27 research outputs found

    Epigenetic silencing of TGFBI confers resistance to trastuzumab in human breast cancer

    Get PDF
    Background: acquired resistance to trastuzumab is a major clinical problem in the treatment of HER2-positive (HER2+) breast cancer patients. The selection of trastuzumab-resistant patients is a great challenge of precision oncology. The aim of this study was to identify novel epigenetic biomarkers associated to trastuzumab resistance in HER2+ BC patients. Methods: we performed a genome-wide DNA methylation (450K array) and a transcriptomic analysis (RNA-Seq) comparing trastuzumab-sensitive (SK) and trastuzumab-resistant (SKTR) HER2+ human breast cancer cell models. The methylation and expression levels of candidate genes were validated by bisulfite pyrosequencing and qRT-PCR, respectively. Functional assays were conducted in the SK and SKTR models by gene silencing and overexpression. Methylation analysis in 24 HER2+ human BC samples with complete response or non-response to trastuzumab-based treatment was conducted by bisulfite pyrosequencing. Results: epigenomic and transcriptomic analysis revealed the consistent hypermethylation and downregulation of TGFBI, CXCL2, and SLC38A1 genes in association with trastuzumab resistance. The DNA methylation and expression levels of these genes were validated in both sensitive and resistant models analyzed. Of the genes, TGFBI presented the highest hypermethylation-associated silencing both at the transcriptional and protein level. Ectopic expression of TGFBI in the SKTR model suggest an increased sensitivity to trastuzumab treatment. In primary tumors, TGFBI hypermethylation was significantly associated with trastuzumab resistance in HER2+ breast cancer patients. Conclusions: our results suggest for the first time an association between the epigenetic silencing of TGFBI by DNA methylation and trastuzumab resistance in HER2+ cell models. These results provide the basis for further clinical studies to validate the hypermethylation of TGFBI promoter as a biomarker of trastuzumab resistance in HER2+ breast cancer patients

    Silibinin Suppresses Tumor Cell-Intrinsic Resistance to Nintedanib and Enhances Its Clinical Activity in Lung Cancer

    Get PDF
    The anti-angiogenic agent nintedanib has been shown to prolong overall and progression-free survival in patients with advanced non-small-cell lung cancer (NSCLC) who progress after first-line platinum-based chemotherapy and second-line immunotherapy. Here, we explored the molecular basis and the clinical benefit of incorporating the STAT3 inhibitor silibinin-a flavonolignan extracted from milk thistle-into nintedanib-based schedules in advanced NSCLC. First, we assessed the nature of the tumoricidal interaction between nintedanib and silibinin and the underlying relevance of STAT3 activation in a panel of human NSCLC cell lines. NSCLC cells with poorer cytotoxic responses to nintedanib exhibited a persistent, nintedanib-unresponsive activated STAT3 state, and deactivation by co-treatment with silibinin promoted synergistic cytotoxicity. Second, we tested whether silibinin could impact the lysosomal sequestration of nintedanib, a lung cancer cell-intrinsic mechanism of nintedanib resistance. Silibinin partially, but significantly, reduced the massive lysosomal entrapment of nintedanib occurring in nintedanib-refractory NSCLC cells, augmenting the ability of nintedanib to reach its intracellular targets. Third, we conducted a retrospective, observational multicenter study to determine the efficacy of incorporating an oral nutraceutical product containing silibinin in patients with NSCLC receiving a nintedanib/docetaxel combination in second- and further-line settings (n = 59). Overall response rate, defined as the combined rates of complete and partial responses, was significantly higher in the study cohort receiving silibinin supplementation (55%) than in the control cohort (22%, p = 0.011). Silibinin therapy was associated with a significantly longer time to treatment failure in multivariate analysis (hazard ratio 0.43, p = 0.013), despite the lack of overall survival benefit (hazard ratio 0.63, p = 0.190). Molecular mechanisms dictating the cancer cell-intrinsic responsiveness to nintedanib, such as STAT3 activation and lysosomal trapping, are amenable to pharmacological intervention with silibinin. A prospective, powered clinical trial is warranted to confirm the clinical relevance of these findings in patients with advanced NSCLC

    New synthetic inhibitors of fatty acid synthase with anticancer activity

    Get PDF
    Fatty acid synthase (FASN) is a lipogenic enzyme that is highly expressed in different human cancers. Here we report the development of a new series of polyphenolic compounds 5-­30 that have been evaluated for their cytotoxic capacity in SK-­Br3 cells, a human breast cancer cell line with high FASN expression. The compounds with an IC50 < 50 M have been tested for their ability to inhibit FASN activity. Among them, derivative 30 blocks the 90% of FASN activity at low concentration (4 M), is highly cytotoxic in a broad panel of tumor cells, induces apoptosis, and blocks the activation of HER2, AKT and ERK pathways. Remarkably, 30 does not activate carnitine palmitoyltransferase-­1 (CPT-­1) nor induces in mice weight loss, which are the main drawbacks of other previously described FASN inhibitors. Thus, FASN inhibitor 30 may aid the validation of this enzyme as a therapeutic target for the treatment of cancer

    Highly sensitive MLH1 methylation analysis in blood identifies a cancer patient with low-level mosaic MLH1 epimutation

    Get PDF
    Constitutional MLH1 methylation (epimutation) is a rare cause of Lynch syndrome. Low-level methylation (<= 10%) has occasionally been described. This study aimed to identify low-level constitutional MLH1 epimutations and determine its causal role in patients with MLH1-hypermethylated colorectal cancer. Eighteen patients with MLH1-hypermethylated colorectal tumors in whom MLH1 methylation was previously undetected in blood by methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) were screened for MLH1 methylation using highly sensitive MS-melting curve analysis (MS-MCA). Constitutional methylation was characterized by different approaches. MS-MCA identified one patient (5.6%) with low-level MLH1 methylation ( 1%) in blood and other normal tissues, which was confirmed by clonal bisulfite sequencing in blood. The patient had developed three clonally related gastrointestinal MLH1-methylated tumor lesions at 22, 24, and 25 years of age. The methylated region in normal tissues overlapped with that reported for other carriers of constitutional MLH1 epimutations. Low-level MLH1 methylation and reduced allelic expression were linked to the same genetic haplotype, whereas the opposite allele was lost in patient's tumors. Mutation screening of MLH1 and other hereditary cancer genes was negative. Herein, a highly sensitive MS-MCA-based approach has demonstrated its utility for the identification of low-level constitutional MLH1 epigenetic mosaicism. The eventual identification and characterization of additional cases will be critical to ascertain the cancer risks associated with constitutional MLH1 epigenetic mosaicism

    Neoadjuvant Metformin Added to Systemic Therapy Decreases the Proliferative Capacity of Residual Breast Cancer

    Get PDF
    The proliferative capacity of residual breast cancer (BC) disease indicates the existence of partial treatment resistance and higher probability of tumor recurrence. We explored the therapeutic potential of adding neoadjuvant metformin as an innovative strategy to decrease the proliferative potential of residual BC cells in patients failing to achieve pathological complete response (pCR) after pre-operative therapy. We performed a prospective analysis involving the intention-to-treat population of the (Metformin and Trastuzumab in Neoadjuvancy) METTEN study, a randomized multicenter phase II trial of women with primary, non-metastatic (human epidermal growth factor receptor 2) HER2-positive BC evaluating the efficacy, tolerability, and safety of oral metformin (850 mg twice-daily) for 24 weeks combined with anthracycline/taxane-based chemotherapy and trastuzumab (arm A) or equivalent regimen without metformin (arm B), before surgery. We centrally evaluated the proliferation marker Ki67 on sequential core biopsies using visual assessment (VA) and an (Food and Drug Administration) FDA-cleared automated digital image analysis (ADIA) algorithm. ADIA-based pre-operative values of high Ki67 (>= 20%), but not those from VA, significantly predicted the occurrence of pCR in both arms irrespective of the hormone receptor status (p = 0.024 and 0.120, respectively). Changes in Ki67 in residual tumors of non-pCR patients were significantly higher in the metformin-containing arm (p = 0.025), with half of all patients exhibiting high Ki67 at baseline moving into the low-Ki67 (<20%) category after neoadjuvant treatment. By contrast, no statistically significant changes in Ki67 occurred in residual tumors of the control treatment arm (p = 0.293). There is an urgent need for innovative therapeutic strategies aiming to provide the protective effects of decreasing Ki67 after neoadjuvant treatment even if pCR is not achieved. Metformin would be evaluated as a safe candidate to decrease the aggressiveness of residual disease after neoadjuvant (pre-operative) systemic therapy of BC patients

    Different fatty acid metabolism effects of (−)-epigallocatechin-3-gallate and C75 in adenocarcinoma lung cancer

    Get PDF
    Background Fatty acid synthase (FASN) is overexpressed and hyperactivated in several human carcinomas, including lung cancer. We characterize and compare the anti-cancer effects of the FASN inhibitors C75 and (−)-epigallocatechin-3-gallate (EGCG) in a lung cancer model. Methods We evaluated in vitro the effects of C75 and EGCG on fatty acid metabolism (FASN and CPT enzymes), cellular proliferation, apoptosis and cell signaling (EGFR, ERK1/2, AKT and mTOR) in human A549 lung carcinoma cells. In vivo, we evaluated their anti-tumour activity and their effect on body weight in a mice model of human adenocarcinoma xenograft. Results C75 and EGCG had comparable effects in blocking FASN activity (96,9% and 89,3% of inhibition, respectively). In contrast, EGCG had either no significant effect in CPT activity, the rate-limiting enzyme of fatty acid β-oxidation, while C75 stimulated CPT up to 130%. Treating lung cancer cells with EGCG or C75 induced apoptosis and affected EGFR-signaling. While EGCG abolished p-EGFR, p-AKT, p-ERK1/2 and p-mTOR, C75 was less active in decreasing the levels of EGFR and p-AKT. In vivo, EGCG and C75 blocked the growth of lung cancer xenografts but C75 treatment, not EGCG, caused a marked animal weight loss. Conclusions In lung cancer, inhibition of FASN using EGCG can be achieved without parallel stimulation of fatty acid oxidation and this effect is related mainly to EGFR signaling pathway. EGCG reduce the growth of adenocarcinoma human lung cancer xenografts without inducing body weight loss. Taken together, EGCG may be a candidate for future pre-clinical development

    Guia de pràctica clínica: tractament del dolor oncològic pediàtric

    Get PDF
    Dolor oncològic pediàtric; Càncer; Tractament; RegistreDolor oncológico pediátrico; Cáncer; Tratamiento; RegistroPediatric oncological pain; Cancer; Treatment; RegistryEl dolor oncològic pediàtric és un problema clínic rellevant a l'Hospital Vall d'hebron i les dades de què es disposa sobre el seu tractament són escasses. Aquest document s'ha desenvolupat per millorar el registre i el tractament del dolor en nens amb càncer atesos a l’Hospital, sobre la base de l’evidència científica de què es disposa. Es pretén aconseguir que, d’una banda, la mesura del dolor es faci de forma regular, amb la metodologia adequada i es registri sistemàticament a la història clínica. De l’altra, utilitzar les estratègies de tractament del dolor, tant farmacològiques com no farmacològiques; més efectives i segures per a aquest tipus de població. La utilització pràctica d’aquesta publicació s’ha de considerar com una guia; no es pretén anteposar-la al judici clínic

    A novel inhibitor of fatty acid synthase shows activity against HER2+ breast cancer xenografts and is active in anti-HER2 drug-resistant cell lines

    Get PDF
    Introduction: Inhibiting the enzyme Fatty Acid Synthase (FASN) leads to apoptosis of breast carcinoma cells, and this is linked to human epidermal growth factor receptor 2 (HER2) signaling pathways in models of simultaneous expression of FASN and HER2. Methods: In a xenograft model of breast carcinoma cells that are FASN+ and HER2+, we have characterised the anticancer activity and the toxicity profile of G28UCM, the lead compound of a novel family of synthetic FASN inhibitors. In vitro, we analysed the cellular and molecular interactions of combining G28UCM with anti-HER drugs. Finally, we tested the cytotoxic ability of G28UCM on breast cancer cells resistant to trastuzumab or lapatinib, that we developed in our laboratory. Results: In vivo, G28UCM reduced the size of 5 out of 14 established xenografts. In the responding tumours, we observed inhibition of FASN activity, cleavage of poly-ADPribose polymerase (PARP) and a decrease of p-HER2, p- protein kinase B (AKT) and p-ERK1/2, which were not observed in the nonresponding tumours. In the G28UCM-treated animals, no significant toxicities occurred, and weight loss was not observed. In vitro, G28UCM showed marked synergistic interactions with trastuzumab, lapatinib, erlotinib or gefitinib (but not with cetuximab), which correlated with increases in apoptosis and with decreases in the activation of HER2, extracellular signal-regulated kinase (ERK)1/2 and AKT. In trastuzumab-resistant and in lapatinib-resistant breast cancer cells, in which trastuzumab and lapatinib were not effective, G28UCM retained the anticancer activity observed in the parental cells. Conclusions: G28UCM inhibits fatty acid synthase (FASN) activity and the growth of breast carcinoma xenografts in vivo, and is active in cells with acquired resistance to anti-HER2 drugs, which make it a candidate for further pre-clinical development

    Guia de pràctica clínica: abordatge del tractament del dolor neuropàtic

    Get PDF
    Dolor neuropàtic; Epidemiologia; Hospital; Atenció primàriaDolor neuropático; Epidemiología; Hospital; Atención primariaNeuropathic pain; Epidemiology; Hospital; Primary careLa Guia pretén estructurar i consensuar l'atenció dels malalts amb dolor neuropàtic dins del nostre territori. S'especifiquen intervencions preventives i terapèutiques, així com a quin nivell el malalts han de ser atesos en funció de la seva situació clínica i la seva complexitat, els fluxos i metodologia bàsica de derivacions. Es vol potenciar un abordatge transversal i integral del dolor neuropàtic, que abasta l'atenció primària i l'atenció especialitzada hospitalària, i promoure la continuïtat assistencial entre ambdós nivells assistencials, tenint en compte intervencions i criteris compartits, per tal de disminuir la variabilitat de la pràctica clínica i millorar la qualitat i seguretat del pacient

    Derivats del (-)3-galat d'epigalocatequina com a nous inhibidors de la sintasa d'àcids grassos amb efectes antitumorals en models cel•lulars i animals de càncer de mama

    Get PDF
    [cat] La sintasa d’àcids grassos (FASN), enzim que realitza la síntesi de novo dels àcids grassos, es sobreexpressa en alguns càncers, sobretot en càncer de mama, i s’associa a un mal pronòstic. La seva regulació està relacionada amb la via de senyalització d’HER2, receptor cel.lular de membrana que pertany a la família dels receptors de creixement epidèrmic (HER), tot i que encara es desconeixen els mecanismes moleculars d’aquesta relació. El coneixement de la implicació del paper del metabolisme dels àcids grassos en la iniciació i la progressió del càncer de mama i el desenvolupament d’inhibidors farmacològics de l’activitat de FASN té un gran interès científico-mèdic. L’ús clínic d’inhibidors de FASN, com la cerulenina o el C75, presenten una baixa estabilitat i efectes col.laterals com l’anorèxia i la pèrdua de pes, ja que activen la Carnitina Palmitoil Transferasa 1 (CPT1), enzim regulador de la β-oxidació dels àcids grassos. Anteriorment el nostre grup va demostrar que el (-)-3-galat d’epigalocatequina (EGCG), un polifenol present en el té verd, inhibeix FASN, té efectes antitumorals i és més selectiu que la cerulenina i el C75 perquè no afecta la CPT1, i en conseqüència no produeix pèrdua de pes in vivo. L’ús terapèutic de l’EGCG com agent antitumoral es veu limitat per l’elevat valor de la IC50 (~ 150 >M) i també per la inestabilitat en condicions fisiològiques. En base a aquests resultats, en aquest treball es presenta el disseny d’una família de compostos polifenòlics relacionats estructuralment amb l’EGCG per millorar-ne la seva activitat antitumoral, la biodistribució i l’estabilitat. In vitro s’ha realitzat l’avaluació de l’activitat biològica i molecular d’aquests compostos per tal de valorar-ne l’especificitat, la capacitat inhibidora específica de FASN i l’activitat citotòxica en diferents models de càncer de mama. S’ha determinat que el compost G28UCM presenta una inhibició de l’activitat de FASN d’un 90% i una elevada citotoxicitat en cèl.lules de càncer de mama (IC50= 30 >M). Per altra banda, s’han analitzat les interaccions cel.lulars i moleculars del composta G28UCM en combinació amb les teràpies anti-HER existents (trastuzumab, lapatinib, erlotinib i cetuximab) en un model cel.lular de càncer de mama FASN/HER2 positiu. El G28UCM mostra un clar sinergisme farmacològic amb els fàrmacs anti-HER, trastuzumab, lapatinib i erlotinib, a diferència de l’EGCG que tant sols mostra additivisme amb el trastuzumab. A més al laboratori s’han desenvolupat models cel.lulars de càncer de mama resistents als fàrmacs anti-HER2, trastuzumab i lapatinib, i s’ha avaluat si el G28UCM té activitat antitumoral en aquests models cel.lulars. Els resultats obtinguts ens mostren que el G28UCM segueix mostrant activitat citotòxica amb les cèl.lules resistents als fàrmacs anti-HER2. In vivo, primer es va desenvolupar el millor model murí de carcinoma de mama FASN i HER2+ utilitzant els compostos inhibidors de FASN de referència EGCG i C75. Un cop establert el model animal es va caracteritzar l’activitat antitumoral i el perfil de toxicitat del compost seleccionat G28UCM. Es va obtenir una inhibició del creixement tumoral en un terç dels ratolins tractats amb el G28UCM, els quals no varen mostrar cap tipus de toxicitat (renal, hepàtica, cardíaca) ni pèrdua de pes. Aquests resultats proporcionen el fonament pel desenvolupament preclínic del G28UCM, ja sigui sol o en combinació amb les teràpies anti-HER existents en els tumors de mama que sobreexpressen HER2 i els que han progressat al tractament.[eng] Fatty Acid Synthase (FASN), an enzyme responsible for the de novo synthesis of fatty acids is overly manifested in certain types of cancer, especially in breast cancer, and it is associated with a poor diagnosis. Its regulation is associated with the signalling of HER2, a cellular receptor belonging to the human epidermal growth receptors family. However, the molecular mechanisms associated with this relationship are not well understood. FASN inhibitors, such as cerulenin or C75, offers low levels of stability and present side effects, like anorexia and weight loss, since they activate Carnitina Palmitoil Transferasa 1 (CPT1), a regulator enzyme of fatty acids’ β-oxidation. Previously our research group has proven that epigallocatechin 3-gallate (EGCG), a polyphenol present in green tea, inhibits FASN, has anti-tumoral effects and since it does not affect CPT1, is more selective than both cerulenin and C75. The therapeutic use of EGCG as an anti-tumoral agent is limited by its high IC50 value, as well as by its instability in physiological conditions. Following these results, a family of polyphenol compounds structurally related to EGCG has been designed with the goal of enhancing its anti-tumoral activity, its biodistribution and its stability. In vitro, we evaluated the biological and molecular activity of these compounds to assessing their specificity, their FASN-specific inhibiting capacity and their cytotoxic activity in different models of breast cancer. G28UCM shows a 90% inhibition of FASN activity and a high cytotoxicity levels in breast cancer cells (IC50= 30 9M). Also, G28UCM were analyzed in combination with already existing anti-HER therapies (trastuzumab, lapatinib, erlotinib, and cetuximab) in a cellular model of FASN/HER2- positive breast cancer. G28UCM shows synergysm with trastuzumab, lapatinib, and erlotinib, as opposed to EGCG, which only shows additivism with trastuzumab. Further, we also developed cellular models of breast cancer, which were resistant to trastuzumab, and lapatinib, and assessed whether G28UCM presents anti-tumoral activity in said cellular models. G28UCM presents cytotoxic activity with anti-HER2 drug resistant cells. In vivo, the tumoral activity and the toxicity profile of G28UCM were characterized in the murine model of FASN and HER2+ breast carcinoma. The results showed an inhibition of tumor growth in mice treated with a third G28UCM and any toxicity or weight loss. In summary, our findings provide the basis for the pre-clinical development of G28UCM
    corecore