelle di Tomaso; Giordi Camponigro; Nicholas C. Turner; Javier Cortes; José Baselga

Endocrine resistance is a clinical challenge for the treatment of estrogen receptor positive (ER+) breast cancer (BC). CDK4/6 blockade in combination with endocrine therapy has shown clinical activity in metastatic ER+ BC refractory to anti-hormonal treatment. However, there is a need for biomarkers that can predict the response to this treatment and improve patient stratification. We aimed to address this issue using xenograft models established from samples of ER+ BC patients. Six ER+ PDXs were treated with continuous doses of a CDK4/6 inhibitor (LEE011, 75mg/kg, 6IW) and a PI3K-alpha inhibitor (BYL719, 35mg/kg, 6IW) as single agents and in combination, and intrinsic sensitivity to these agents was evaluated. The models were then genomically characterized using a capture-based sequencing panel and by digital PCR. One PDX model was intrinsically sensitive to single-agent CDK4/6 inhibition and experienced tumor regression, but all individual tumors eventually escaped therapy after 50 days of treatment. This particular model harbored an ESR1-mutation and concomitant losses of CDKN2A/B. At relapse, we identified the acquisition of an RB1 frameshift mutation. Interestingly, upfront combined treatment with a PI3K-alpha inhibitor delayed the onset of tumor progression. Two out of the remaining five CDK4/6-resistant PDXs harbored either a frameshift mutation in RB1 (plus loss of heterozygosity) or had low pRb protein expression. Two other resistant models harbored CCND1 and MYC amplifications. The remaining one harbored a TSC1 loss. In all the CDK4/6-resistant PDX, however, the combination of CDK4/6 and PI3K-alpha inhibition resulted in tumor regression. From our results, we conclude that loss of G1-cell cycle checkpoint control, such as mutation/loss of RB1 and CCND1-amplification, is associated with lack of response to CDK4/6 blockade in ER+ BC PDX. The addition of a PI3K-alpha inhibitor results in improvement of disease control in all experimental models tested. Citation Format: Violeta Serra, Marta Palafox, Maria-Teresa Herrera, Martin A Rivas, Marta Guzman, Olga Rodriguez, Judit Grueso, Meritxell Bellet, Mafalda Oliveira, Cristina Saura, Emmanuelle di Tomaso, Giordi Camponigro, Nicholas C. Turner, Javier Cortes, Jose Baselga. Identification of CDK4/6-response biomarkers using estrogen receptor-positive breast cancer patient-derived xenografts (PDX). [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2825.


Cancer Research | 2012

Abstract 4655: Aplidin induces a non-canonical ER stress response in HeLa cells response in HeLa cells

Alejandro Losada; Alberto Bejarano; Marta Palafox; David Moreno-Mateos; Victoria Moneo; Antonio Jiménez-Ruiz; Federico Gago; Carmen Cuevas; Luis García-Fernández; Juan Leal; Carlos M. Galmarini

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Aplidin (APL), a marine cyclic depsipeptide originally found in the Mediterranean tunicate Aplidium albicans, is currently under phase II/III clinical investigation for cancer therapy. The mechanism of action of the compound includes the oxidative-stress mediated activation of Rac1 and JNK1, which rapidly trigger the mitochondrial apoptotic pathway. Comparing the protein expression profiles of HeLa wt cells and an APL-resistant subclone previously generated in our lab (HeLa-APL), we identified a subset of differentially expressed proteins the function of which was related to the unfolded protein response (UPR). HeLa-APL resistant cells showed reduced levels of BiP (GRP78) and relatively higher levels of Ero1α and phospho-eIF2α, indicating a higher basal endoplasmic reticulum (ER) stress. To investigate whether APL was inducing a bona fide ER stress response in HeLa cells and whether this process was essential in the mechanism of action of this compound, we compared the molecular and cellular effects elicited by APL with those induced by two well-known ER-stress inducing agents, thapsigargin and tunicamycin. Basically, while these agents elicited a complete canonical UPR response, APL only triggered part of the stress signaling cascade, including the phosphorylation of eIF2α and JNK1 and a rapid inhibition of protein synthesis. By contrast, CHOP, a transcription factor involved in launching apoptosis by the UPR, was not induced by APL. Rather, it seemed to be slightly reduced in treated cells. Similarly, while tunicamycin induced the alternative splicing of XBP1 by IRE1 and the activation of the ER-related caspase-4, APL failed to induce the same response in HeLa cells. At present, the precise connection between the partial activation of the ER stress signaling pathway and the rapid induction of apoptosis by APL remains poorly understood, although it could represent a new, aberrant UPR response Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4655. doi:1538-7445.AM2012-4655

Collaboration


Dive into the Marta Palafox's collaboration.

Top Co-Authors

Avatar

Marta Guzman

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicholas C. Turner

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Joaquín Arribas

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

José Baselga

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ander Urruticoechea

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana Lluch

University of Valencia

View shared research outputs
Top Co-Authors

Avatar

Ana Rovira

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Irene Ferrer

Spanish National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge