Browse
Search for other papers by K E Lines in
Google Scholar
PubMed
Search for other papers by P Filippakopoulos in
Google Scholar
PubMed
Search for other papers by M Stevenson in
Google Scholar
PubMed
Search for other papers by S Müller in
Google Scholar
PubMed
Search for other papers by H E Lockstone in
Google Scholar
PubMed
Search for other papers by B Wright in
Google Scholar
PubMed
Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
Search for other papers by S Knapp in
Google Scholar
PubMed
Search for other papers by D Buck in
Google Scholar
PubMed
Search for other papers by C Bountra in
Google Scholar
PubMed
Search for other papers by R V Thakker in
Google Scholar
PubMed
Medical treatments for corticotrophinomas are limited, and we therefore investigated the effects of epigenetic modulators, a new class of anti-tumour drugs, on the murine adrenocorticotropic hormone (ACTH)-secreting corticotrophinoma cell line AtT20. We found that AtT20 cells express members of the bromo and extra-terminal (BET) protein family, which bind acetylated histones, and therefore, studied the anti-proliferative and pro-apoptotic effects of two BET inhibitors, referred to as (+)-JQ1 (JQ1) and PFI-1, using CellTiter Blue and Caspase Glo assays, respectively. JQ1 and PFI-1 significantly decreased proliferation by 95% (P < 0.0005) and 43% (P < 0.0005), respectively, but only JQ1 significantly increased apoptosis by >50-fold (P < 0.0005), when compared to untreated control cells. The anti-proliferative effects of JQ1 and PFI-1 remained for 96 h after removal of the respective compound. JQ1, but not PFI-1, affected the cell cycle, as assessed by propidium iodide staining and flow cytometry, and resulted in a higher number of AtT20 cells in the sub G1 phase. RNA-sequence analysis, which was confirmed by qRT-PCR and Western blot analyses, revealed that JQ1 treatment significantly altered expression of genes involved in apoptosis, such as NFκB, and the somatostatin receptor 2 (SSTR2) anti-proliferative signalling pathway, including SSTR2. JQ1 treatment also significantly reduced transcription and protein expression of the ACTH precursor pro-opiomelanocortin (POMC) and ACTH secretion by AtT20 cells. Thus, JQ1 treatment has anti-proliferative and pro-apoptotic effects on AtT20 cells and reduces ACTH secretion, thereby indicating that BET inhibition may provide a novel approach for treatment of corticotrophinomas.
Search for other papers by Jesús Morillo-Bernal in
Google Scholar
PubMed
Molecular Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Madrid, Spain
Search for other papers by Lara P Fernández in
Google Scholar
PubMed
Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
Search for other papers by Pilar Santisteban in
Google Scholar
PubMed
FOXE1 is a thyroid-specific transcription factor essential for thyroid gland development and maintenance of the differentiated state. Interestingly, a strong association has been recently described between FOXE1 expression and susceptibility to thyroid cancer, but little is known about the mechanisms underlying FOXE1-induced thyroid tumorigenesis. Here, we used a panel of human thyroid cancer-derived cell lines covering the spectrum of thyroid cancer phenotypes to examine FOXE1 expression and to test for correlations between FOXE1 expression, the allele frequency of two SNPs and a length polymorphism in or near the FOXE1 locus associated with cancer susceptibility, and the migration ability of thyroid cancer cell lines. Results showed that FOXE1 expression correlated with differentiation status according to histological sub-type, but not with SNP genotype or cell migration ability. However, loss-and-gain-of-function experiments revealed that FOXE1 modulates cell migration, suggesting a role in epithelial-to-mesenchymal transition (EMT). Our previous genome-wide expression analysis identified Zeb1, a major EMT inducer, as a putative Foxe1 target gene. Indeed, gene silencing of FOXE1 decreased ZEB1 expression, whereas its overexpression increased ZEB1 transcriptional activity. FOXE1 was found to directly interact with the ZEB1 promoter. Lastly, ZEB1 silencing decreased the ability of thyroid tumoral cells to migrate and invade, pointing to its importance in thyroid tumor mestastases. In conclusion, we have identified ZEB1 as a bona fide target of FOXE1 in thyroid cancer cells, which provides new insights into the role of FOXE1 in regulating cell migration and invasion in thyroid cancer.
Search for other papers by Martina Gruber in
Google Scholar
PubMed
Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
Search for other papers by Lavinia Ferrone in
Google Scholar
PubMed
Search for other papers by Martin Puhr in
Google Scholar
PubMed
Search for other papers by Frédéric R Santer in
Google Scholar
PubMed
Search for other papers by Tobias Furlan in
Google Scholar
PubMed
Search for other papers by Iris E Eder in
Google Scholar
PubMed
Search for other papers by Natalie Sampson in
Google Scholar
PubMed
Search for other papers by Georg Schäfer in
Google Scholar
PubMed
Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
Search for other papers by Florian Handle in
Google Scholar
PubMed
Search for other papers by Zoran Culig in
Google Scholar
PubMed
Administration of the microtubule inhibitor docetaxel is a common treatment for metastatic castration-resistant prostate cancer (mCRPC) and results in prolonged patient overall survival. Usually, after a short period of time chemotherapy resistance emerges and there is urgent need to find new therapeutic targets to overcome therapy resistance. The lysine-acetyltransferase p300 has been correlated to prostate cancer (PCa) progression. Here, we aimed to clarify a possible function of p300 in chemotherapy resistance and verify p300 as a target in chemoresistant PCa. Immunohistochemistry staining of tissue samples revealed significantly higher p300 protein expression in patients who received docetaxel as a neoadjuvant therapy compared to control patients. Elevated p300 expression was confirmed by analysis of publicly available patient data, where significantly higher p300 mRNA expression was found in tissue of mCRPC tumors of docetaxel-treated patients. Consistently, docetaxel-resistant PCa cells showed increased p300 protein expression compared to docetaxel-sensitive counterparts. Docetaxel treatment of PCa cells for 72 h resulted in elevated p300 expression. shRNA-mediated p300 knockdown did not alter colony formation efficiency in docetaxel-sensitive cells, but significantly reduced clonogenic potential of docetaxel-resistant cells. Downregulation of p300 in docetaxel-resistant cells also impaired cell migration and invasion. Taken together, we showed that p300 is upregulated by docetaxel, and our findings suggest that p300 is a possible co-target in treatment of chemoresistant PCa.
Search for other papers by Frances Collins in
Google Scholar
PubMed
Search for other papers by Nozomi Itani in
Google Scholar
PubMed
Search for other papers by Arantza Esnal-Zufiaurre in
Google Scholar
PubMed
Search for other papers by Douglas A Gibson in
Google Scholar
PubMed
Search for other papers by Carol Fitzgerald in
Google Scholar
PubMed
Search for other papers by Philippa T K Saunders in
Google Scholar
PubMed
Endometrial cancer is a common gynaeological malignancy: life time exposure to oestrogen is a key risk factor. Oestrogen action is mediated by receptors encoded by ESR1 (ERα) and ESR2 (ERβ): ERα plays a key role in regulating endometrial cell proliferation. A truncated splice variant isoform (ERβ5) encoded by ESR2 is highly expressed in cancers. This study explored whether ERβ5 alters oestrogen responsiveness of endometrial epithelial cells. Immunhistochemistry profiling of human endometrial cancer tissue biopsies identified epithelial cells co-expressing ERβ5 and ERα in stage I endometrial adenocarcinomas and post menopausal endometrium. Induced co-expression of ERβ5 in ERαpos endometrial cancer cells (Ishikawa) significantly increased ligand-dependent activation of an ERE-luciferase reporter stimulated by either E2 or the ERα-selective agonist 1,3,5-(4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT) compared to untransfected cells. Fluorescence recovery after photobleaching (FRAP) analysis of tagged yellow fluorescent protein (YFP)-ERβ5 transfected into Ishikawa cells revealed that incubation with E2 induced a transient reduction in intra-nuclear mobility characterised by punctate protein redistribution which phenocopied the behaviour of ERα following ligand activation with E2. In ERαneg MDA-MD-231 breast cancer cells, there was no E2-dependent change in mobility of YFP-ERβ5 and no activation of the ERE reporter in cells expressing ERβ5. In conclusion, we demonstrate that ERβ5 can act as heterodimeric partner to ERα in Ishikawa cells and increases their sensitivity to E2. We speculate that expression of ERβ5 in endometrial epithelial cells may increase the risk of malignant transformation and suggest that immunostaining for ERβ5 should be included in diagnostic assessment of women with early grade cancers.
Search for other papers by Jonathan Wesley Nyce in
Google Scholar
PubMed
We have recently described in this journal our detection of an anthropoid primate-specific, adrenal androgen-dependent, p53-mediated, ‘kill switch’ tumor suppression mechanism that reached its fullest expression only in humans, as a result of human-specific exposure to polycyclic aromatic hydrocarbons caused by the harnessing of fire – but which has components reaching all the way back to the origin of the primate lineage. We proposed that species-specific mechanisms of tumor suppression are a generalized requirement for vertebrate species to increase in body size or lifespan beyond those of species basal to their lineage or to exploit environmental niches which increase exposure to carcinogenic substances. Using empirical dynamic modeling, we have also reported our detection of a relationship between body size, lifespan, and species-specific mechanism of tumor suppression (and here add carcinogen exposure), such that a change in any one of these variables requires an equilibrating change in one or more of the others in order to maintain lifetime cancer risk at a value of about 4%, as observed in virtually all larger, longer-lived species under natural conditions. Here we show how this relationship, which we refer to as the lex naturalis of vertebrate speciation, elucidates the evolutionary steps underlying an adrenal androgen-dependent, human-specific ‘kill switch’ tumor suppression mechanism; and further, how it prescribes a solution to ‘normalize’ lifetime cancer risk in our species from its current aberrant 40% to the 4% that characterized primitive humans. We further argue that this prescription writ by the lex naturalis represents the only tenable strategy for meaningful suppression of the accelerating impact of cancer upon our species.
Search for other papers by Rajeev Mishra in
Google Scholar
PubMed
Search for other papers by Subhash Haldar in
Google Scholar
PubMed
Search for other papers by Surabhi Suchanti in
Google Scholar
PubMed
Department of Research, Greater Los Angeles Veterans Administration, Los Angeles, California, USA
Search for other papers by Neil A Bhowmick in
Google Scholar
PubMed
Genomic changes that drive cancer initiation and progression contribute to the co-evolution of the adjacent stroma. The nature of the stromal reprogramming involves differential DNA methylation patterns and levels that change in response to the tumor and systemic therapeutic intervention. Epigenetic reprogramming in carcinoma-associated fibroblasts are robust biomarkers for cancer progression and have a transcriptional impact that support cancer epithelial progression in a paracrine manner. For prostate cancer, promoter hypermethylation and silencing of the RasGAP, RASAL3 that resulted in the activation of Ras signaling in carcinoma-associated fibroblasts. Stromal Ras activity initiated a process of macropinocytosis that provided prostate cancer epithelia with abundant glutamine for metabolic conversion to fuel its proliferation and a signal to transdifferentiate into a neuroendocrine phenotype. This epigenetic oncogenic metabolic/signaling axis seemed to be further potentiated by androgen receptor signaling antagonists and contributed to therapeutic resistance. Intervention of stromal signaling may complement conventional therapies targeting the cancer cell.
Search for other papers by S Felder in
Google Scholar
PubMed
Search for other papers by H Jann in
Google Scholar
PubMed
Search for other papers by R Arsenic in
Google Scholar
PubMed
Search for other papers by T Denecke in
Google Scholar
PubMed
Klinik für Nuklearmedizin, Universitätklinikum Ulm, Ulm, Germany
Search for other papers by V Prasad in
Google Scholar
PubMed
Search for other papers by B Knappe-Drzikova in
Google Scholar
PubMed
Innere Medizin und Gastroenterologie, Asklepios Klinik St. Georg, Asklepios Medical School, Hamburg, Germany
Search for other papers by S Maasberg in
Google Scholar
PubMed
Search for other papers by B Wiedenmann in
Google Scholar
PubMed
Medizinische Klinik 1, Gastroenterologie, Pneumologie und Endokrinologie, Universitätsklinikum der Friedrich-Alexander Universität Erlangen, Erlangen, Germany
Search for other papers by M Pavel in
Google Scholar
PubMed
Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Uinversitätsklinikum Münster, Münster, Germany
Search for other papers by A Pascher in
Google Scholar
PubMed
Innere Medizin und Gastroenterologie, Asklepios Klinik St. Georg, Asklepios Medical School, Hamburg, Germany
Search for other papers by U F Pape in
Google Scholar
PubMed
Although gastric neuroendocrine neoplasias (gNEN) are an orphan disease, their incidence is rising. The heterogeneous clinical course powers the ongoing discussion of the most appropriate classification system and management. Prognostic relevance of proposed classifications was retrospectively analysed in 142 patients from a single tertiary referral centre. Baseline, management and survival data were acquired for statistical analyses. The distribution according to the clinicopathological typification was gNEN-1 (n = 86/60.6%), gNEN-2 (n = 7/4.9%), gNEN-3 (n = 24/16.9%) and gNEN-4 (n = 25/17.6%), while hypergastrinemia-associated gNEN-1 and -2 were all low-grade tumours (NET-G1/2), formerly termed sporadic gNEN-3 could be subdivided into gNEN-3 with grade 1 or 2 and gNEN-4 with grade 3 (NEC-G3). During follow-up 36 patients died (25%). The mean overall survival (OS) of all gNEN was 14.2 years. The OS differed statistically significant across all subgroups with either classification system. According to UICC 2017 TNM classification, OS differed for early and advanced stages, while WHO grading indicated poorer prognosis for NEC-G3. Cox regression analysis confirmed the independent prognostic validity of either classification system for survival. Particularly careful analysis of the clinical course of gNEN-1 (ECLomas, gastric carcinoids) confirmed their mostly benign, but recurrent and extremely slowly progressive behaviour with low risk of metastasis (7%) and an efficient long-term control by repetitive endoscopic procedures. Our study provides evidence for the validity of current classifications focusing on typing, grading and staging. These are crucial tools for risk stratification, especially to differentiate gNEN-1 as well as sporadic gNET and gNEC (gNEN-3 vs -4).
Search for other papers by Tobias Hofving in
Google Scholar
PubMed
Search for other papers by Viktor Sandblom in
Google Scholar
PubMed
Search for other papers by Yvonne Arvidsson in
Google Scholar
PubMed
Search for other papers by Emman Shubbar in
Google Scholar
PubMed
Search for other papers by Gülay Altiparmak in
Google Scholar
PubMed
Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
Search for other papers by John Swanpalmer in
Google Scholar
PubMed
Search for other papers by Bilal Almobarak in
Google Scholar
PubMed
Search for other papers by Anna-Karin Elf in
Google Scholar
PubMed
Search for other papers by Viktor Johanson in
Google Scholar
PubMed
Search for other papers by Erik Elias in
Google Scholar
PubMed
Search for other papers by Erik Kristiansson in
Google Scholar
PubMed
Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
Search for other papers by Eva Forssell-Aronsson in
Google Scholar
PubMed
Search for other papers by Ola Nilsson in
Google Scholar
PubMed
177Lu-octreotate is an FDA-approved radionuclide therapy for patients with gastroenteropancreatic neuroendocrine tumours (NETs) expressing somatostatin receptors. The 177Lu-octreotate therapy has shown promising results in clinical trials by prolonging progression-free survival, but complete responses are still uncommon. The aim of this study was to improve the 177Lu-octreotate therapy by means of combination therapy. To identify radiosensitising inhibitors, two cell lines, GOT1 and P-STS, derived from small intestinal neuroendocrine tumours (SINETs), were screened with 1224 inhibitors alone or in combination with external radiation. The screening revealed that inhibitors of Hsp90 can potentiate the tumour cell-killing effect of radiation in a synergistic fashion (GOT1; false discovery rate <3.2 × 10−11). The potential for Hsp90 inhibitor ganetespib to enhance the anti-tumour effect of 177Lu-octreotate in an in vivo setting was studied in the somatostatin receptor-expressing GOT1 xenograft model. The combination led to a larger decrease in tumour volume relative to monotherapies and the tumour-reducing effect was shown to be synergistic. Using patient-derived tumour cells from eight metastatic SINETs, we could show that ganetespib enhanced the effect of 177Lu-octreotate therapy for all investigated patient tumours. Levels of Hsp90 protein expression were evaluated in 767 SINETs from 379 patients. We found that Hsp90 expression was upregulated in tumour cells relative to tumour stroma in the vast majority of SINETs. We conclude that Hsp90 inhibitors enhance the tumour-killing effect of 177Lu-octreotate therapy synergistically in SINET tumour models and suggest that this potentially promising combination should be further evaluated.
Search for other papers by James Yao in
Google Scholar
PubMed
Search for other papers by Abhishek Garg in
Google Scholar
PubMed
Search for other papers by David Chen in
Google Scholar
PubMed
Search for other papers by Jaume Capdevila in
Google Scholar
PubMed
Search for other papers by Paul Engstrom in
Google Scholar
PubMed
Search for other papers by Rodney Pommier in
Google Scholar
PubMed
Search for other papers by Eric Van Cutsem in
Google Scholar
PubMed
Search for other papers by Simron Singh in
Google Scholar
PubMed
Search for other papers by Nicola Fazio in
Google Scholar
PubMed
Search for other papers by Wei He in
Google Scholar
PubMed
Search for other papers by Markus Riester in
Google Scholar
PubMed
Search for other papers by Parul Patel in
Google Scholar
PubMed
Search for other papers by Maurizio Voi in
Google Scholar
PubMed
Search for other papers by Michael Morrissey in
Google Scholar
PubMed
Search for other papers by Marianne Pavel in
Google Scholar
PubMed
Search for other papers by Matthew Helmut Kulke in
Google Scholar
PubMed
Neuroendocrine tumors (NETs) have historically been subcategorized according to histologic features and the site of anatomic origin. Here, we characterize the genomic alterations in patients enrolled in three phase 3 clinical trials of NET of different anatomic origins and assess the potential correlation with clinical outcomes. Whole-exome and targeted panel sequencing was used to characterize 225 NET samples collected in the RADIANT series of clinical trials. Genomic profiling of NET was analyzed along with nongenomic biomarker data on the tumor grade and circulating chromogranin A (CgA) and neuron-specific enolase (NSE) levels from these patients enrolled in clinical trials. Our results highlight recurrent large-scale chromosomal alterations as a common theme among NET. Although the specific pattern of chromosomal alterations differed between tumor subtypes, the evidence for generalized chromosomal instability (CIN) was observed across all primary sites of NET. In pancreatic NET, although the P value was not significant, higher CIN suggests a trend toward longer survival (HR, 0.55, P = 0.077), whereas in the gastrointestinal NET, lower CIN was associated with longer survival (HR, 0.44, P = 0.0006). Our multivariate analyses demonstrated that when combined with other clinical data among patients with progressive advanced NETs, chromosomal level alteration adds important prognostic information. Large-scale CIN is a common feature of NET, and specific patterns of chromosomal gain and loss appeared to have independent prognostic value in NET subtypes. However, whether CIN in general has clinical significance in NET requires validation in larger patient cohort and warrants further mechanistic studies.
Search for other papers by Fabia De Oliveira Andrade in
Google Scholar
PubMed
Search for other papers by Wei Yu in
Google Scholar
PubMed
Search for other papers by Xiyuan Zhang in
Google Scholar
PubMed
Search for other papers by Elissa Carney in
Google Scholar
PubMed
Search for other papers by Rong Hu in
Google Scholar
PubMed
Search for other papers by Robert Clarke in
Google Scholar
PubMed
Search for other papers by Kevin FitzGerald in
Google Scholar
PubMed
Search for other papers by Leena Hilakivi-Clarke in
Google Scholar
PubMed
Resistance to endocrine therapy remains a clinical challenge in the treatment of estrogen receptor-positive (ER+) breast cancer. We investigated if adding a traditional Asian herbal mixture consisting of 12 herbs, called Jaeumkanghwa-tang (JEKHT), to tamoxifen (TAM) therapy might prevent resistance and recurrence in the ER+ breast cancer model of 7,12-dimethylbenz[a]anthracene (DMBA)-exposed Sprague–Dawley rats. Rats were divided into four groups treated as follows: 15 mg/kg TAM administered via diet as TAM citrate (TAM only); 500 mg/kg JEKHT administered via drinking water (JEKHT only group); TAM + JEKHT and no treatment control group. The study was replicated using two different batches of JEKHT. In both studies, a significantly higher proportion of ER+ mammary tumors responded to TAM if animals also were treated with JEKHT (experiment 1: 47% vs 65%, P = 0.015; experiment 2: 43% vs 77%, P < 0.001). The risk of local recurrence also was reduced (31% vs 12%, P = 0.002). JEKHT alone was mostly ineffective. In addition, JEKHT prevented the development of premalignant endometrial lesions in TAM-treated rats (20% in TAM only vs 0% in TAM + JEKHT). Co-treatment of antiestrogen-resistant LCC9 human breast cancer cells with 1.6 mg/mL JEKHT reversed their TAM resistance in dose–response studies in vitro. Several traditional herbal medicine preparations can exhibit anti-inflammatory properties and may increase anti-tumor immune activities in the tumor microenvironment. In the tumors of rats treated with both JEKHT and TAM, expression of Il-6 (P = 0.03), Foxp3/T regulatory cell (Treg) marker (P = 0.033) and Tgfβ1 that activates Tregs (P < 0.001) were significantly downregulated compared with TAM only group. These findings indicate that JEKHT may prevent TAM-induced evasion of tumor immune responses.