Browse
Search for other papers by Marcia S Brose in
Google Scholar
PubMed
Search for other papers by Johannes Smit in
Google Scholar
PubMed
Search for other papers by Chia-Chi Lin in
Google Scholar
PubMed
Search for other papers by Fabian Pitoia in
Google Scholar
PubMed
Search for other papers by Marc Fellous in
Google Scholar
PubMed
Search for other papers by Yoriko DeSanctis in
Google Scholar
PubMed
Search for other papers by Martin Schlumberger in
Google Scholar
PubMed
Search for other papers by Masayuki Tori in
Google Scholar
PubMed
Search for other papers by Iwao Sugitani in
Google Scholar
PubMed
There are limited treatment options for patients with radioactive iodine refractory, progressive differentiated thyroid cancer. Although there is consensus that multikinase inhibitor therapy should be considered in patients with progressive disease with considerable tumor load or symptomatic disease, uncertainty exists on the optimal timing to treat with a multikinase inhibitor, especially for asymptomatic patients. RIFTOS MKI is an international, prospective, open-label, multicenter, noninterventional study with the primary objective to compare the time to symptomatic progression from study entry in asymptomatic patients with radioactive iodine refractory, progressive differentiated thyroid cancer for whom there is a decision to initiate multikinase inhibitors at study entry (cohort 1) with those for whom there is a decision to not initiate multikinase inhibitors at study entry (cohort 2). Secondary endpoints are overall survival and progression-free survival, which will be compared between cohorts 1 and 2. Additional secondary endpoints are postprogression survival from time of symptomatic progression, duration of and response to each systemic treatment regimen and dosing of sorafenib throughout the treatment period. Asymptomatic, multikinase inhibitor-naive patients aged ≥18 years with histologically/cytologically documented differentiated thyroid cancer that is radioactive iodine refractory are eligible. Patients may receive any therapy for differentiated thyroid cancer, including sorafenib or other multikinase inhibitors if indicated and decided on by the treating physician. In total, 700 patients are estimated to be enrolled from >20 countries. Final analysis will be performed once the last enrolled patient has been followed up with for 24 months (ClinicalTrials.gov identifier: Nbib2303444).
Cancer Biology Laboratory, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
Search for other papers by Andrew M K Law in
Google Scholar
PubMed
St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
Search for other papers by Elgene Lim in
Google Scholar
PubMed
St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
Search for other papers by Christopher J Ormandy in
Google Scholar
PubMed
St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
Search for other papers by David Gallego-Ortega in
Google Scholar
PubMed
A cancer cell-centric view has long dominated the field of cancer biology. Research efforts have focussed on aberrant cancer cell signalling pathways and on changes to cancer cell DNA. Mounting evidence demonstrates that many cancer-associated cell types within the tumour stroma co-evolve and support tumour growth and development, greatly modifying cancer cell behaviour, facilitating invasion and metastasis and controlling dormancy and sensitivity to drug therapy. Thus, these stromal cells represent potential targets for cancer therapy. Among these cell types, immune cells have emerged as a promising target for therapy. The adaptive and the innate immune system play an important role in normal mammary development and breast cancer. The number of infiltrating adaptive immune system cells with tumour-rejecting capacity, primarily, T lymphocytes, is lower in breast cancer compared with other cancer types, but infiltration occurs in a large proportion of cases. There is strong evidence demonstrating the importance of the immunosuppressive role of the innate immune system during breast cancer progression. A consideration of components of both the innate and the adaptive immune system is essential for the design and development of immunotherapies in breast cancer. In this review, we focus on the importance of immunosuppressive myeloid-derived suppressor cells (MDSCs) as potential targets for breast cancer therapy.
Search for other papers by K M Biernacka in
Google Scholar
PubMed
Search for other papers by R A Persad in
Google Scholar
PubMed
Search for other papers by A Bahl in
Google Scholar
PubMed
Search for other papers by D Gillatt in
Google Scholar
PubMed
Search for other papers by J M P Holly in
Google Scholar
PubMed
Search for other papers by C M Perks in
Google Scholar
PubMed
The incidence of many common cancers varies between different populations and appears to be affected by a Western lifestyle. Highly proliferative malignant cells require sufficient levels of nutrients for their anabolic activity. Therefore, targeting genes and pathways involved in metabolic pathways could yield future therapeutics. A common pathway implicated in energetic and nutritional requirements of a cell is the LKB1/AMPK pathway. Metformin is a widely studied anti-diabetic drug, which improves glycaemia in patients with type 2 diabetes by targeting this pathway. We investigated the effect of metformin on prostate cancer cell lines and evaluated its mechanism of action using DU145, LNCaP, PC3 and VCaP prostate cancer cell lines. Trypan blue dye-exclusion assay was used to assess levels of cell death. Western immunoblotting was used to determine the abundance of proteins. Insulin-like growth factor-binding protein-2 (IGFBP-2) and AMPK genes were silenced using siRNA. Effects on cell morphology were visualised using microscopy. IGFBP-2 gene expression was assessed using real-time RT-PCR. With DU145 and LNCaP cells metformin alone induced cell death, but this was reduced in hyperglycaemic conditions. Hyperglycaemia also reduced the sensitivity to Docetaxel, but this was countered by co-treatment with metformin. LKB1 was required for the activation of AMPK but was not essential to mediate the induction of cell death. An alternative pathway by which metformin exerted its action was through downregulation of IGFBP-2 in DU145 and LNCaP cells, independently of AMPK. This finding could have important implications in relation to therapeutic strategies in prostate cancer patients presenting with diabetes.
Search for other papers by John W Cassidy in
Google Scholar
PubMed
Search for other papers by Ankita S Batra in
Google Scholar
PubMed
Search for other papers by Wendy Greenwood in
Google Scholar
PubMed
Search for other papers by Alejandra Bruna in
Google Scholar
PubMed
Despite remarkable advances in our understanding of the drivers of human malignancies, new targeted therapies often fail to show sufficient efficacy in clinical trials. Indeed, the cost of bringing a new agent to market has risen substantially in the last several decades, in part fuelled by extensive reliance on preclinical models that fail to accurately reflect tumour heterogeneity. To halt unsustainable rates of attrition in the drug discovery process, we must develop a new generation of preclinical models capable of reflecting the heterogeneity of varying degrees of complexity found in human cancers. Patient-derived tumour xenograft (PDTX) models prevail as arguably the most powerful in this regard because they capture cancer’s heterogeneous nature. Herein, we review current breast cancer models and their use in the drug discovery process, before discussing best practices for developing a highly annotated cohort of PDTX models. We describe the importance of extensive multidimensional molecular and functional characterisation of models and combination drug–drug screens to identify complex biomarkers of drug resistance and response. We reflect on our own experiences and propose the use of a cost-effective intermediate pharmacogenomic platform (the PDTX-PDTC platform) for breast cancer drug and biomarker discovery. We discuss the limitations and unanswered questions of PDTX models; yet, still strongly envision that their use in basic and translational research will dramatically change our understanding of breast cancer biology and how to more effectively treat it.
Search for other papers by Ju-Yeon Moon in
Google Scholar
PubMed
Search for other papers by Man Ho Choi in
Google Scholar
PubMed
Department of Medicine, University of California, Los Angeles, California, USA
Search for other papers by Jayoung Kim in
Google Scholar
PubMed
Cholesterol and sex steroid hormones including androgens and estrogens play a critical role in the development and progression of urological diseases such as prostate cancer. This disease remains the most commonly diagnosed malignant tumor in men and is the leading cause of death from different cancers. Attempts to understand the role of cholesterol and steroid metabolism in urological diseases have been ongoing for many years, but despite this, our mechanistic and translational understanding remains elusive. In order to further evaluate the problem, we have taken an interest in metabolomics; a discipline dedicated to the systematic study of biologically active metabolites in cells, tissues, hair and biofluids. Recently, we provided evidence that a quantitative measurement of cholesterol and sex steroid metabolites can be successfully achieved using hair of human and mouse models. The overall goal of this short review article is to introduce current metabolomic technologies for the quantitative biomarker assay development and also to provide new insight into understanding the underlying mechanisms that trigger the pathological condition. Furthermore, this review will place a particular emphasis on how to prepare biospecimens (e.g., hair fiber), quantify molecular profiles and assess their clinical significance in various urological diseases.
Search for other papers by Sakine Sever in
Google Scholar
PubMed
Clinical Epidemiology and Comparative Effectiveness Program, Section of Health Services Research (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, HSR&D Center of Innovation (152), Houston, Texas, USA
Texas Medical Center Digestive Disease Center, Baylor College of Medicine, Houston, Texas, USA
Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
Search for other papers by Donna L White in
Google Scholar
PubMed
Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington, USA
Search for other papers by José M Garcia in
Google Scholar
PubMed
Ghrelin is a hormone with multiple physiologic functions, including promotion of growth hormone release, stimulation of appetite and regulation of energy homeostasis. Treatment with ghrelin/ghrelin-receptor agonists is a prospective therapy for disease-related cachexia and malnutrition. In vitro studies have shown high expression of ghrelin in cancer tissue, although its role including its impact in cancer risk and progression has not been established. We performed a systematic literature review to identify peer-reviewed human or animal in vivo original research studies of ghrelin, ghrelin-receptor agonists, or ghrelin genetic variants and the risk, presence, or growth of cancer using structured searches in PubMed database as well as secondary searches of article reference lists, additional reviews and meta-analyses. Overall, 45 (73.8%) of the 61 studies reviewed, including all 11 involving exogenous ghrelin/ghrelin-receptor agonist treatment, reported either a null (no statistically significant difference) or inverse association of ghrelin/ghrelin-receptor agonists or ghrelin genetic variants with cancer risk, presence or growth; 10 (16.7%) studies reported positive associations; and 6 (10.0%) reported both negative or null and positive associations. Differences in serum ghrelin levels in cancer cases vs controls (typically lower) were reported for some but not all cancers. The majority of in vivo studies showed a null or inverse association of ghrelin with risk and progression of most cancers, suggesting that ghrelin/ghrelin-receptor agonist treatment may have a favorable safety profile to use for cancer cachexia. Additional large-scale prospective clinical trials as well as basic bioscientific research are warranted to further evaluate the safety and benefits of ghrelin treatment in patients with cancer.
Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
Search for other papers by Hyun Ho Han in
Google Scholar
PubMed
Search for other papers by Baek Gil Kim in
Google Scholar
PubMed
Search for other papers by Joo Hyun Lee in
Google Scholar
PubMed
Search for other papers by Suki Kang in
Google Scholar
PubMed
Search for other papers by Ji Eun Kim in
Google Scholar
PubMed
Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
Severance Biomedical Science Institute (SBSI), Yonsei University College of Medicine, Seoul, South Korea
Global 5-5-10 System Biology, Yonsei University, Seoul, South Korea
Search for other papers by Nam Hoon Cho in
Google Scholar
PubMed
In estrogen receptor-positive (ER+) breast cancer, it is recognized that metastases may develop after a long period of dormancy. Bone marrow (BM) vascular niche is where the dormant tumor cells are most likely to reside. So far, it is not fully understood why the dormant tumor cells become proliferative and eventually generate tumor. We hypothesized that therapeutic or menopause-related estrogen depletion may be the switch behind dormant ER+ tumor cell awakening in BM. We utilized an existing experimental model of BM endothelial niche that can simulate ER+ tumor cell dormancy to test our hypothesis. In results, estrogen depletion paradoxically promoted ER+ tumor cell proliferation in the BM endothelial niche, and their molecular phenotype shifted from dormant to awaken. Following estrogen depletion, the BM niche cells produced angiopoietin-2 (ANGPT2), which destabilized niche endothelium by interfering ANGPT1/Tie2 signaling, and promoted ER+ tumor cell survival under estrogen deficiency via cell surface integrin β1. Knockdown of ANGPT2 completely negated ER+ tumor cell awakening in the niche. Furthermore, ANGPT2 expression in ER+ tumor human samples was associated with increased risk of distant metastasis only in those who underwent adjuvant estrogen depletion therapy, not in those who did not undergo adjuvant therapy. In conclusion, we demonstrate that ANGPT2 signaling activated after estrogen depletion paradoxically triggers ER+ tumor cell awakening from dormancy in their BM niche, partly indirectly via endothelial Tie2 receptor and partly directly via tumor cell surface integrin β1.
Search for other papers by Milena Rondón-Lagos in
Google Scholar
PubMed
Natural and Mathematical Sciences Faculty, Universidad del Rosario, Bogotá, Colombia
Search for other papers by Nelson Rangel in
Google Scholar
PubMed
Search for other papers by Ludovica Verdun Di Cantogno in
Google Scholar
PubMed
Search for other papers by Laura Annaratone in
Google Scholar
PubMed
Search for other papers by Isabella Castellano in
Google Scholar
PubMed
Search for other papers by Rosalia Russo in
Google Scholar
PubMed
Search for other papers by Tilde Manetta in
Google Scholar
PubMed
Search for other papers by Caterina Marchiò in
Google Scholar
PubMed
Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
Search for other papers by Anna Sapino in
Google Scholar
PubMed
Evidence supports a role of 17&-estradiol (E2) in carcinogenesis and the large majority of breast carcinomas are dependent on estrogen. The anti-estrogen tamoxifen (TAM) is widely used for both treatment and prevention of breast cancer; however, it is also carcinogenic in human uterus and rat liver, highlighting the profound complexity of its actions. The nature of E2- or TAM-induced chromosomal damage has been explored using relatively high concentrations of these agents, and only some numerical aberrations and chromosomal breaks have been analyzed. This study aimed to determine the effects of low doses of E2 and TAM (10&8 mol L&1 and 10&6 mol L&1 respectively) on karyotypes of MCF7, T47D, BT474, and SKBR3 breast cancer cells by comparing the results of conventional karyotyping and multi-FISH painting with cell proliferation. Estrogen receptor (ER)-positive (+) cells showed an increase in cell proliferation after E2 treatment (MCF7, T47D, and BT474) and a decrease after TAM treatment (MCF7 and T47D), whereas in ER& cells (SKBR3), no alterations in cell proliferation were observed, except for a small increase at 96 h. Karyotypes of both ER+ and ER& breast cancer cells increased in complexity after treatments with E2 and TAM leading to specific chromosomal abnormalities, some of which were consistent throughout the treatment duration. This genotoxic effect was higher in HER2+ cells. The ER&/HER2+ SKBR3 cells were found to be sensitive to TAM, exhibiting an increase in chromosomal aberrations. These in vitro results provide insights into the potential role of low doses of E2 and TAM in inducing chromosomal rearrangements in breast cancer cells.
Search for other papers by Thomas Cuny in
Google Scholar
PubMed
Search for other papers by Caroline Zeiller in
Google Scholar
PubMed
Search for other papers by Martin Bidlingmaier in
Google Scholar
PubMed
Search for other papers by Céline Défilles in
Google Scholar
PubMed
Search for other papers by Catherine Roche in
Google Scholar
PubMed
Search for other papers by Marie-Pierre Blanchard in
Google Scholar
PubMed
Search for other papers by Marily Theodoropoulou in
Google Scholar
PubMed
APHM Timone, Department of Neurosurgery, Marseille, France
Search for other papers by Thomas Graillon in
Google Scholar
PubMed
Search for other papers by Morgane Pertuit in
Google Scholar
PubMed
Search for other papers by Dominique Figarella-Branger in
Google Scholar
PubMed
APHM, Conception, Laboratory of Molecular Biology, Marseille, France
Search for other papers by Alain Enjalbert in
Google Scholar
PubMed
APHM Conception, Department of Endocrinology, Marseille, France
Search for other papers by Thierry Brue in
Google Scholar
PubMed
APHM, Conception, Laboratory of Molecular Biology, Marseille, France
Search for other papers by Anne Barlier in
Google Scholar
PubMed
Pegvisomant (PEG), an antagonist of growth hormone (GH)-receptor (GHR), normalizes insulin-like growth factor 1 (IGF1) oversecretion in most acromegalic patients unresponsive to somatostatin analogs (SSAs) and/or uncontrolled by transsphenoidal surgery. The residual GH-secreting tumor is therefore exposed to the action of circulating PEG. However, the biological effect of PEG at the pituitary level remains unknown. To assess the impact of PEG in vitro on the hormonal secretion (GH and prolactin (PRL)), proliferation and cellular viability of eight human GH-secreting tumors in primary cultures and of the rat somatolactotroph cell line GH4C1. We found that the mRNA expression levels of GHR were characterized in 31 human GH-secreting adenomas (0.086 copy/copy β-Gus) and the GHR was identified by immunocytochemistry staining. In 5/8 adenomas, a dose-dependent inhibition of GH secretion was observed under PEG with a maximum of 38.2±17% at 1μg/mL (P<0.0001 vs control). A dose-dependent inhibition of PRL secretion occurred in three mixed GH/PRL adenomas under PEG with a maximum of 52.8±11.5% at 10μg/mL (P<0.0001 vs control). No impact on proliferation of either human primary tumors or GH4C1 cell line was observed. We conclude that PEG inhibits the secretion of GH and PRL in primary cultures of human GH(/PRL)-secreting pituitary adenomas without effect on cell viability or cell proliferation.
Search for other papers by Birgit Luef in
Google Scholar
PubMed
Search for other papers by Florian Handle in
Google Scholar
PubMed
Search for other papers by Gvantsa Kharaishvili in
Google Scholar
PubMed
Search for other papers by Martina Hager in
Google Scholar
PubMed
Search for other papers by Johannes Rainer in
Google Scholar
PubMed
Search for other papers by Günter Janetschek in
Google Scholar
PubMed
Search for other papers by Stephan Hruby in
Google Scholar
PubMed
Search for other papers by Christine Englberger in
Google Scholar
PubMed
Search for other papers by Jan Bouchal in
Google Scholar
PubMed
Search for other papers by Frédéric R Santer in
Google Scholar
PubMed
Search for other papers by Zoran Culig in
Google Scholar
PubMed
Due to the urgent need for new prostate cancer (PCa) therapies, the role of androgen receptor (AR)-interacting proteins should be investigated. In this study we aimed to address whether the AR coactivator nuclear receptor coactivator 1 (NCOA1) is involved in PCa progression. Therefore, we tested the effect of long-term NCOA1 knockdown on processes relevant to metastasis formation. [3H]-thymidine incorporation assays revealed a reduced proliferation rate in AR-positive MDA PCa 2b and LNCaP cells upon knockdown of NCOA1, whereas AR-negative PC3 cells were not affected. Furthermore, Boyden chamber assays showed a strong decrease in migration and invasion upon NCOA1 knockdown, independently of the cell line’s AR status. In order to understand the mechanistic reasons for these changes, transcriptome analysis using cDNA microarrays was performed. Protein kinase D1 (PRKD1) was found to be prominently up-regulated by NCOA1 knockdown in MDA PCa 2b, but not in PC3 cells. Inhibition of PRKD1 reverted the reduced migratory potential caused by NCOA1 knockdown. Furthermore, PRKD1 was negatively regulated by AR. Immunohistochemical staining of PCa patient samples revealed a strong increase in NCOA1 expression in primary tumors compared with normal prostate tissue, while no final conclusion could be drawn for PRKD1 expression in tumor specimens. Thus, our findings directly associate the AR/NCOA1 complex with PRKD1 regulation and cellular migration and support the concept of therapeutic inhibition of NCOA1 in PCa.