36
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Metformin as an Adjuvant Drug against Pediatric Sarcomas: Hypoxia Limits Therapeutic Effects of the Drug

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Metformin, a well-known insulin-sensitizer commonly used for type 2 diabetes therapy, has recently emerged as potentially very attractive drug also in oncology. It is cheap, it is relatively safe and many reports have indicated effects in cancer prevention and therapy. These desirable features are particularly interesting for pediatric sarcomas, a group of rare tumors that have been shown to be dependent on IGF and insulin system for pathogenesis and progression. Metformin exerts anti-mitogenic activity in several cancer histotypes through several molecular mechanisms. In this paper, we analyzed its effects against osteosarcoma, Ewing sarcoma and rhabdomyosarcoma, the three most common pediatric sarcomas. Despite in vitro metformin gave remarkable antiproliferative and chemosensitizing effects both in sensitive and chemoresistant cells, its efficacy was not confirmed against Ewing sarcoma xenografts neither as single agent nor in combination with vincristine. This discrepancy between in vitro and in vivo effects may be due to hypoxia, a common feature of solid tumors. We provide evidences that in hypoxia conditions metformin was not able to activate AMPK and inhibit mTOR signaling, which likely prevents the inhibitory effects of metformin on tumor growth. Thus, although metformin may be considered a useful complement of conventional chemotherapy in normoxia, its therapeutic value in highly hypoxic tumors may be more limited. The impact of hypoxia should be considered when novel therapies are planned for pediatric sarcomas.

          Related collections

          Most cited references52

          • Record: found
          • Abstract: found
          • Article: not found

          The insulin and insulin-like growth factor receptor family in neoplasia: an update.

          Although several early phase clinical trials raised enthusiasm for the use of insulin-like growth factor I receptor (IGF1R)-specific antibodies for cancer treatment, initial Phase III results in unselected patients have been disappointing. Further clinical studies may benefit from the use of predictive biomarkers to identify probable responders, the use of rational combination therapies and the consideration of alternative targeting strategies, such as ligand-specific antibodies and receptor-specific tyrosine kinase inhibitors. Targeting insulin and IGF signalling also needs to be considered in the broader context of the pathophysiology that relates obesity and diabetes to neoplasia, and the effects of anti-diabetic drugs, including metformin, on cancer risk and prognosis. The insulin and IGFI receptor family is also relevant to the development of PI3K-AKT pathway inhibitors.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease.

            In mammals, the insulin receptor (IR) gene has acquired an additional exon, exon 11. This exon may be skipped in a developmental and tissue-specific manner. The IR, therefore, occurs in two isoforms (exon 11 minus IR-A and exon 11 plus IR-B). The most relevant functional difference between these two isoforms is the high affinity of IR-A for IGF-II. IR-A is predominantly expressed during prenatal life. It enhances the effects of IGF-II during embryogenesis and fetal development. It is also significantly expressed in adult tissues, especially in the brain. Conversely, IR-B is predominantly expressed in adult, well-differentiated tissues, including the liver, where it enhances the metabolic effects of insulin. Dysregulation of IR splicing in insulin target tissues may occur in patients with insulin resistance; however, its role in type 2 diabetes is unclear. IR-A is often aberrantly expressed in cancer cells, thus increasing their responsiveness to IGF-II and to insulin and explaining the cancer-promoting effect of hyperinsulinemia observed in obese and type 2 diabetic patients. Aberrant IR-A expression may favor cancer resistance to both conventional and targeted therapies by a variety of mechanisms. Finally, IR isoforms form heterodimers, IR-A/IR-B, and hybrid IR/IGF-IR receptors (HR-A and HR-B). The functional characteristics of such hybrid receptors and their role in physiology, in diabetes, and in malignant cells are not yet fully understood. These receptors seem to enhance cell responsiveness to IGFs.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1.

              Metformin is a widely prescribed antidiabetic drug associated with a reduced risk of cancer. Many studies show that metformin inhibits cancer cell viability through the inhibition of mTOR. We recently showed that antiproliferative action of metformin in prostate cancer cell lines is not mediated by AMP-activated protein kinase (AMPK). We identified REDD1 (also known as DDIT4 and RTP801), a negative regulator of mTOR, as a new molecular target of metformin. We show that metformin increases REDD1 expression in a p53-dependent manner. REDD1 invalidation, using siRNA or REDD1(-/-) cells, abrogates metformin inhibition of mTOR. Importantly, inhibition of REDD1 reverses metformin-induced cell-cycle arrest and significantly protects from the deleterious effects of metformin on cell transformation. Finally, we show the contribution of p53 in mediating metformin action in prostate cancer cells. These results highlight the p53/REDD1 axis as a new molecular target in anticancer therapy in response to metformin treatment. ©2011 AACR.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS One
                PLoS ONE
                plos
                plosone
                PLoS ONE
                Public Library of Science (San Francisco, USA )
                1932-6203
                2013
                31 December 2013
                : 8
                : 12
                : e83832
                Affiliations
                [1 ]CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
                [2 ]Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
                [3 ]Department of Health, University of Catanzaro, Catanzaro, Italy
                [4 ]Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
                University of Navarra, Spain
                Author notes

                Competing Interests: The anti-IGF-IR drugs used in this study were kindly provided by: Immunogen (AVE1642 HAb; Waltham, AM), Pfizer (CP-751,871/Figitumumab; San Diego, California), and Novartis (NVP-AEW541 and NVP-BEZ235; Basel, Switzerland). An ifosfamide analog not requiring metabolic activation used in this study was kindly provided by Baxter Oncology GmbH (Frankfurt, Germany), and Actinomycin-D was provided by Ovation Healthcare International (Dublin, Ireland). There are no further patents, products in development or marketed products to declare. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

                Conceived and designed the experiments: CG KS. Performed the experiments: CG MC MCM CM LL. Analyzed the data: CG MC MCM CM LL AB KS. Contributed reagents/materials/analysis tools: KS PP PLL AB. Wrote the paper: CG KS.

                Article
                PONE-D-13-30753
                10.1371/journal.pone.0083832
                3877110
                24391834
                11dbb885-e99e-44a5-9a54-2e7f34d6793f
                Copyright @ 2013

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 29 July 2013
                : 8 November 2013
                Page count
                Pages: 12
                Funding
                This work was funded by: Italian Association for Cancer Research grant (MFAG 11584 to CG; IG 10452 to KS; IG 10353 to PLL); The Italian Ministry of Health (Ricerca Finalizzata 2009 to KS 1628/2010); The Italian Ministry of Research and Instruction grant PRIN2009 cod. SFC2EK to KS. PRIN2009 to PLL. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Medicine
                Clinical Research Design
                Preclinical Models
                Drugs and Devices
                Drug Research and Development
                Endocrinology
                Diabetic Endocrinology
                Diabetes Mellitus Type 2
                Insulin
                Oncology
                Cancers and Neoplasms
                Bone and Soft Tissue Sarcomas
                Basic Cancer Research
                Pediatric Oncology

                Uncategorized
                Uncategorized

                Comments

                Comment on this article