101
|
Hvilsom GB, Londero SC, Hahn CH, Schytte S, Pedersen HB, Christiansen P, Kiss K, Larsen SR, Jespersen ML, Lelkaitis G, Godballe C. Anaplastic thyroid carcinoma in Denmark 1996-2012: A national prospective study of 219 patients. Cancer Epidemiol 2018; 53:65-71. [PMID: 29414634 DOI: 10.1016/j.canep.2018.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is the least common but most malignant thyroid cancer. We aimed to examine the characteristics as well as evaluate the incidence, prognostic factors, and if introduction of a fast track cancer program might influence survival in a cohort of ATC patients. METHODS A cohort study based on prospective data from the national Danish thyroid cancer database DATHYRCA and the national Danish Pathology Register including 219 patients diagnosed from 1996 to 2012, whom were followed until death or through September 2014. RESULTS We found the median age in the 7th decade, the majority of patients being women presenting with a growing mass at the neck, diagnosed with stage T4b disease. At diagnosis, 56% of the patients had lymph node metastasis and 38% distant metastasis. We observed one- and five-year survival of 20.7% and 11.0%, respectively. Both univariate and multivariate analyses showed age (above 73.6 years), respiratory impairment, T4b stage, and distant metastasis at diagnosis to be significant prognostic factors. Further, introduction of a national fast track cancer program increased survival nearly two-fold. CONCLUSION As new information, our study adds "respiratory impairment at diagnosis" and "introduction of a national fast track cancer program" to the list of already established prognostic indicators for ATC.
Collapse
Affiliation(s)
- Gitte Bjørn Hvilsom
- Odense University Hospital, Department of ENT, Head & Neck Surgery, Sdr Boulevard 29, 5000 Odense C, Denmark.
| | - Stefano Christian Londero
- Odense University Hospital, Department of ENT, Head & Neck Surgery, Sdr Boulevard 29, 5000 Odense C, Denmark; Aarhus University Hospital, Department of ENT Head & Neck Surgery, Nørrebrogade 44, 8000 Aarhus C, Denmark.
| | - Christoffer Holst Hahn
- Copenhagen University Hospital, Rigshospitalet, Department of ENT Head & Neck Surgery, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Sten Schytte
- Aarhus University Hospital, Department of ENT Head & Neck Surgery, Nørrebrogade 44, 8000 Aarhus C, Denmark.
| | - Henrik Baymler Pedersen
- Aalborg University Hospital, Syd, Department of ENT Head & Neck Surgery, Hobrovej 18-22, 9000 Aalborg, Denmark.
| | - Peer Christiansen
- Aarhus University Hospital, Department of Surgery, Tage-Hansens Gade 2, 8000 Aarhus C, Denmark.
| | - Katalin Kiss
- Copenhagen University Hospital, Rigshospitalet, Department of Pathology, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Stine Rosenkilde Larsen
- Odense University Hospital, Department of Pathology, Sdr Boulevard 29, 5000 Odense C, Denmark.
| | - Marie Louise Jespersen
- Aarhus University Hospital, Department of Pathology, Tage-Hansens Gade 2, 8000 Aarhus C, Denmark.
| | - Giedrius Lelkaitis
- Aalborg University Hospital, Nord, Department of Pathology, Ladegårdsgade 3, 9000 Aalborg, Denmark.
| | - Christian Godballe
- Odense University Hospital, Department of ENT, Head & Neck Surgery, Sdr Boulevard 29, 5000 Odense C, Denmark.
| |
Collapse
|
102
|
Oh JM, Kalimuthu S, Gangadaran P, Baek SH, Zhu L, Lee HW, Rajendran RL, Hong CM, Jeong SY, Lee SW, Lee J, Ahn BC. Reverting iodine avidity of radioactive-iodine refractory thyroid cancer with a new tyrosine kinase inhibitor (K905-0266) excavated by high-throughput NIS (sodium iodide symporter) enhancer screening platform using dual reporter gene system. Oncotarget 2018; 9:7075-7087. [PMID: 29467951 PMCID: PMC5805537 DOI: 10.18632/oncotarget.24159] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/21/2017] [Indexed: 11/25/2022] Open
Abstract
Radioactive-iodine (RAI) therapy is typically unprevailing as anaplastic thyroid cancer (ATC) management, owing to the decrease in the endogenous sodium iodide symporter (NIS) expression. Therefore, new strategies for NIS re-induction are required to improve the efficacy of RAI therapy in ATC. In this study, we developed a novel high-throughput NIS enhancer screening platform using a dual reporter gene system to identify a potent tyrosine kinase inhibitor (TKI) and selected a new hit compound, K905-0266 TKI. The effects of K905-0266 TKI treatment was validated as RAI accumulation, changes in signalling pathway related to thyroid pathogenesis, and cytotoxicity of RAI depending on re-induction of endogenous NIS expression in ATC. Furthermore, we evaluated enhancement of NIS promoter and therapeutic efficacy of RAI in ATC tumour xenograft mice. After K905-0266 TKI treatment, the expression of endogenous NIS was significantly increased, while phosphorylated-ERK was decreased. In addition, the thyroid-metabolising protein expressions were upregulated and increased of RAI accumulation and its therapeutic effects in ATC. Moreover, K905-0266 TKI increased therapeutic efficacy of RAI in ATC tumour in vivo. In conclusion, we successfully established a novel high-throughput NIS enhancer screening platform to excavate a NIS enhancer and identified K905-0266 TKI among TKI candidates and it's proven to increase the endogenous NIS expression and therapeutic efficacy of RAI in ATC. These findings suggest that a novel high-throughput NIS enhancer screening platform is useful for selecting of NIS promoter enhancers. In addition, K905-0266 TKI can be used to re-induce endogenous NIS expression and recover RAI therapy in ATC.
Collapse
Affiliation(s)
- Ji Min Oh
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Liya Zhu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University School of Medicine and Hospital, Daegu 41944, Republic of Korea
| |
Collapse
|
103
|
A selective cyclin-dependent kinase 4, 6 dual inhibitor, Ribociclib (LEE011) inhibits cell proliferation and induces apoptosis in aggressive thyroid cancer. Cancer Lett 2018; 417:131-140. [PMID: 29306020 DOI: 10.1016/j.canlet.2017.12.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/16/2017] [Accepted: 12/21/2017] [Indexed: 12/23/2022]
Abstract
The RB-E2F1 pathway is an important mechanism of cell-cycle control, and deregulation of this pathway is one of the key factors contributing to tumorigenesis. Cyclin-dependent kinases (CDKs) and Cyclin D have been known to increase in aggressive thyroid cancer. However, there has been no study to investigate effects of a selective CDK 4/6 inhibitor, Ribociclib (LEE011), in thyroid cancer. Performing Western blotting, we found that RB phosphorylation and the expression of Cyclin D are significantly higher in papillary thyroid cancer (PTC) cell lines as well as anaplastic thyroid cancer (ATC) cell lines, compared with normal thyroid cell line and follicular thyroid cancer cell line. LEE011 dose-dependently inhibited RB phosphorylation and also decreased the expressions of its target genes such as FOXM1, Cyclin A1, and Myc in ATC. Furthermore, LEE011 induced cell cycle arrest in G0-G1 phase and cell apoptosis, and inhibited cell proliferation in ATC. Consistently, oral administration of LEE011 to ATC xenograft models strongly inhibited tumor growth with decreased expressions of pRB, pAKT and Ki-67, and also significantly increased tumor cell apoptosis. Taken together, our data support the rationale for clinical development of the CDK4/6 inhibitor as a therapy for patients with aggressive thyroid cancer.
Collapse
|
104
|
Ahmed S, Ghazarian MP, Cabanillas ME, Zafereo ME, Williams MD, Vu T, Schomer DF, Debnam JM. Imaging of Anaplastic Thyroid Carcinoma. AJNR Am J Neuroradiol 2017; 39:547-551. [PMID: 29242360 DOI: 10.3174/ajnr.a5487] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/22/2017] [Indexed: 02/02/2023]
Abstract
Anaplastic thyroid carcinoma is fatal if unresectable. However, improved survival has been reported after gross total resection and multimodality therapy. In this report, we describe the contrast-enhanced high-resolution CT characteristics of anaplastic thyroid carcinoma in 57 patients. Anaplastic thyroid carcinoma presented as a large neck mass with necrosis in 82% of cases. The tumors demonstrated common extrathyroidal extension (91%). Sixty-two percent of tumors demonstrated calcification. Visceral space invasion involved the esophagus (62%), trachea (57%), and larynx (29%). Carotid artery encasement was present in 42%, and 43% involved the internal jugular vein. Sixty-three percent had lateral compartment lymphadenopathy; 58% of these nodes were necrotic, and 11% were cystic. No metastatic nodes had calcification. Central compartment lymphadenopathy was seen in 56% of cases, and lateral retropharyngeal lymphadenopathy was detected in 12%. Knowledge of these imaging features aids in guiding the approach to the initial tissue diagnosis with either fine-needle aspiration or core biopsy, assessing the feasibility of surgical resection, and determining prognosis.
Collapse
Affiliation(s)
- S Ahmed
- From the Departments of Diagnostic Radiology (S.A., T.V., D.F.S., J.M.D.)
| | - M P Ghazarian
- Department of Diagnostic and Interventional Imaging (M.P.G.), University of Texas Houston Medical School, Houston, Texas
| | | | | | - M D Williams
- Pathology (M.D.W.), University of Texas MD Anderson Cancer Center, Houston, Texas
| | - T Vu
- From the Departments of Diagnostic Radiology (S.A., T.V., D.F.S., J.M.D.)
| | - D F Schomer
- From the Departments of Diagnostic Radiology (S.A., T.V., D.F.S., J.M.D.)
| | - J M Debnam
- From the Departments of Diagnostic Radiology (S.A., T.V., D.F.S., J.M.D.)
| |
Collapse
|
105
|
Schmohl KA, Dolp P, Schug C, Knoop K, Klutz K, Schwenk N, Bartenstein P, Nelson PJ, Ogris M, Wagner E, Spitzweg C. Reintroducing the Sodium-Iodide Symporter to Anaplastic Thyroid Carcinoma. Thyroid 2017; 27:1534-1543. [PMID: 29032724 DOI: 10.1089/thy.2017.0290] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC), the most aggressive form of thyroid cancer, is unresponsive to radioiodine therapy. The current study aimed to extend the diagnostic and therapeutic application of radioiodine beyond the treatment of differentiated thyroid cancer by targeting the functional sodium-iodide symporter (NIS) to ATC. METHODS The study employed nanoparticle vectors (polyplexes) based on linear polyethylenimine (LPEI), shielded by polyethylene glycol (PEG) and coupled to the synthetic peptide GE11 as an epidermal growth factor receptor (EGFR)-specific ligand in order to target a NIS-expressing plasmid (LPEI-PEG-GE11/NIS) to EGFR overexpressing human thyroid carcinoma cell lines. Using ATC xenograft mouse models, transfection efficiency by 123I scintigraphy and potential for systemic radioiodine therapy after systemic polyplex application were evaluated. RESULTS In vitro iodide uptake studies in SW1736 and Hth74 ATC cells, and, for comparison, in more differentiated follicular (FTC-133) and papillary (BCPAP) thyroid carcinoma cells demonstrated high transfection efficiency and EGFR-specificity of LPEI-PEG-GE11/NIS that correlated well with EGFR expression levels. After systemic polyplex injection, in vivo 123I gamma camera imaging revealed significant tumor-specific accumulation of radioiodine in an SW1736 and an Hth74 xenograft mouse model. Radioiodine accumulation was found to be higher in SW1736 tumors, reflecting in vitro results, EGFR expression levels, and results from ex vivo analysis of NIS staining. Administration of 131I in LPEI-PEG-GE11/NIS-treated SW1736 xenograft mice resulted in significantly reduced tumor growth associated with prolonged survival compared to control animals. CONCLUSIONS The data open the exciting prospect of NIS-mediated radionuclide imaging and therapy of ATC after non-viral reintroduction of the NIS gene. The high tumor specificity after systemic application makes the strategy an attractive alternative for the treatment of highly metastatic ATC.
Collapse
Affiliation(s)
- Kathrin A Schmohl
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Patrick Dolp
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Christina Schug
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kerstin Knoop
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kathrin Klutz
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Nathalie Schwenk
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Peter Bartenstein
- 2 Department of Nuclear Medicine, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Peter J Nelson
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Manfred Ogris
- 3 Department of Pharmaceutical Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna , Vienna, Austria
| | - Ernst Wagner
- 4 Department of Pharmaceutical Biotechnology, Department of Pharmacy, Center for System-Based Drug Research and Center for Nanoscience , LMU Munich, Munich, Germany
| | - Christine Spitzweg
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| |
Collapse
|
106
|
Mishall KM, Beadnell TC, Kuenzi BM, Klimczak DM, Superti-Furga G, Rix U, Schweppe RE. Sustained activation of the AKT/mTOR and MAP kinase pathways mediate resistance to the Src inhibitor, dasatinib, in thyroid cancer. Oncotarget 2017; 8:103014-103031. [PMID: 29262541 PMCID: PMC5732707 DOI: 10.18632/oncotarget.20488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/23/2017] [Indexed: 12/11/2022] Open
Abstract
New targeted therapies are needed for advanced thyroid cancer. Our lab has shown that Src is a key mediator of tumorigenic processes in thyroid cancer. However, single-agent Src inhibitors have had limited efficacy in solid tumors. In order to more effectively target Src in the clinic, our lab has previously generated four thyroid cancer cell lines that are resistant to dasatinib through gradual dose escalation. We further tested two additional Src inhibitors and shown the dasatinib-resistant (DasRes) cells exhibit cross-resistance to saracatinib, but are sensitive to bosutinib, suggesting that unique off-targets of bosutinib play an important role in mediating sensitivity to bosutinib. To identify the kinases targeted by dasatinib and bosutinib, we utilized an unbiased compound centric chemical proteomics screen. We identified 33 kinases that were enriched in the bosutinib pull down. Using the STRING database to map protein-protein interactions of the unique bosutinib targets, we identified a signaling axis which included mTOR, FAK, and MEK. Inhibition of the mTOR, MEK, and Src/FAK nodes simultaneously was the most effective at reducing cell growth and survival. Overall, these studies have identified key mediators of Src inhibitor resistance, and show that targeting these signaling nodes are necessary for anti-tumor efficacy.
Collapse
Affiliation(s)
- Katie M. Mishall
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas C. Beadnell
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brent M. Kuenzi
- Department of Drug Discovery, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Cancer Biology PhD Program, University of South Florida, Tampa, Florida, USA
| | - Dorothy M. Klimczak
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Uwe Rix
- Department of Drug Discovery, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Rebecca E. Schweppe
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
107
|
Zhang L, Boufraqech M, Lake R, Kebebew E. Carfilzomib potentiates CUDC-101-induced apoptosis in anaplastic thyroid cancer. Oncotarget 2017; 7:16517-28. [PMID: 26934320 PMCID: PMC4941332 DOI: 10.18632/oncotarget.7760] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/08/2016] [Indexed: 12/19/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is one of the most aggressive human malignancies, with no effective treatment currently available. Previously, we identified agents active against ATC cells, both in vitro and in vivo, using quantitative high-throughput screening of 3282 clinically approved drugs and small molecules. Here, we report that combining two of these active agents, carfilzomib, a second-generation proteasome inhibitor, and CUDC-101, a histone deacetylase and multi-kinase inhibitor, results in increased, synergistic activity in ATC cells. The combination of carfilzomib and CUDC-101 synergistically inhibited cellular proliferation and caused cell death in multiple ATC cell lines harboring various driver mutations observed in human ATC tumors. This increased anti-ATC effect was associated with a synergistically enhanced G2/M cell cycle arrest and increased caspase 3/7 activity induced by the drug combination. Mechanistically, treatment with carfilzomib and CUDC-101 increased p21 expression and poly (ADP-ribose) polymerase protein cleavage. Our results suggest that combining carfilzomib and CUDC-101 would offer an effective therapeutic strategy to treat ATC.
Collapse
Affiliation(s)
- Lisa Zhang
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Myriem Boufraqech
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ross Lake
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Electron Kebebew
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
108
|
Molinaro E, Romei C, Biagini A, Sabini E, Agate L, Mazzeo S, Materazzi G, Sellari-Franceschini S, Ribechini A, Torregrossa L, Basolo F, Vitti P, Elisei R. Anaplastic thyroid carcinoma: from clinicopathology to genetics and advanced therapies. Nat Rev Endocrinol 2017; 13:644-660. [PMID: 28707679 DOI: 10.1038/nrendo.2017.76] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare malignancy, accounting for 1-2% of all thyroid cancers. Although rare, ATC accounts for the majority of deaths from thyroid carcinoma. ATC often originates in a pre-existing thyroid cancer lesion, as suggested by the simultaneous presence of areas of differentiated or poorly differentiated thyroid carcinoma. ATC is characterized by the accumulation of several oncogenic alterations, and studies have shown that an increased number of oncogenic alterations equates to an increased level of dedifferentiation and aggressiveness. The clinical management of ATC requires a multidisciplinary approach; according to recent American Thyroid Association guidelines, surgery, radiotherapy and/or chemotherapy should be considered. In addition to conventional therapies, novel molecular targeted therapies are the most promising emerging treatment modalities. These drugs are often multiple receptor tyrosine kinase inhibitors, several of which have been tested in clinical trials with encouraging results so far. Accordingly, clinical trials are ongoing to evaluate the safety, efficacy and effectiveness of these new agents. This Review describes the updated clinical and pathological features of ATC and provides insight into the molecular biology of this disease. The most recent literature regarding conventional, newly available and future therapies for ATC is also discussed.
Collapse
Affiliation(s)
- Eleonora Molinaro
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Cristina Romei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Agnese Biagini
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Elena Sabini
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Laura Agate
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Salvatore Mazzeo
- Diagnostic and Interventional Radiology Department of Translational Research and New Technologies in Medicine and Surgery, University Hospital of Pisa
| | - Gabriele Materazzi
- Division of Endocrine Surgery, Department of Surgical Pathology, University Hospital of Pisa
| | | | | | - Liborio Torregrossa
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Fulvio Basolo
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Paolo Vitti
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| |
Collapse
|
109
|
Di Desidero T, Antonelli A, Orlandi P, Ferrari SM, Fioravanti A, Alì G, Fontanini G, Basolo F, Francia G, Bocci G. Synergistic efficacy of irinotecan and sunitinib combination in preclinical models of anaplastic thyroid cancer. Cancer Lett 2017; 411:35-43. [PMID: 28964784 DOI: 10.1016/j.canlet.2017.09.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/18/2017] [Accepted: 09/21/2017] [Indexed: 01/19/2023]
Abstract
The identification of new therapeutic strategies is urgently needed for the management of patients affected by anaplastic thyroid cancer (ATC) due to their short survival and poor prognosis. Aim of the study was to determine the activity of the combination irinotecan/sunitinib on ATC cell growth in vitro and the antitumor effects in vivo. Proliferation assays were performed for 72 h on ATC cell lines exposed to the combination of SN-38, the active metabolite of irinotecan, and sunitinib. The simultaneous combination of sunitinib and SN-38, quantified by the combination index, determined a high synergism on ATC cells, increasing the intracellular concentrations of SN-38. Moreover, the synergistic combination greatly decreases the gene expression and the protein levels of vascular endothelial growth factor, colony stimulating factor 1 and ATP-binding cassette transporter G2 in ATC cells. A significant in vivo antitumor effect was observed in ATC xenografts with the simultaneous combination of irinotecan and sunitinib if compared to monotherapy. The simultaneous combination of irinotecan and sunitinib, in vitro and in vivo demonstrated a significant, synergistic ATC antitumor activity, suggesting a possible and rapid translation of this schedule into the clinics.
Collapse
Affiliation(s)
- Teresa Di Desidero
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Alessandro Antonelli
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Paola Orlandi
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Silvia Martina Ferrari
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Anna Fioravanti
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Fulvio Basolo
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, University of Pisa, Pisa, I-56126 Pisa, Italy
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso, Texas, USA
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, I-56126 Pisa, Italy.
| |
Collapse
|
110
|
Anania MC, Cetti E, Lecis D, Todoerti K, Gulino A, Mauro G, Di Marco T, Cleris L, Pagliardini S, Manenti G, Belmonte B, Tripodo C, Neri A, Greco A. Targeting COPZ1 non-oncogene addiction counteracts the viability of thyroid tumor cells. Cancer Lett 2017; 410:201-211. [PMID: 28951131 DOI: 10.1016/j.canlet.2017.09.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 01/19/2023]
Abstract
Thyroid carcinoma is generally associated with good prognosis, but no effective treatments are currently available for aggressive forms not cured by standard therapy. To find novel therapeutic targets for this tumor type, we had previously performed a siRNA-based functional screening to identify genes essential for sustaining the oncogenic phenotype of thyroid tumor cells, but not required to the same extent for the viability of normal cells (non-oncogene addiction paradigm). Among those, we found the coatomer protein complex ζ1 (COPZ1) gene, which is involved in intracellular traffic, autophagy and lipid homeostasis. In this paper, we investigated the mechanisms through which COPZ1 depletion leads to thyroid tumor cell death. We showed that siRNA-mediated COPZ1 depletion causes abortive autophagy, endoplasmic reticulum stress, unfolded protein response and apoptosis. Interestingly, we observed that mouse tumor xenografts, locally treated with siRNA targeting COPZ1, showed a significant reduction of tumor growth. On the whole, we demonstrated for the first time the crucial role of COPZ1 in the viability of thyroid tumor cells, suggesting that it may be considered an attractive target for novel therapeutic approaches for thyroid cancer.
Collapse
Affiliation(s)
- Maria Chiara Anania
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Elena Cetti
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Daniele Lecis
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Katia Todoerti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Alessandro Gulino
- Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - Giuseppe Mauro
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Tiziana Di Marco
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Loredana Cleris
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Sonia Pagliardini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Giacomo Manenti
- Department of Predictive and Preventive Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Beatrice Belmonte
- Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - Claudio Tripodo
- Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy; Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Angela Greco
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.
| |
Collapse
|
111
|
Fallahi P, Ruffilli I, Elia G, Ragusa F, Ulisse S, Baldini E, Miccoli M, Materazzi G, Antonelli A, Ferrari SM. Novel treatment options for anaplastic thyroid cancer. Expert Rev Endocrinol Metab 2017; 12:279-288. [PMID: 30058884 DOI: 10.1080/17446651.2017.1340155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Several genetic alterations have been identified in different molecular pathways ofanaplastic thyroid cancer (ATC) and associated with tumor aggressiveness and progression (BRAF, p53,RAS, EGFR, VEGFR-1, VEGFR-2, etc). New drugs targeting these molecular pathways have beenrecently evaluated in ATC. Areas covered: We review the new targeted therapies of ATC. Interesting results have been reported with molecules targeting different pathways, as: a-BRAF (dabrafenib/trametinib, vemurafenib); b-angiogenesis (sorafenib, combretastatin, vandetanib, sunitinib, lenvatinib, CLM3, etc); c-EGFR (gefitinib); d- PPARγ agonists (rosiglitazone, pioglitazone, efatutazone). In patients with ATC treated with lenvatinib, a median overall survival of 10.6 (3.8-19.8) months was reported. In order to bypass the resistance to the single drug, the capability of targeted drugs to synergize with radiation, or chemotherapy, or other targeted drugs is explored. Expert commentary: New, affordable and individual genomic analysis combined with the opportunity to test these new treatments in primary cell cultures from every ATC patient in vitro, may permit the personalization of therapy. Increasing the therapeutic effectiveness and avoiding the use of ineffective drugs. The identification of new treatments is necessary, to extend life duration guaranteing a good quality of life. To bypass the resistance to asingle drug, the capability of targeted drugs to synergize with radiation, or chemotherapy, or othertargeted drugs is explored. Moreover, new affordable individual genomic analysis and the opportunity totest these novel treatments in primary cell cultures from every ATC patient in vitro, might permit topersonalize the therapy, increasing the therapeutic effectiveness and avoiding the use of ineffectivedrugs.
Collapse
Affiliation(s)
- Poupak Fallahi
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Ilaria Ruffilli
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Giusy Elia
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Francesca Ragusa
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Salvatore Ulisse
- b Department of Experimental Medicine , Sapienza University of Rome , Rome , Italy
| | - Enke Baldini
- b Department of Experimental Medicine , Sapienza University of Rome , Rome , Italy
| | - Mario Miccoli
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Gabriele Materazzi
- c Department of Surgical, Medical, Molecular Pathology and Critical Area , University of Pisa , Pisa , Italy
| | - Alessandro Antonelli
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | | |
Collapse
|
112
|
Vulpe H, Kwan JYY, McNiven A, Brierley JD, Tsang R, Chan B, Goldstein DP, Le LW, Hope A, Giuliani M. Patterns of failure in anaplastic and differentiated thyroid carcinoma treated with intensity-modulated radiotherapy. ACTA ACUST UNITED AC 2017; 24:e226-e232. [PMID: 28680291 DOI: 10.3747/co.24.3551] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The radiotherapy (rt) volumes in anaplastic (atc) and differentiated thyroid carcinoma (dtc) are controversial. METHODS We retrospectively examined the patterns of failure after postoperative intensity-modulated rt for atc and dtc. Computed tomography images were rigidly registered with the original rt plans. Recurrences were considered in-field if more than 95% of the recurrence volume received 95% of the prescribed dose, out-of-field if less than 20% received 95% of the dose, and marginal otherwise. RESULTS Of 30 dtc patients, 4 developed regional recurrence: 1 being in-field (level iii), and 3 being out-of-field (all level ii). Of 5 atc patients, all 5 recurred at 7 sites: 2 recurrences being local, and 5 being regional [2 marginal (intramuscular to the digastric and sternocleidomastoid), 3 out-of-field (retropharyngeal, soft tissues near the manubrium, and lateral to the sternocleidomastoid)]. CONCLUSIONS In dtc, locoregional recurrence is unusual after rt. Out-of-field dtc recurrences infrequently occurred in level ii. Enlarged treatment volumes to level ii must be balanced against a potentially greater risk of toxicity.
Collapse
Affiliation(s)
- H Vulpe
- Department of Radiation Oncology, Princess Margaret Cancer Centre
| | - J Y Y Kwan
- Department of Radiation Oncology, Princess Margaret Cancer Centre
| | - A McNiven
- Department of Radiation Oncology, Princess Margaret Cancer Centre
| | - J D Brierley
- Department of Radiation Oncology, Princess Margaret Cancer Centre
| | - R Tsang
- Department of Radiation Oncology, Princess Margaret Cancer Centre
| | - B Chan
- Radiation Medicine Program, Princess Margaret Cancer Centre
| | - D P Goldstein
- Department of Otolaryngology-Head and Neck Surgery; and
| | - L W Le
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON
| | - A Hope
- Department of Radiation Oncology, Princess Margaret Cancer Centre
| | - M Giuliani
- Department of Radiation Oncology, Princess Margaret Cancer Centre
| |
Collapse
|
113
|
Maggisano V, Celano M, Lombardo GE, Lepore SM, Sponziello M, Rosignolo F, Verrienti A, Baldan F, Puxeddu E, Durante C, Filetti S, Damante G, Russo D, Bulotta S. Silencing of hTERT blocks growth and migration of anaplastic thyroid cancer cells. Mol Cell Endocrinol 2017; 448:34-40. [PMID: 28288903 DOI: 10.1016/j.mce.2017.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 02/09/2017] [Accepted: 03/07/2017] [Indexed: 12/21/2022]
Abstract
Mutations in the hTERT promoter responsible for constitutive telomerase activity are the most frequent genetic alteration detected in anaplastic thyroid cancer (ATC), and proposed as diagnostic and prognostic biomarker in these tumours. In this study we analyzed hTERT expression in a series of human ATCs and investigated the effects of small-interfering RNA-mediated silencing of hTERT on viability and migration and invasive properties of three human ATC cell lines. Expression of hTERT mRNA resulted increased in 8/10 ATCs compared to normal thyroid tissues. Silencing of hTERT in CAL-62, 8505C and SW1736 cells did not modify telomere length but determined a significant decrease (about 50%) of cell proliferation in all cell lines and a great reduction (about 50%) of migration and invasion capacity. These finding demonstrate that hTERT may be considered as a molecular target for ATC treatment.
Collapse
Affiliation(s)
- Valentina Maggisano
- Department of Health Sciences, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Marilena Celano
- Department of Health Sciences, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | | | - Saverio Massimo Lepore
- Department of Health Sciences, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Marialuisa Sponziello
- Department of Internal Medicine and Medical Specialties, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Francesca Rosignolo
- Department of Internal Medicine and Medical Specialties, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Antonella Verrienti
- Department of Internal Medicine and Medical Specialties, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Federica Baldan
- Department of Medical Area, University of Udine, 33100 Udine, Italy
| | - Efisio Puxeddu
- Department of Medicine, University of Perugia, 06100 Perugia, Italy
| | - Cosimo Durante
- Department of Internal Medicine and Medical Specialties, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Sebastiano Filetti
- Department of Internal Medicine and Medical Specialties, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Giuseppe Damante
- Department of Medical Area, University of Udine, 33100 Udine, Italy
| | - Diego Russo
- Department of Health Sciences, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy.
| | - Stefania Bulotta
- Department of Health Sciences, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
114
|
Unusual presentation of anaplastic thyroid carcinoma with diffuse neck and thoracic nodules and hyperthyroidism. DERMATOL SIN 2017. [DOI: 10.1016/j.dsi.2017.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
115
|
Cabanillas ME, Williams MD, Gunn GB, Weitzman SP, Burke L, Busaidy NL, Ying AK, Yiin YH, William WN, Lu C, Lai SY. Facilitating anaplastic thyroid cancer specialized treatment: A model for improving access to multidisciplinary care for patients with anaplastic thyroid cancer. Head Neck 2017; 39:1291-1295. [PMID: 28452157 DOI: 10.1002/hed.24784] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 12/21/2016] [Accepted: 02/08/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC) is a highly aggressive thyroid cancer. Several treatment trials are available, but the number of eligible patients to participate is very low because of the rarity and aggressiveness of the disease. METHODS Facilitating Anaplastic Thyroid Cancer Specialized Treatment (FAST) is a quality improvement project aimed at decreasing time from referral to disposition (scheduling of first appointment) to our institution. After identifying reasons for delays, we created a new process flow specifically for patients with ATC allowing patients to be scheduled immediately. RESULTS Historical data revealed a mean referral to disposition time for patients with ATC of 8.7 days before our intervention. After the intervention, the mean referral to disposition time was reduced to 0.5 days. Participation in treatment trials for all patients with ATC was 34%. CONCLUSION Since the implementation of FAST, the access time has decreased and the number of successful referrals for ATC has increased significantly.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michelle D Williams
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - G Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven P Weitzman
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura Burke
- Department of Quality Measurement and Engineering, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anita K Ying
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yeh Hung Yiin
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William N William
- Department of Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Charles Lu
- Department of Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
116
|
Cutignano A, Seetharamsingh B, D'Angelo D, Nuzzo G, Khairnar PV, Fusco A, Reddy DS, Fontana A. Identification and Synthesis of Mycalol Analogues with Improved Potency against Anaplastic Thyroid Carcinoma Cell Lines. JOURNAL OF NATURAL PRODUCTS 2017; 80:1125-1133. [PMID: 28263603 DOI: 10.1021/acs.jnatprod.7b00043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The marine metabolite mycalol (1) has a specific inhibitory activity on cells of anaplastic thyroid carcinoma (ATC), a very aggressive and rare cancer that does not have effective conventional therapy. In this study, we describe six new related analogues (2-7) that differ in the length of the terminal alkyl residue and the presence of acetate or 3S-hydroxybutyrate (3S)-3HB as a substituent at C-19. Despite the structural analogies, some of the new members were significantly more cytotoxic than 1 on cell lines derived from human ATC. Structures inclusive of the 2'R,3R,4S,7R,8S,19R absolute configuration were assigned to 2-7 on the basis of detailed spectroscopic analysis, synthesis of different isomers, and application of ECD and Mosher's methods. This work led to the identification of mycalol-578 (3) as the most potent analogue, with an IC50 of 2.3 μM on FRO cells.
Collapse
Affiliation(s)
- Adele Cutignano
- Istituto di Chimica Biomolecolare, Bio-Organic Chemistry Unit, Consiglio Nazionale delle Ricerche , Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - B Seetharamsingh
- Division of Organic Chemistry, CSIR-National Chemical Laboratory , Dr. Homi Bhabha Road, Pune, 411008, India
| | - Daniela D'Angelo
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche , Via Pansini 5, 80131, Napoli, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita' degli Studi di Napoli "Federico II" , Via Pansini 5, 80131, Napoli, Italy
| | - Genoveffa Nuzzo
- Istituto di Chimica Biomolecolare, Bio-Organic Chemistry Unit, Consiglio Nazionale delle Ricerche , Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Pankaj V Khairnar
- Division of Organic Chemistry, CSIR-National Chemical Laboratory , Dr. Homi Bhabha Road, Pune, 411008, India
| | - Alfredo Fusco
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche , Via Pansini 5, 80131, Napoli, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita' degli Studi di Napoli "Federico II" , Via Pansini 5, 80131, Napoli, Italy
| | - D Srinivasa Reddy
- Division of Organic Chemistry, CSIR-National Chemical Laboratory , Dr. Homi Bhabha Road, Pune, 411008, India
| | - Angelo Fontana
- Istituto di Chimica Biomolecolare, Bio-Organic Chemistry Unit, Consiglio Nazionale delle Ricerche , Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| |
Collapse
|
117
|
Preparation of human single-chain variable fragment antibodies against anaplastic thyroid carcinoma, and single photon emission‑computed tomography/computed tomography imaging in tumor-bearing nude mice. Oncol Rep 2017; 37:2980-2986. [PMID: 28393196 DOI: 10.3892/or.2017.5550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/07/2016] [Indexed: 11/05/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is the most aggressive malignant thyroid tumor with the worst prognosis, and the response to treatment is poor. We investigated soluble human single-chain variable fragment (scFv) which provides unique information for diagnostics, and for monitoring and optimizing responses to therapy. Enzyme-linked immunosorbent assay (ELISA) was used to detect the expression of scFv. The expression and relative molecular mass of soluble scFv were assessed by sodium dodecyl sulfate‑polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting, respectively. The chloramine T method was used to label scFv with 131I. The labeling rate and the radiochemical purity were determined. To analyze the distribution of 131I‑scFv in body tissues and organs of nude mice, static single photon emission-computed tomography (SPECT) imaging and single photon emission‑computed tomography/computed tomography (SPECT/CT) image fusion were performed. The relative molecular mass of soluble scFv was ~29 kDa. The labeling rate was 91.64% and the radiochemical purity was 93.3±0.32%. SPECT imaging revealed that 131I-scFv selectively accumulated in tumor tissue. In addition, SPECT/CT image fusion results were in agreement with the biodistribution results. In conclusion, the human scFv antibodies against ATC were successfully generated, and clear imaging of 131I‑scFv in a nude mouse model at 48 h was obtained.
Collapse
|
118
|
Expression profile of biomarkers altered in papillary and anaplastic thyroid carcinoma: Contribution of Tunisian patients. Bull Cancer 2017; 104:433-441. [PMID: 28185633 DOI: 10.1016/j.bulcan.2016.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 11/09/2016] [Accepted: 12/07/2016] [Indexed: 01/04/2023]
Abstract
AIMS The objective of this study was to compare the protein expression profile between well-differentiated (papillary) and undifferentiated (anaplastic) thyroid carcinoma in Tunisian patients. METHODS This first Tunisian retrospective study concerned data of 38 thyroid cancer cases (19 papillary carcinoma PTC and 19 anaplastic carcinoma ATC) collected at Salah Azaiez Institute of Tunisia. Immunohistochemistry was used to evaluate tumor expression of different molecular markers (p53, Ki67, E-cadherin, cyclin D1, bcl2, S100 and Her-2). The molecular expression was correlated with the clinicopathological characteristics of the tumors. RESULTS There were 6 differentially expressed markers when comparing anaplastic thyroid carcinoma ATC with papillary thyroid carcinoma PTC. Expression of p53 and Ki67 were significantly increased in 16 and 18 ATC cases respectively, the Ki67 expression was lost in PTC. Cyclin D1, E-cadherin, bcl2 and S100 were overexpressed in PTC tumors; however, they were significantly decreased in ATC. The last marker, Her-2 was expressed in one case of PTC only. CONCLUSION Our results, similar with findings of other ethnic groups, showed alteration in expression of molecular markers associated with tumor dedifferentiation, indicating loss of cell cycle control with increased proliferative activity in ATC carcinoma. These data support the hypothesis that ATC may derive from dedifferentiation of preexisting PTC tumor.
Collapse
|
119
|
Weinberger P, Ponny SR, Xu H, Bai S, Smallridge R, Copland J, Sharma A. Cell Cycle M-Phase Genes Are Highly Upregulated in Anaplastic Thyroid Carcinoma. Thyroid 2017; 27:236-252. [PMID: 27796151 PMCID: PMC5314727 DOI: 10.1089/thy.2016.0285] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) accounts for only 3% of thyroid cancers, yet strikingly, it accounts for almost 40% of thyroid cancer deaths. Currently, no effective therapies exist. In an effort to identify ATC-specific therapeutic targets, we analyzed global gene expression data from multiple studies to identify ATC-specific dysregulated genes. METHODS The National Center for Biotechnology Information Gene Expression Omnibus database was searched for high-throughput gene expression microarray studies from human ATC tissue along with normal thyroid and/or papillary thyroid cancer (PTC) tissue. Gene expression levels in ATC were compared with normal thyroid or PTC using seven separate comparisons, and an ATC-specific gene set common in all seven comparisons was identified. We investigated these genes for their biological functions and pathways. RESULTS There were three studies meeting inclusion criteria, (including 32 ATC patients, 69 PTC, and 75 normal). There were 259 upregulated genes and 286 downregulated genes in ATC with at least two-fold change in all seven comparisons. Using a five-fold filter, 36 genes were upregulated in ATC, while 40 genes were downregulated. Of the 10 top globally upregulated genes in ATC, 4/10 (MMP1, ANLN, CEP55, and TFPI2) are known to play a role in ATC progression; however, 6/10 genes (TMEM158, CXCL5, E2F7, DLGAP5, MME, and ASPM) had not been specifically implicated in ATC. Similarly, 3/10 (SFTA3, LMO3, and C2orf40) of the most globally downregulated genes were novel in this context, while 7/10 genes (SLC26A7, TG, TSHR, DUOX2, CDH1, PDE8B, and FOXE1) have been previously identified in ATC. We experimentally validated a significant correlation for seven transcription factors (KLF16, SP3, ETV6, FOXC1, SP1, EGFR1, and MAFK) with the ATC-specific genes using microarray analysis of ATC cell lines. Ontology clustering of globally altered genes revealed that "mitotic cell cycle" is highly enriched in the globally upregulated gene set (44% of top upregulated genes, p-value <10-30). CONCLUSIONS By focusing on globally altered genes, we have identified a set of consistently altered biological processes and pathways in ATC. Our data are consistent with an important role for M-phase cell cycle genes in ATC, and may provide direction for future studies to identify novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Paul Weinberger
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Otolaryngology, Medical College of Georgia at Augusta University, Augusta, Georgia
- Augusta University Cancer Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Sithara Raju Ponny
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Hongyan Xu
- Department of Biostatistics and Epidemiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | | | - John Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Biostatistics and Epidemiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
120
|
Morandi L, Righi A, Maletta F, Rucci P, Pagni F, Gallo M, Rossi S, Caporali L, Sapino A, Lloyd RV, Asioli S. Somatic mutation profiling of hobnail variant of papillary thyroid carcinoma. Endocr Relat Cancer 2017; 24:107-117. [PMID: 28062544 DOI: 10.1530/erc-16-0546] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 01/06/2017] [Indexed: 02/06/2023]
Abstract
Hobnail variant of papillary thyroid carcinoma (HPTC) represents a recently described, aggressive and rare group of thyroid tumors with poorly understood pathogenesis. Molecular data about this group of cancers are few, and a more detailed molecular characterization of these tumors is needed. The main objective of the study is to define a comprehensive molecular typing of HPTC. Eighteen patients affected by HPTC, including eighteen primary tumors and four lymph node metastases, were screened for NRAS, KRAS, HRAS, BRAF, TP53, PIK3CA, hTERT, PTEN, CDKN2A, EGFR, AKT1, CTNNB1 and NOTCH1 gene mutations. Sequencing is conducted on the MiSEQ system, and molecular data are compared with clinical-pathologic data and follow-up. The patients include 14 women and 4 men. Ages range from 23 to 87 years. All 18 primary tumors of HPTC showed ≥30% hobnail features. BRAF and TP53 mutations are by far the most common genetic alterations in primary HPTC (72.2% and 55.6%, respectively), followed by hTERT (44.4%), PIK3CA (27.8%), CTNNB1 (16.7%), EGFR (11.1%), AKT1 (5.5%) and NOTCH1 (5.5%). The mutational pattern in primary tumors and metastasis was usually maintained. Univariate Cox regression analyses with bootstrap procedure indicated a significantly increased mortality risk in patients harboring BRAF mutation and BRAF mutation associated with TP53 and/or PIK3CA mutations. The detection of these multiple mutations appears to allow the identification of a subset of more aggressive tumors within the group and to bear information that should be useful for prognostic stratification of these patients including the planning of adjuvant therapy.
Collapse
Affiliation(s)
- Luca Morandi
- Department of Biomedical and Neuromotor SciencesSection of Anatomic Pathology 'M. Malpighi' at Bellaria Hospital, University of Bologna, Bologna, Italy
| | - Alberto Righi
- Department of PathologyRizzoli Institute, (IRCCS), Bologna, Italy
| | | | - Paola Rucci
- Section of Hygiene and BiostatisticsUniversity of Bologna, Bologna, Italy
| | - Fabio Pagni
- Department of PathologyUniversity of Milano Bicocca, Monza, Italy
| | - Marco Gallo
- Oncological Endocrinology UnitDepartment of Medical Sciences, AOU Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Sabrina Rossi
- Department of PathologyRegional Hospital, Treviso, Italy
| | - Leonardo Caporali
- Istituto delle Scienze Neurologiche di Bologna (IRCCS)Bellaria Hospital, Bologna, Italy
| | - Anna Sapino
- Institute for Cancer Research and Treatment (IRCCS)Candiolo, Italy
| | - Ricardo V Lloyd
- University of Wisconsin School of Medicine and Public HealthMadison, Wisconsin, USA
| | - Sofia Asioli
- Department of Biomedical and Neuromotor SciencesSection of Anatomic Pathology 'M. Malpighi' at Bellaria Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
121
|
Schmid KW. [Lymph node and distant metastases of thyroid gland cancer. Metastases in the thyroid glands]. DER PATHOLOGE 2016; 36 Suppl 2:171-5. [PMID: 26357953 DOI: 10.1007/s00292-015-0071-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The different biological features of the various major entities of thyroid cancer, e.g. papillary, follicular, poorly differentiated, anaplastic and medullary, depend to a large extent on their different metastatic spread. Papillary thyroid cancer (PTC) has a propensity for cervical lymphatic spread that occurs in 20-50 % of patients whereas distant metastasis occurs in < 5 % of cases. Cervical lymphadenopathy may be the first symptom particularly of (micro) PTC. In contrast follicular thyroid cancer (FTC) has a marked propensity for vascular but not lymphatic invasion and 10-20 % of FTC develop distant metastases. At the time of diagnosis approximately one third of medullary thyroid cancer (MTC) cases show lymph node metastases, in 10-15 % distant metastases and 25 % develop metastases during the course of the disease. Poorly differentiated (PDTC) and anaplastic thyroid cancer (ATC) spread via both lymphatic and vascular invasion. Thus distant metastases are relatively uncommon in DTC and when they occur, long-term stable disease is the typical clinical course. The major sites of distant metastases are the lungs and bone. Metastases to the brain, breasts, liver, kidneys, muscle and skin are relatively rare or even rare. The thyroid gland itself can be a site of metastases from a variety of other tumors. In autopsy series of patients with disseminated cancer disease, metastases to the thyroid gland were found in up to 10 % of cases. Metastases from other primary tumors to the thyroid gland have been reported in 1.4-3 % of patients who have surgery for suspected cancer of the thyroid gland. The most common primary cancers that metastasize to the thyroid gland are renal cell (48.1 %), colorectal (10.4 %), lung (8.3 %) and breast cancer (7.8 %) and surprisingly often sarcomas (4.0 %).
Collapse
Affiliation(s)
- K W Schmid
- Institut für Pathologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Deutschland.
| |
Collapse
|
122
|
Huang Y, Tao Y, Li X, Chang S, Jiang B, Li F, Wang ZM. Bioinformatics analysis of key genes and latent pathway interactions based on the anaplastic thyroid carcinoma gene expression profile. Oncol Lett 2016; 13:167-176. [PMID: 28428828 PMCID: PMC5396846 DOI: 10.3892/ol.2016.5447] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/10/2016] [Indexed: 01/03/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is an aggressive malignant disease in older adults
with a high mortality rate. The present study aimed to examine several key genes and
pathways, which are associated with ATC. The GSE33630 gene expression profile was
downloaded from the Gene Expression Omnibus database, which included 11 ATC and 45
normal thyroid samples. The differentially expressed genes (DEGs) in ATC were
identified using the Limma package in R. The Gene Ontology functions and Kyoto
Encyclopedia of Genes and Genomes pathways of the selected DEGs were enriched using
the Database for Annotation, Visualization and Integrated Discovery. A
protein-protein interaction (PPI) network of the DEGs was constructed to select
significant modules. Furthermore, a latent pathway interactive network was
constructed to select the significant pathways associated with ATC. A total of 665
DEGs in the ATC samples were screened, and four significant modules were selected
from the PPI network. The DEGs in the four modules were enriched in several functions
and pathways. In addition, 29 significant pathways associated with ATC were selected,
and he Toll-like receptor (TLR) signaling pathway, extracellular matrix
(ECM)-receptor interaction and cytokine-cytokine interaction pathway were identified
as important pathways. FBJ murine osteosarcoma viral oncogene homolog (FOS),
chemokine C-X-C motif ligand 10 (CXCL10), collagen type V α1 (COL5A1) and
chemokine (C-C motif) ligand 28 (CCL28) were the key DEGs involved in these
significant pathways. The data obtained in the present study revealed that the TLR
signaling pathway, ECM-receptor interaction and cytokine-cytokine receptor
interaction pathway, and the FOS, CXCL10, COL5A1, COL11A1 and CCL28 genes have
different roles in the progression of ATC, and these may be used as therapeutic
targets for ATC.
Collapse
Affiliation(s)
- Yun Huang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yiming Tao
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xinying Li
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Shi Chang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Bo Jiang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Feng Li
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zhi-Ming Wang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
123
|
Patel SG, Carty SE, McCoy KL, Ohori NP, LeBeau SO, Seethala RR, Nikiforova MN, Nikiforov YE, Yip L. Preoperative detection of RAS mutation may guide extent of thyroidectomy. Surgery 2016; 161:168-175. [PMID: 27863786 DOI: 10.1016/j.surg.2016.04.054] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/25/2016] [Accepted: 04/30/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Preoperative detection of RAS mutations can contribute to cancer risk assessment in indeterminate thyroid nodules, although RAS is not always associated with malignancy. METHODS Fine-needle aspiration samples classified in 1 of 3 indeterminate cytology categories were prospectively tested for N-, H-, and K-RAS mutations using next-generation sequencing assay. RESULTS In the study, 93 patients with 94 nodules had preoperative RAS detected, of whom 86 patients had an operation (69% total thyroidectomy, 29% lobectomy). In total, 76% of RAS-positive nodules were malignant and follicular variant papillary thyroid cancer was the most common cancer type (83%). HRAS mutations had the greatest risk of cancer (92%) followed by NRAS (74%) and KRAS (64%; P = .05). No preoperative variables were associated with malignancy including age (P = .07), sex (P = .49), RAS isoform (P = .05), mutational allelic frequency (P = .49), nodule size (P = .14), cytology category (P = .63), or ultrasound bilaterality (P = .24), multifocality (P = .23), or presence of ≥1 suspicious feature (P = .86). Only 60% of patients with a unifocal nodule on ultrasound had single focus low-risk encapsulated follicular variant papillary thyroid cancer or benign disease. CONCLUSION Preoperative RAS mutation detection in thyroid nodules carries a substantial risk of cancer with a greater risk associated with HRAS and NRAS. Most RAS malignancies are follicular variant papillary thyroid cancer, which may inform the extent of operation.
Collapse
Affiliation(s)
- Snehal G Patel
- Department of Surgery, Division of Endocrine Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Sally E Carty
- Department of Surgery, Division of Endocrine Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Kelly L McCoy
- Department of Surgery, Division of Endocrine Surgery, University of Pittsburgh, Pittsburgh, PA
| | - N Paul Ohori
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Shane O LeBeau
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA
| | - Raja R Seethala
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | | | - Yuri E Nikiforov
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Linwah Yip
- Department of Surgery, Division of Endocrine Surgery, University of Pittsburgh, Pittsburgh, PA.
| |
Collapse
|
124
|
Su X, Jiang X, Wang W, Wang H, Xu X, Lin A, Teng X, Wu H, Teng L. Association of telomerase reverse transcriptase promoter mutations with clinicopathological features and prognosis of thyroid cancer: a meta-analysis. Onco Targets Ther 2016; 9:6965-6976. [PMID: 27956840 PMCID: PMC5113912 DOI: 10.2147/ott.s116594] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The clinicopathological and prognostic significance of telomerase reverse transcriptase (TERT) promoter mutations have been widely investigated in thyroid cancer; however, the results are still discrepant. Systematic searches were performed in PubMed, Web of Science, Scopus, Ovid, and the Cochran Library databases for relevant articles prior to April 2016. Mutation rates were synthesized by R statistical software. The odds ratio or standardized mean difference with 95% confidence interval was pooled by Stata. A total of 22 studies with 4,907 cases were included in this meta-analysis. TERT promoter mutations tended to present in aggressive histological types including poorly differentiated thyroid cancer (33.37%), anaplastic thyroid cancer (38.69%), and tall-cell variant papillary thyroid cancer (30.23%). These promoter mutations were likely to exist in older patients and males and were well associated with larger tumor size, extrathyroidal extension, vascular invasion, lymph node metastasis, distant metastasis, advanced tumor stage, disease recurrence/persistence, and mortality. In addition, TERT promoter mutations (especially C228T) tended to coexist with BRAFV600E mutation, which indicated more aggressive tumor behavior. Therefore, TERT promoter mutations may be promising biomarkers for early diagnosis, risk stratification, prognostic prediction, and management of thyroid cancer.
Collapse
Affiliation(s)
| | | | | | | | - Xin Xu
- Department of Medical Oncology
| | | | | | - Huiling Wu
- Department of Plastic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | | |
Collapse
|
125
|
Han J, Gao W, Su D, Liu Y. Silencing of A-Kinase Anchor Protein 4 (AKAP4) Inhibits Proliferation and Progression of Thyroid Cancer. Oncol Res 2016; 25:873-878. [PMID: 27983916 PMCID: PMC7841067 DOI: 10.3727/096504016x14783701102564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A-kinase anchor protein 4 (AKAP4), a member of the A-kinase anchor family of proteins, plays a role in tumor development and progression. However, its expression pattern and function in human thyroid cancer remain obscure. Here we examined AKAP4 expression in thyroid cancer cell lines as well as the effects of AKAP4 on the proliferation and metastasis of thyroid cancer cells. We also explored the molecular mechanism by which AKAP4 mediates the metastatic potential of thyroid cancer cells. Our results revealed that the transcript and protein levels of AKAP4 were significantly upregulated in thyroid cancer cell lines. In vitro experiments showed that knockdown of AKAP4 significantly inhibited the proliferation, migration, invasion, and epithelial–mesenchymal transition (EMT) process in thyroid cancer cells. Additionally, knockdown of AKAP4 greatly decreased the protein expression of Shh as well as Smo, Ptc, and Gli-1 in ACT-1 cells. Finally, the in vivo nude mice model confirmed that knockdown of AKAP4 attenuated tumor growth. In conclusion, our findings demonstrated that knockdown of AKAP4 inhibited proliferation and metastasis, likely through suppressing the Shh signaling pathway, in thyroid cancer cells. Thus, AKAP4 may act as a potential therapeutic target for human thyroid cancer.
Collapse
|
126
|
Affiliation(s)
- James A Fagin
- From the Department of Medicine and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York (J.A.F.); and the Genetics Branch, National Cancer Institute, Bethesda, MD (S.A.W.)
| | - Samuel A Wells
- From the Department of Medicine and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York (J.A.F.); and the Genetics Branch, National Cancer Institute, Bethesda, MD (S.A.W.)
| |
Collapse
|
127
|
Liu G, Wu K, Sheng Y. Elucidation of the molecular mechanisms of anaplastic thyroid carcinoma by integrated miRNA and mRNA analysis. Oncol Rep 2016; 36:3005-3013. [PMID: 27599582 DOI: 10.3892/or.2016.5064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/24/2016] [Indexed: 11/06/2022] Open
Abstract
To elucidate the complex molecular mechanisms of anaplastic thyroid carcinoma (ATC), the mRNA and miRNA expression profiles of ATC were systematically explored. A total of 55 common differentially expressed genes (DEGs) were obtained from two mRNA expression datasets including 23 ATC samples and 24 paired normal samples. Gene expression levels of three randomly selected DEGs, VCAN, COL5A1 and KCNJ16, were examined using RT-PCR in 10 ATC samples. Notably, the ATC and normal samples were clearly classified into two groups based on their common DEGs. Moreover 23 common DEGs, such as TG, NKX2-1, KCNJ16 and CTHRC1, were predicted to be the potential targets of 17 identified miRNAs in ATC. Meanwhile, several miRNA target genes were associated with biological processes related to tumor progression such as angiogenesis, cell migration or growth and potassium channel regulation. In summary, the poor prognosis of ATC is possibly caused via complex biological processes. Firstly, angiogenesis was activated by the high expression of CTHRC1, VCAN and POSTN, providing necessary nutrition for tumor cells. Then tumor distant metastasis was induced via stimulation of cell migration and cell growth or regulation of cell-cell interaction. Moreover, intracellular potassium concentration changes promoted ATC progression indirectly. Hence, identification of these critical DEGs was valuable in understanding the molecular mechanisms of ATC.
Collapse
Affiliation(s)
- Guoping Liu
- Department of Breast Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Kainan Wu
- Department of Breast Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Yuan Sheng
- Department of Breast Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
128
|
Anaplastic Transformation of Papillary Thyroid Carcinoma Only Seen in Pleural Metastasis: A Case Report with Review of the Literature. Head Neck Pathol 2016; 11:162-167. [PMID: 27550513 PMCID: PMC5429274 DOI: 10.1007/s12105-016-0751-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/09/2016] [Indexed: 10/21/2022]
Abstract
Anaplastic transformation of papillary thyroid carcinoma (PTC) at distant metastatic sites is extremely rare, and there have been fewer than 20 reported cases in the literature. A 61-year-old woman presented with 1-week history of dyspnea. Her past medical history was remarkable because, 19 years ago, she underwent nearly total thyroidectomy and radical neck dissection due to PTC. Computed tomography of the chest revealed a 1.7 cm nodule in the lung and diffuse pleural thickening. Gun biopsy of the lung nodule revealed metastatic PTC with typical histology. However, the pleural biopsy predominantly showed anaplastic pleomorphic and spindle sarcomatoid carcinoma with microscopic focus of PTC. Immunohistochemical results showed both anaplastic sarcomatoid and PTC components positive for TTF-1, galectin-3 and PAX-8, thus supporting anaplastic transformation of PTC at the metastatic site. Subsequently the patient received 1 cycle of cisplatin-based chemotherapy but died from the disease 4 months after diagnosis. Although it is rare, anaplastic transformation of PTC should be considered during differential diagnosis of patients who present with exclusive sarcomatoid morphology at metastatic sites and have a history of PTC. We report another case of anaplastic transformation of PTC, found at pleural metastasis, together with the immunohistochemical profile and a literature review.
Collapse
|
129
|
Cabanillas ME, Busaidy NL, Khan SA, Gunn GB, Dadu R, Rao SN, Waguespack SG. Molecular diagnostics and anaplastic thyroid carcinoma: the time has come to harvest the high hanging fruit. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016. [DOI: 10.2217/ije-2016-0007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeted therapies have played a major role in cancer therapeutics, starting with the discovery of a drug against BCR–ABL rearrangements in chronic myelogenous leukemia. This led to the first approval of a targeted agent in cancer and since, many others have followed. Anaplastic thyroid cancer (ATC) is an aggressive carcinoma with few curative options. Although previous cytotoxic chemotherapy and kinase inhibitor therapies have not proven efficacious in ATC, some of the newer drugs appear to be promising. A case report and a comprehensive review of the current standard of care, genetics, modern therapeutic drugs and clinical trials are presented, in order to outline where we currently stand and where the future lies in the quest for a cure for ATC.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Saad A Khan
- The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - G Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Sarika N Rao
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Steven G Waguespack
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| |
Collapse
|
130
|
Aherne ST, Smyth P, Freeley M, Smith L, Spillane C, O'Leary J, Sheils O. Altered expression of mir-222 and mir-25 influences diverse gene expression changes in transformed normal and anaplastic thyroid cells, and impacts on MEK and TRAIL protein expression. Int J Mol Med 2016; 38:433-45. [PMID: 27353001 PMCID: PMC4935456 DOI: 10.3892/ijmm.2016.2653] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 03/21/2016] [Indexed: 12/25/2022] Open
Abstract
Thyroid cancer is the most common endocrine malignancy and accounts for the majority of endocrine cancer-related deaths each year. Our group and others have previously demonstrated dysfunctional microRNA (miRNA or miR) expression in the context of thyroid cancer. The objective of the present study was to investigate the impact of synthetic manipulation of expression of miR-25 and miR-222 in benign and malignant thyroid cells. miR-25 and miR-222 expression was upregulated in 8505C (an anaplastic thyroid cell line) and Nthy-ori (a SV40-immortalised thyroid cell line) cells, respectively. A transcriptomics-based approach was utilised to identify targets of the two miRNAs and real-time PCR and western blotting were used to validate a subset of the targets. Almost 100 mRNAs of diverse functions were found to be either directly or indirectly targeted by both miR-222 and miR-25 [fold change ≥2, false discovery rate (FDR) ≤0.05]. Gene ontology analysis showed the miR-25 gene target list to be significantly enriched for genes involved in cell adhesion. Fluidigm real-time PCR technologies were used to validate the downregulation of 23 and 22 genes in response to miR-25 and miR-222 overexpression, respectively. The reduction of the expression of two miR-25 protein targets, TNF-related apoptosis-inducing ligand (TRAIL) and mitogen-activated protein kinase kinase 4 (MEK4), was also validated. Manipulating the expression of both miR-222 and miR-25 influenced diverse gene expression changes in thyroid cells. Increased expression of miR-25 reduced MEK4 and TRAIL protein expression, and cell adhesion and apoptosis are important aspects of miR-25 functioning in thyroid cells.
Collapse
Affiliation(s)
- Sinéad T Aherne
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Republic of Ireland
| | - Paul Smyth
- Department of Histopathology, Trinity College Dublin, Dublin 8, Republic of Ireland
| | - Michael Freeley
- Department of Clinical Medicine and Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Republic of Ireland
| | - Leila Smith
- Fluidigm Corporation, Suite 100, South San Francisco, CA 94080, USA
| | - Cathy Spillane
- Department of Histopathology, Trinity College Dublin, Dublin 8, Republic of Ireland
| | - John O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin 8, Republic of Ireland
| | - Orla Sheils
- Department of Histopathology, Trinity College Dublin, Dublin 8, Republic of Ireland
| |
Collapse
|
131
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. Synergistic cytotoxicity of BIIB021 with triptolide through suppression of PI3K/Akt/mTOR and NF-κB signal pathways in thyroid carcinoma cells. Biomed Pharmacother 2016; 83:22-32. [PMID: 27470546 DOI: 10.1016/j.biopha.2016.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/04/2016] [Accepted: 06/09/2016] [Indexed: 01/24/2023] Open
Abstract
The effec.t of BIIB021, a novel heat shock protein 90 (hsp90) inhibitor, on survival of thyroid carcinoma cells has not been evaluated. In this study, the impact of BIIB021 alone or in combination with the histone acetyltransferase inhibitor triptolide on survival of thyroid carcinoma cells was identified. In 8505C and TPC-1 thyroid carcinoma cells, BIIB021 caused cell death in conjunction with alterations in expression of hsp90 client proteins. Cotreatment of both BIIB021 and triptolide, compared with treatment of BIIB021 alone, decreased cell viability, and increased the percentage of dead cells and cytotoxic activity. All of the combination index values were lower than 1.0, suggesting synergistic activity of BIIB021 with triptolide in induction of cytotoxicity. In treatment of both BIIB021 and triptolide, compared with treatment of BIIB021 alone, the protein levels of total and phospho-p53, and cleaved caspase-3 were elevated, while those of total Akt, phospho-mTOR, phospho-4EBP1, phospho-S6K, phospho-NF-κB, survivin, X-linked inhibitor of apoptosis protein (xIAP), cellular inhibitor of apoptosis protein (cIAP) and acetyl. histone H4 were reduced. These results suggest that BIIB021 has a cytotoxic activity accompanied by regulation of hsp90 client proteins in thyroid carcinoma cells. Moreover, the synergism between BIIB021 and triptolide in induction of cytotoxicity is associated with the inhibition of PI3K/Akt/mTOR and NF-κB signal pathways, the underexpression of survivin and the activation of DNA damage response in thyroid carcinoma cells.
Collapse
Affiliation(s)
- Si Hyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jun Goo Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Chul Sik Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Sung-Hee Ihm
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Moon Gi Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Hyung Joon Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Seong Jin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea.
| |
Collapse
|
132
|
Abdulghani J, Gokare P, Gallant JN, Dicker D, Whitcomb T, Cooper T, Liao J, Derr J, Liu J, Goldenberg D, Finnberg NK, El-Deiry WS. Sorafenib and Quinacrine Target Anti-Apoptotic Protein MCL1: A Poor Prognostic Marker in Anaplastic Thyroid Cancer (ATC). Clin Cancer Res 2016; 22:6192-6203. [PMID: 27307592 DOI: 10.1158/1078-0432.ccr-15-2792] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 04/21/2016] [Accepted: 05/21/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE AND EXPERIMENTAL DESIGN Anaplastic thyroid cancer (ATC) comprises approximately 2% of all thyroid cancers, and its median survival rate remains poor. It is responsible for more than one third of thyroid cancer-related deaths. ATC is frequently resistant to conventional therapy, and NFκB signaling has been proposed to be a feature of the disease. We aimed to assess the activity of the antimalaria drug quinacrine known to target NFκB signaling in combination with the clinically relevant kinase inhibitor sorafenib in ATC cells. The presence of NFκB-p65/RELA and its target MCL1 was demonstrated in ATC by meta-data gene set enrichment analysis and IHC. We assessed the responses of a panel of human ATC cell lines to quinacrine and sorafenib in vitro and in vivo RESULTS: We detected increased expression of NFκB-p65/RELA and MCL1 in the nucleus of a subset of ATC compared with non-neoplastic thyroid. ATC cells were found to respond with additive/synergistic tumor cell killing to the combination of sorafenib plus quinacrine in vitro, and the drug combination improves survival of immunodeficient mice injected orthotopically with ATC cells as compared with mice administered either compound alone or doxorubicin. We also demonstrate that the combination of sorafenib and quinacrine is well tolerated in mice. At the molecular level, quinacrine and sorafenib inhibited expression of prosurvival MCL1, pSTAT3, and dampened NFκB signaling. CONCLUSIONS The combination of quinacrine and sorafenib targets emerging molecular hallmarks of ATC and shows promising results in clinically relevant models for the disease. Further testing of sorafenib plus quinacrine can be conducted in ATC patients. Clin Cancer Res; 22(24); 6192-203. ©2016 AACR.
Collapse
Affiliation(s)
- Junaid Abdulghani
- Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, Pennsylvania.,Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Prashanth Gokare
- Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, Pennsylvania.,Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Jean-Nicolas Gallant
- Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, Pennsylvania
| | - David Dicker
- Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, Pennsylvania.,Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Tiffany Whitcomb
- Department of Comparative Medicine, Penn State Hershey Medical Center, Hershey, Pennsylvania
| | - Timothy Cooper
- Department of Comparative Medicine, Penn State Hershey Medical Center, Hershey, Pennsylvania
| | - Jiangang Liao
- Department of Public Health Sciences, Penn State Hershey Medical Center, Hershey, Pennsylvania
| | - Jonathan Derr
- Department of Surgery; Division of Otolaryngology-Head and Neck Surgery, Penn State Hershey Medical Center, Hershey, Pennsylvania
| | - Jing Liu
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - David Goldenberg
- Department of Surgery; Division of Otolaryngology-Head and Neck Surgery, Penn State Hershey Medical Center, Hershey, Pennsylvania
| | - Niklas K Finnberg
- Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, Pennsylvania. .,Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Wafik S El-Deiry
- Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, Pennsylvania. .,Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
133
|
Giuffrida R, Adamo L, Iannolo G, Vicari L, Giuffrida D, Eramo A, Gulisano M, Memeo L, Conticello C. Resistance of papillary thyroid cancer stem cells to chemotherapy. Oncol Lett 2016; 12:687-691. [PMID: 27347201 DOI: 10.3892/ol.2016.4666] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 03/01/2016] [Indexed: 12/30/2022] Open
Abstract
Thyroid carcinoma is the most common endocrine neoplasm, with the highest mortality rate of all the endocrine cancers. Among the endocrine malignancies, ~80% are papillary thyroid carcinomas (PTCs). In the initiation and progression of this tumor, genetic alterations in the mitogen-associated protein kinase pathway, including RAS point mutations, RET/PTC oncogene rearrangements and BRAF point mutations, play an important role, particularly in deciding targeted therapy. In the present study, a small population of thyroid tumor cells, known as tumor spheres, were isolated and characterized from PTC surgical samples. These spheres can be expanded indefinitely in vitro and give rise to differentiated adherent cells when cultivated in differentiative conditions. The present study showed by reverse transcription-polymerase chain reaction and flow cytometric analysis that the undifferentiated PTC cells exhibited a characteristic antigen expression profile of adult progenitor/stem cells. The cells were more resistant to chemotherapeutics, including bortezomib, taxol, cisplatin, etoposide, doxorubicin and vincristine, than differentiated PTC cells and the majority possessed a quiescent status, as revealed by the various cell cycle characteristics and anti-apoptotic protein expression. Such advances in cancer thyroid stem cell biology may provide relevant information for future targeted therapies.
Collapse
Affiliation(s)
| | - Luana Adamo
- Cell Biology Unit, IOM Ricerca Srl, Viagrande, I-95029 Catania, Italy
| | - Gioacchin Iannolo
- Cell Biology Unit, IOM Ricerca Srl, Viagrande, I-95029 Catania, Italy; Regenerative Medicine and Biomedical Technologies Unit, Department of Laboratory Medicine and Advanced Biotechnologies, Mediterranean Institute for Transplantation and Advanced Specialized Therapies, I-90127 Palermo, Italy
| | - Luisa Vicari
- Cell Biology Unit, IOM Ricerca Srl, Viagrande, I-95029 Catania, Italy
| | - Dario Giuffrida
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, I-95029 Catania, Italy
| | - Adriana Eramo
- Department of Hematology, Oncology and Molecular Medicine, National Institute of Health, I-00161 Rome, Italy
| | - Massimo Gulisano
- Cell Biology Unit, IOM Ricerca Srl, Viagrande, I-95029 Catania, Italy
| | - Lorenzo Memeo
- Cell Biology Unit, IOM Ricerca Srl, Viagrande, I-95029 Catania, Italy; Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, I-95029 Catania, Italy
| | - Concetta Conticello
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, I-95029 Catania, Italy
| |
Collapse
|
134
|
Zhao HD, Tang HL, Liu NN, Zhao YL, Liu QQ, Zhu XS, Jia LT, Gao CF, Yang AG, Li JT. Targeting ubiquitin-specific protease 22 suppresses growth and metastasis of anaplastic thyroid carcinoma. Oncotarget 2016; 7:31191-31203. [PMID: 27145278 PMCID: PMC5058749 DOI: 10.18632/oncotarget.9098] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/11/2016] [Indexed: 11/25/2022] Open
Abstract
Ubiquitin-specific protease 22 (USP22) aberrance has been implicated in several malignancies; however, whether USP22 plays a role in anaplastic thyroid carcinoma (ATC) remains unclear. Here, we report that USP22 expression is highly elevated in ATC tissues, which positively correlated with tumor size, extracapsular invasion, clinical stages, and poor prognosis of ATC patients. In vitro assays showed that USP22 depletion suppressed ATC cell survival and proliferation by decreasing Rb phosphorylation and cyclin D2, inactivating Akt, and simultaneously upregulating Rb; USP22 silencing restrained cell migration and invasion by inhibiting epithelial-mesenchymal transition; USP22 knockdown promoted mitochondrion- mediated and caspase-dependent apoptosis by upregulating Bax and Bid and promoting caspase-3 activation. Consistent with in vitro findings, downregulation of USP22 in ATC cells impeded tumor growth and lung metastasis in vivo. These results raise the applicability for USP22 as a useful predictor of ATC prognosis and a potential therapeutic target for ATC.
Collapse
Affiliation(s)
- Hua-Dong Zhao
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hai-Li Tang
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ning-Ning Liu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Ya-Li Zhao
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Qin-Qin Liu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Xiao-Shan Zhu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Lin-Tao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Chun-Fang Gao
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jun-Tang Li
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
135
|
Sadowski SM, Boufraqech M, Zhang L, Mehta A, Kapur P, Zhang Y, Li Z, Shen M, Kebebew E. Torin2 targets dysregulated pathways in anaplastic thyroid cancer and inhibits tumor growth and metastasis. Oncotarget 2016; 6:18038-49. [PMID: 25945839 PMCID: PMC4627234 DOI: 10.18632/oncotarget.3833] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/26/2015] [Indexed: 12/18/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is rare but it is one of the most lethal human malignancies with no effective therapy. There is a pressing need to identify new therapeutic agents for ATC. We performed quantitative high-throughput screening (qHTS) in ATC cell lines using a compound library of 3,282 drugs. qHTS identified 100 pan-active agents. Enrichment analysis of qHTS data showed drugs targeting mTOR were one of the most active drug categories, and Torin2 showed the highest efficacy. We found mTOR to be upregulated in ATC. Treatment of multiple ATC cell lines with Torin2 showed significant dose-dependent inhibition of cellular proliferation with caspase-dependent apoptosis and G1/S phase arrest. Torin2 inhibited cellular migration and inhibited the phosphorylation of key effectors of the mTOR-pathway (AKT, 4E-BP1 and 70S6K), as well as claspin and survivin expression, regulators of cell cycle and apoptosis. In our in vivo mouse model of metastatic ATC, Torin2 inhibited tumor growth and metastasis and significantly prolonged overall survival. Our findings suggest that Torin2 is a promising agent for ATC therapy and that it effectively targets upregulated pathways in human ATC.
Collapse
Affiliation(s)
- Samira M Sadowski
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Myriem Boufraqech
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Zhang
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amit Mehta
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yaqin Zhang
- Division of Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Zhuyin Li
- Division of Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Min Shen
- Division of Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Electron Kebebew
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
136
|
Jeon MJ, Chun SM, Kim D, Kwon H, Jang EK, Kim TY, Kim WB, Shong YK, Jang SJ, Song DE, Kim WG. Genomic Alterations of Anaplastic Thyroid Carcinoma Detected by Targeted Massive Parallel Sequencing in a BRAF(V600E) Mutation-Prevalent Area. Thyroid 2016; 26:683-90. [PMID: 26980298 DOI: 10.1089/thy.2015.0506] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC), the most aggressive type of thyroid cancer, has no effective therapy. Due to its dismal prognosis, it is vital to understand the genetic alterations of ATC and identify effective molecular targets. Targeted next-generation sequencing was performed to investigate the mutational profile of ATC using a massive parallel sequencing approach. METHODS DNA from formalin-fixed, paraffin-embedded archival samples of 11 ATCs and normal matched pairs were used. A total of 48 genetic alterations were identified by targeted exome sequencing. These alterations were validated by mass spectrometric genotyping and direct Sanger sequencing. RESULTS The most commonly mutated gene was BRAF, identified in 10 samples (91%), all showing the V600E point mutation. A KRAS point mutation was observed in the one sample (9%) without the BRAF(V600E) mutation. All 11 ATCs harbored BRAF or RAS mutations, reflecting the possibility that differentiated thyroid carcinomas progress to ATCs after the accumulation of mutations. A loss of function mutation of TP53 was observed in eight samples (73%), a PIK3CA mutation was observed in two samples (18%), and a frameshift mutation of PTEN was observed in one sample (9%). Twenty-eight novel mutated genes were found that had not previously been associated with ATC. Of these, loss of function mutations of NF2, KMT2D, and PKHD1 were repeatedly seen in three samples (27%), two samples (18%), and two samples (18%), respectively. Using direct Sanger sequencing, two samples (18%) were also found with a RASAL1 mutation. KMT2D and RASAL1 mutations were significantly associated with shorter ATC patient survival. CONCLUSIONS This comprehensive analysis of ATCs using targeted massive parallel sequencing identified several novel mutations in ATCs, such as loss of function mutations of NF2 or KMT2D. Future studies are needed to confirm the role of these novel mutations as independent drivers of ATC development.
Collapse
Affiliation(s)
- Min Ji Jeon
- 1 Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Sung-Min Chun
- 2 Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Deokhoon Kim
- 3 Center for Cancer Genome Discovery, Asan Institute for Life Science , Seoul, Korea
| | - Hyemi Kwon
- 1 Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Eun Kyung Jang
- 1 Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Tae Yong Kim
- 1 Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Won Bae Kim
- 1 Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Young Kee Shong
- 1 Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Se Jin Jang
- 2 Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Dong Eun Song
- 2 Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Won Gu Kim
- 1 Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| |
Collapse
|
137
|
Broecker-Preuss M, Becher-Boveleth N, Müller S, Mann K. The BH3 mimetic drug ABT-737 induces apoptosis and acts synergistically with chemotherapeutic drugs in thyroid carcinoma cells. Cancer Cell Int 2016; 16:27. [PMID: 27042160 PMCID: PMC4818940 DOI: 10.1186/s12935-016-0303-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/24/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with dedifferentiated and anaplastic thyroid carcinomas that do not take up radioiodine are resistant to chemotherapeutic treatment and external irradiation and thus are difficult to treat. Direct induction of apoptosis is a promising approach in these apoptosis-resistant tumor cells. The BH3 mimetic ABT-737 belongs to a new class of drugs that target anti-apoptotic proteins of the BCL-2 family and facilitate cell death. The purpose of this study was to investigate the effect of ABT-737 alone or in combination with chemotherapeutic drugs on thyroid carcinoma cell lines. METHODS A total of 16 cell lines derived from follicular, papillary, and anaplastic thyroid carcinomas were treated with ABT-737. Cell viability was measured with MTT assay. Cell death was determined by cell cycle phase distribution and subG1 peak analyses, determination of caspase 3/7 activity and caspase cleavage products, lactate dehydrogenase (LDH) liberation assays and LC3 analysis by western blot. RESULTS The number of viable cells was decreased in all cell lines examined after ABT-737 treatment, with IC50 values ranging from 0.73 to 15.6 μM. Biochemical markers of apoptosis like caspase activities, caspase cleavage products and DNA fragmentation determined as SubG1 peak were elevated after ABT-737 treatment, but no LC3 cleavage was induced by ABT-737 indicating no autophagic processes. In combination with doxorubicin and gemcitabine, ABT-737 showed synergistic effects on cell viability. CONCLUSIONS With these experiments we demonstrated the efficacy of the BH3 mimetic drug ABT-737 against dedifferentiated thyroid carcinoma cells of various histological origins and showed synergistic effects with chemotherapeutic drugs. ABT-737-treated cells underwent an apoptotic cell death. ABT-737 and related BH3 mimetic drugs, alone or in combination, may thus be of value as a new therapeutic option for dedifferentiated thyroid carcinomas.
Collapse
Affiliation(s)
- Martina Broecker-Preuss
- Department of Endocrinology and Metabolism, Division of Laboratory Research, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany ; Department of Clinical Chemistry, University Hospital Essen, Hufelandstr. 55, Essen, Germany
| | - Nina Becher-Boveleth
- Department of Endocrinology and Metabolism, Division of Laboratory Research, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany ; Clinic of Nuclear Medicine, University Hospital Essen, Hufelandstr. 55, Essen, Germany
| | - Stefan Müller
- Department of Nuclear Medicine, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Klaus Mann
- Department of Endocrinology and Metabolism, Division of Laboratory Research, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany ; Center of Endocrinology Alter Hof München, Dienerstr. 12, Munich, Germany
| |
Collapse
|
138
|
Viola D, Valerio L, Molinaro E, Agate L, Bottici V, Biagini A, Lorusso L, Cappagli V, Pieruzzi L, Giani C, Sabini E, Passannati P, Puleo L, Matrone A, Pontillo-Contillo B, Battaglia V, Mazzeo S, Vitti P, Elisei R. Treatment of advanced thyroid cancer with targeted therapies: ten years of experience. Endocr Relat Cancer 2016; 23:R185-205. [PMID: 27207700 DOI: 10.1530/erc-15-0555] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/27/2016] [Indexed: 12/13/2022]
Abstract
Thyroid cancer is rare, but it is the most frequent endocrine malignancy. Its prognosis is generally favorable, especially in cases of well-differentiated thyroid cancers (DTCs), such as papillary and follicular cancers, which have survival rates of approximately 95% at 40 years. However, 15-20% of cases became radioiodine refractory (RAI-R), and until now, no other treatments have been effective. The same problems are found in cases of poorly differentiated (PDTC) and anaplastic (ATC) thyroid cancers and in at least 30% of medullary thyroid cancer (MTC) cases, which are very aggressive and not sensitive to radioiodine. Tyrosine kinase inhibitors (TKIs) represent a new approach to the treatment of advanced cases of RAI-R DTC, MTC, PDTC, and, possibly, ATC. In the past 10 years, several TKIs have been tested for the treatment of advanced, progressive, and RAI-R thyroid tumors, and some of them have been recently approved for use in clinical practice: sorafenib and lenvatinib for DTC and PDTC and vandetanib and cabozantinib for MTC. The objective of this review is to present the current status of the treatment of advanced thyroid cancer with the use of innovative targeted therapies by describing both the benefits and the limits of their use based on the experiences reported so far. A comprehensive analysis and description of the molecular basis of these therapies, as well as new therapeutic perspectives, are reported. Some practical suggestions are given for both the choice of patients to be treated and their management, with particular regard to the potential side effects.
Collapse
Affiliation(s)
- David Viola
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Laura Valerio
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Eleonora Molinaro
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Laura Agate
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Valeria Bottici
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Agnese Biagini
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Loredana Lorusso
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Virginia Cappagli
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Letizia Pieruzzi
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Carlotta Giani
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Elena Sabini
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Paolo Passannati
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Luciana Puleo
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Antonio Matrone
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Benedetta Pontillo-Contillo
- Diagnostic and Interventional RadiologyDepartment of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Valentina Battaglia
- Diagnostic and Interventional RadiologyDepartment of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Salvatore Mazzeo
- Diagnostic and Interventional RadiologyDepartment of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Paolo Vitti
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| | - Rossella Elisei
- Department of Clinical and Experimental MedicineSection of Endocrinology, University of Pisa, Pisa, Italy
| |
Collapse
|
139
|
Mio C, Lavarone E, Conzatti K, Baldan F, Toffoletto B, Puppin C, Filetti S, Durante C, Russo D, Orlacchio A, Di Cristofano A, Di Loreto C, Damante G. MCM5 as a target of BET inhibitors in thyroid cancer cells. Endocr Relat Cancer 2016; 23:335-47. [PMID: 26911376 PMCID: PMC4891972 DOI: 10.1530/erc-15-0322] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/24/2016] [Indexed: 12/21/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is an extremely aggressive thyroid cancer subtype, refractory to the current medical treatment. Among various epigenetic anticancer drugs, bromodomain and extra-terminal inhibitors (BETis) are considered to be an appealing novel class of compounds. BETi target the bromodomain and extra-terminal of BET proteins that act as regulators of gene transcription, interacting with histone acetyl groups. The goal of this study is to delineate which pathway underlies the biological effects derived from BET inhibition, in order to find new potential therapeutic targets in ATC. We investigated the effects of BET inhibition on two human anaplastic thyroid cancer-derived cell lines (FRO and SW1736). The treatment with two BETis, JQ1 and I-BET762, decreased cell viability, reduced cell cycle S-phase, and determined cell death. In order to find BETi effectors, FRO and SW1736 were subjected to a global transcriptome analysis after JQ1 treatment. A significant portion of deregulated genes belongs to cell cycle regulators. Among them, MCM5 was decreased at both mRNA and protein levels in both tested cell lines. Chromatin immunoprecipitation (ChIP) experiments indicate that MCM5 is directly bound by the BET protein BRD4. MCM5 silencing reduced cell proliferation, thus underlining its involvement in the block of proliferation induced by BETis. Furthermore, MCM5 immunohistochemical evaluation in human thyroid tumor tissues demonstrated its overexpression in several papillary thyroid carcinomas and in all ATCs. MCM5 was also overexpressed in a murine model of ATC, and JQ1 treatment reduced Mcm5 mRNA expression in two murine ATC cell lines. Thus, MCM5 could represent a new target in the therapeutic approach against ATC.
Collapse
Affiliation(s)
- Catia Mio
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| | - Elisa Lavarone
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| | - Ketty Conzatti
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| | - Federica Baldan
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| | - Barbara Toffoletto
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| | - Cinzia Puppin
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| | - Sebastiano Filetti
- Department of Internal Medicine and Medical SpecialtiesUniversity 'Sapienza', Rome, Italy
| | - Cosimo Durante
- Department of Internal Medicine and Medical SpecialtiesUniversity 'Sapienza', Rome, Italy
| | - Diego Russo
- Department of Health SciencesUniversity of Catanzaro 'Magna Graecia', Catanzaro, Italy
| | - Arturo Orlacchio
- Department of Developmental and Molecular BiologyAlbert Einstein College of Medicine, Bronx, New York, USA
| | - Antonio Di Cristofano
- Department of Developmental and Molecular BiologyAlbert Einstein College of Medicine, Bronx, New York, USA
| | - Carla Di Loreto
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| | - Giuseppe Damante
- Department of Medical and Biological SciencesUniversity of Udine, Udine, Italy
| |
Collapse
|
140
|
Zhang L, Zhang Y, Mehta A, Boufraqech M, Davis S, Wang J, Tian Z, Yu Z, Boxer MB, Kiefer JA, Copland JA, Smallridge RC, Li Z, Shen M, Kebebew E. Dual inhibition of HDAC and EGFR signaling with CUDC-101 induces potent suppression of tumor growth and metastasis in anaplastic thyroid cancer. Oncotarget 2016; 6:9073-85. [PMID: 25940539 PMCID: PMC4496203 DOI: 10.18632/oncotarget.3268] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 01/31/2015] [Indexed: 12/20/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is one of the most lethal human malignancies that currently has no effective therapy. We performed quantitative high-throughput screening (qHTS) in three ATC cell lines using 3,282 clinically approved drugs and drug candidates, and identified 100 active agents. Enrichment analysis of active compounds showed that inhibitors of EGFR and histone deacetylase (HDAC) were most active. Of these, the first-in-class dual inhibitor of EGFR, HER2 and HDACs, CUDC-101, had the highest efficacy and lower IC50 than established drugs. We validated that CUDC-101 inhibited cellular proliferation and resulted in cell death by inducing cell cycle arrest and caspase-dependent apoptosis. CUDC-101 also inhibited cellular migration in vitro. Mechanistically, CUDC-101 inhibited MAPK signaling and histone deacetylation in ATC cell lines with multiple driver mutations present in human ATC. The anticancer effect of CUDC-101 was associated with increased expression of p21 and E-cadherin, and reduced expression of survivin, XIAP, β-catenin, N-cadherin, and Vimentin. In an in vivo mouse model of metastatic ATC, CUDC-101 inhibited tumor growth and metastases, and significantly prolonged survival. Response to CUDC-101 treatment in vivo was associated with increased histone 3 acetylation and reduced survivin expression. Our findings provide a preclinical basis to evaluate CUDC-101 therapy in ATC.
Collapse
Affiliation(s)
- Lisa Zhang
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yaqin Zhang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Amit Mehta
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Myriem Boufraqech
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sean Davis
- Cancer Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jing Wang
- Curis Inc., Translational Science, Lexington, MA, USA
| | - Ze Tian
- Curis Inc., Translational Science, Lexington, MA, USA
| | - Zhiya Yu
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Matthew B Boxer
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey A Kiefer
- Division of Information Sciences, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - John A Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Robert C Smallridge
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Endocrinology Division, Internal Medicine Department, Mayo Clinic, Jacksonville, FL, USA
| | - Zhuyin Li
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Min Shen
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Electron Kebebew
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
141
|
Bible KC, Ryder M. Evolving molecularly targeted therapies for advanced-stage thyroid cancers. Nat Rev Clin Oncol 2016; 13:403-16. [PMID: 26925962 DOI: 10.1038/nrclinonc.2016.19] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Increased understanding of disease-specific molecular targets of therapy has led to the regulatory approval of two drugs (vandetanib and cabozantinib) for the treatment of medullary thyroid cancer (MTC), and two agents (sorafenib and lenvatinib) for the treatment of radioactive- iodine refractory differentiated thyroid cancer (DTC) in both the USA and in the EU. The effects of these and other therapies on overall survival and quality of life among patients with thyroid cancer, however, remain to be more-clearly defined. When applied early in the disease course, intensive multimodality therapy seems to improve the survival outcomes of patients with anaplastic thyroid cancer (ATC), but salvage therapies for ATC are of uncertain benefit. Additional innovative, rationally designed therapeutic strategies are under active development both for patients with DTC and for patients with ATC, with multiple phase II and phase III randomized clinical trials currently ongoing. Continued effort is being made to identify further signalling pathways with potential therapeutic relevance in thyroid cancers, as well as to elaborate on the complex interactions between signalling pathways, with the intention of translating these discoveries into effective and personalized therapies. Herein, we summarize the progress made in molecular medicine for advanced-stage thyroid cancers of different histotypes, analyse how these developments have altered - and might further refine - patient care, and identify open questions for future research.
Collapse
Affiliation(s)
- Keith C Bible
- Division of Medical Oncology, Department of Oncology, and Endocrine Malignancies Disease Oriented Group, Mayo Clinic Cancer Center, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Mabel Ryder
- Division of Medical Oncology, Department of Oncology, and Endocrine Malignancies Disease Oriented Group, Mayo Clinic Cancer Center, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA.,Division of Endocrinology, Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| |
Collapse
|
142
|
Landa I, Ibrahimpasic T, Boucai L, Sinha R, Knauf JA, Shah RH, Dogan S, Ricarte-Filho JC, Krishnamoorthy GP, Xu B, Schultz N, Berger MF, Sander C, Taylor BS, Ghossein R, Ganly I, Fagin JA. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J Clin Invest 2016; 126:1052-66. [PMID: 26878173 DOI: 10.1172/jci85271] [Citation(s) in RCA: 846] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC) are rare and frequently lethal tumors that so far have not been subjected to comprehensive genetic characterization. METHODS We performed next-generation sequencing of 341 cancer genes from 117 patient-derived PDTCs and ATCs and analyzed the transcriptome of a representative subset of 37 tumors. Results were analyzed in the context of The Cancer Genome Atlas study (TCGA study) of papillary thyroid cancers (PTC). RESULTS Compared to PDTCs, ATCs had a greater mutation burden, including a higher frequency of mutations in TP53, TERT promoter, PI3K/AKT/mTOR pathway effectors, SWI/SNF subunits, and histone methyltransferases. BRAF and RAS were the predominant drivers and dictated distinct tropism for nodal versus distant metastases in PDTC. RAS and BRAF sharply distinguished between PDTCs defined by the Turin (PDTC-Turin) versus MSKCC (PDTC-MSK) criteria, respectively. Mutations of EIF1AX, a component of the translational preinitiation complex, were markedly enriched in PDTCs and ATCs and had a striking pattern of co-occurrence with RAS mutations. While TERT promoter mutations were rare and subclonal in PTCs, they were clonal and highly prevalent in advanced cancers. Application of the TCGA-derived BRAF-RAS score (a measure of MAPK transcriptional output) revealed a preserved relationship with BRAF/RAS mutation in PDTCs, whereas ATCs were BRAF-like irrespective of driver mutation. CONCLUSIONS These data support a model of tumorigenesis whereby PDTCs and ATCs arise from well-differentiated tumors through the accumulation of key additional genetic abnormalities, many of which have prognostic and possible therapeutic relevance. The widespread genomic disruptions in ATC compared with PDTC underscore their greater virulence and higher mortality. FUNDING This work was supported in part by NIH grants CA50706, CA72597, P50-CA72012, P30-CA008748, and 5T32-CA160001; the Lefkovsky Family Foundation; the Society of Memorial Sloan Kettering; the Byrne fund; and Cycle for Survival.
Collapse
|
143
|
Kasaian K, Wiseman SM, Walker BA, Schein JE, Zhao Y, Hirst M, Moore RA, Mungall AJ, Marra MA, Jones SJM. The genomic and transcriptomic landscape of anaplastic thyroid cancer: implications for therapy. BMC Cancer 2015; 15:984. [PMID: 26680454 PMCID: PMC4683857 DOI: 10.1186/s12885-015-1955-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/25/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Anaplastic thyroid carcinoma is the most undifferentiated form of thyroid cancer and one of the deadliest of all adult solid malignancies. Here we report the first genomic and transcriptomic profile of anaplastic thyroid cancer including those of several unique cell lines and outline novel potential drivers of malignancy and targets of therapy. METHODS We describe whole genomic and transcriptomic profiles of 1 primary anaplastic thyroid tumor and 3 authenticated cell lines. Those profiles augmented by the transcriptomes of 4 additional and unique cell lines were compared to 58 pairs of papillary thyroid carcinoma and matched normal tissue transcriptomes from The Cancer Genome Atlas study. RESULTS The most prevalent mutations were those of TP53 and BRAF; repeated alterations of the epigenetic machinery such as frame-shift deletions of HDAC10 and EP300, loss of SMARCA2 and fusions of MECP2, BCL11A and SS18 were observed. Sequence data displayed aneuploidy and large regions of copy loss and gain in all genomes. Common regions of gain were however evident encompassing chromosomes 5p and 20q. We found novel anaplastic gene fusions including MKRN1-BRAF, FGFR2-OGDH and SS18-SLC5A11, all expressed in-frame fusions involving a known proto-oncogene. Comparison of the anaplastic thyroid cancer expression datasets with the papillary thyroid cancer and normal thyroid tissue transcriptomes suggested several known drug targets such as FGFRs, VEGFRs, KIT and RET to have lower expression levels in anaplastic specimens compared with both papillary thyroid cancers and normal tissues, confirming the observed lack of response to therapies targeting these pathways. Further integrative data analysis identified the mTOR signaling pathway as a potential therapeutic target in this disease. CONCLUSIONS Anaplastic thyroid carcinoma possessed heterogeneous and unique profiles revealing the significance of detailed molecular profiling of individual tumors and the treatment of each as a unique entity; the cell line sequence data promises to facilitate the more accurate and intentional drug screening studies for anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Katayoon Kasaian
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Sam M Wiseman
- Department of Surgery, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada.
| | - Blair A Walker
- Department of Pathology and Laboratory Medicine, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada.
| | - Jacqueline E Schein
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Martin Hirst
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada. .,, 570 West 7th Ave, Vancouver, British Columbia, V5Z 4S6, Canada.
| |
Collapse
|
144
|
Rahman MA, Salajegheh A, Smith RA, Lam AKY. MicroRNA-126 suppresses proliferation of undifferentiated (BRAF(V600E) and BRAF(WT)) thyroid carcinoma through targeting PIK3R2 gene and repressing PI3K-AKT proliferation-survival signalling pathway. Exp Cell Res 2015; 339:342-350. [PMID: 26384552 DOI: 10.1016/j.yexcr.2015.09.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/04/2015] [Accepted: 09/13/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND The objectives of this study are to investigate the expression of miR-126 and evaluate its effect on proliferation in undifferentiated thyroid carcinoma. METHODS miR-126 expression of undifferentiated thyroid carcinoma cell lines 8505C (BRAF(V600E/V600E)), BHT-101 (BRAF(V600E/WT)) and MB-1 (BRAF(WT/WT)) were quantified with q-PCR. These cell lines were transiently transfected with exogenous miR-126 (mimic). Following transfection, proliferation effects were observed through MTS proliferation assay and colony formation abilities. Immunofluorescence imaging and Western blot assay were also done to check target proteins expression. RESULTS Under-expression (p<0.05) of miR-126 was noted in BRAF(V600E) mutated undifferentiated thyroid carcinoma cells (8505C and BHT-101), but no change in expression was noted in non BRAF(V600E) mutated undifferentiated thyroid carcinoma cells (MB-1). In addition, a 30-50% drop in proliferation ability and a 35-45% reduction in colony formation capability were noticed in miR-126 mimic transfected group when compared to control group. Furthermore, immunofluorescence images showed reduced expression of p85β and p-AKT protein in miR-126 mimic transfected cells when compared to un-transfected cells. Also, Western blot analysis revealed a 34-40% suppression of p85β protein and a 21-53% drop in active AKT kinase (p-AKT) protein in miR-126 mimic transfected group when compared to control group. CONCLUSIONS Expression of miR-126 was down-regulated in BRAF(V600E) mutated undifferentiated thyroid carcinoma. In addition, miR-126 was found to act as proliferation suppressor targeting PIK3R2 gene and reducing p85β (a regulatory subunit of PI3K kinase) protein translation and lower AKT kinase activity. Therefore, miR-126 could be a potential therapeutic tool in the treatment of undifferentiated thyroid carcinoma.
Collapse
Affiliation(s)
- Md Atiqur Rahman
- Cancer Molecular Pathology, School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Ali Salajegheh
- Cancer Molecular Pathology, School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Robert Anthony Smith
- Cancer Molecular Pathology, School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia; Genomics Research Centre, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Alfred King-yin Lam
- Cancer Molecular Pathology, School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia; Pathology Queensland and Gold Coast University Hospital, Gold Coast, Queensland, Australia.
| |
Collapse
|
145
|
Garcia-Rendueles MER, Ricarte-Filho JC, Untch BR, Landa I, Knauf JA, Voza F, Smith VE, Ganly I, Taylor BS, Persaud Y, Oler G, Fang Y, Jhanwar SC, Viale A, Heguy A, Huberman KH, Giancotti F, Ghossein R, Fagin JA. NF2 Loss Promotes Oncogenic RAS-Induced Thyroid Cancers via YAP-Dependent Transactivation of RAS Proteins and Sensitizes Them to MEK Inhibition. Cancer Discov 2015; 5:1178-93. [PMID: 26359368 PMCID: PMC4642441 DOI: 10.1158/2159-8290.cd-15-0330] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/08/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Ch22q LOH is preferentially associated with RAS mutations in papillary and in poorly differentiated thyroid cancer (PDTC). The 22q tumor suppressor NF2, encoding merlin, is implicated in this interaction because of its frequent loss of function in human thyroid cancer cell lines. Nf2 deletion or Hras mutation is insufficient for transformation, whereas their combined disruption leads to murine PDTC with increased MAPK signaling. Merlin loss induces RAS signaling in part through inactivation of Hippo, which activates a YAP-TEAD transcriptional program. We find that the three RAS genes are themselves YAP-TEAD1 transcriptional targets, providing a novel mechanism of promotion of RAS-induced tumorigenesis. Moreover, pharmacologic disruption of YAP-TEAD with verteporfin blocks RAS transcription and signaling and inhibits cell growth. The increased MAPK output generated by NF2 loss in RAS-mutant cancers may inform therapeutic strategies, as it generates greater dependency on the MAPK pathway for viability. SIGNIFICANCE Intensification of mutant RAS signaling through copy-number imbalances is commonly associated with transformation. We show that NF2/merlin inactivation augments mutant RAS signaling by promoting YAP/TEAD-driven transcription of oncogenic and wild-type RAS, resulting in greater MAPK output and increased sensitivity to MEK inhibitors.
Collapse
MESH Headings
- Animals
- Binding Sites
- Cell Cycle Proteins
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Chromosome Deletion
- Chromosomes, Human, Pair 22
- DNA Copy Number Variations
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Gene Deletion
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Order
- Gene Targeting
- Genes, ras
- Humans
- Mice
- Mice, Transgenic
- Mitogen-Activated Protein Kinases/antagonists & inhibitors
- Models, Biological
- Neoplasm Staging
- Neurofibromin 2/genetics
- Nuclear Proteins/metabolism
- Nucleotide Motifs
- Position-Specific Scoring Matrices
- Promoter Regions, Genetic
- Protein Binding
- Protein Kinase Inhibitors/pharmacology
- Signal Transduction/drug effects
- Thyroid Neoplasms/drug therapy
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Transcription Factors/metabolism
- Transcriptional Activation
Collapse
Affiliation(s)
| | - Julio C Ricarte-Filho
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian R Untch
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Iňigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeffrey A Knauf
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Francesca Voza
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vicki E Smith
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yogindra Persaud
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gisele Oler
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuqiang Fang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Suresh C Jhanwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Agnes Viale
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Adriana Heguy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kety H Huberman
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Filippo Giancotti
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
146
|
Marlow LA, Bok I, Smallridge RC, Copland JA. RhoB upregulation leads to either apoptosis or cytostasis through differential target selection. Endocr Relat Cancer 2015. [PMID: 26206775 PMCID: PMC4559850 DOI: 10.1530/erc-14-0302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Anaplastic thyroid carcinoma is a highly aggressive undifferentiated carcinoma with a mortality rate near 100% due to an assortment of genomic abnormalities which impede the success of therapeutic options. Our laboratory has previously identified that RhoB upregulation serves as a novel molecular therapeutic target and agents upregulating RhoB combined with paclitaxel lead to antitumor synergy. Knowing that histone deacetylase 1 (HDAC1) transcriptionally suppresses RhoB, we sought to extend our findings to other HDACs and to identify the HDAC inhibitor (HDACi) that optimally synergize with paclitaxel. Here we identify HDAC6 as a newly discovered RhoB repressor. By using isoform selective HDAC inhibitors (HDACi) and shRNAs, we show that RhoB has divergent downstream signaling partners, which are dependent on the HDAC isoform that is inhibited. When RhoB upregulates only p21 (cyclin kinase inhibitor) using a class I HDACi (romidepsin), cells undergo cytostasis. When RhoB upregulates BIMEL using class II/(I) HDACi (belinostat or vorinostat), apoptosis occurs. Combinatorial synergy with paclitaxel is dependent upon RhoB and BIMEL while upregulation of RhoB and only p21 blocks synergy. This bifurcated regulation of the cell cycle by RhoB is novel and silencing either p21 or BIMEL turns the previously silenced pathway on, leading to phenotypic reversal. This study intimates that the combination of belinostat/vorinostat with paclitaxel may prove to be an effective therapeutic strategy via the novel observation that class II/(I) HDACi antagonize HDAC6-mediated suppression of RhoB and subsequent BIMEL, thereby promoting antitumor synergy. These overall observations may provide a mechanistic understanding of optimal therapeutic response.
Collapse
Affiliation(s)
- Laura A Marlow
- Departments of Cancer BiologyInternal MedicineDivision of EndocrinologyEndocrine Malignancy Working GroupMayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Ilah Bok
- Departments of Cancer BiologyInternal MedicineDivision of EndocrinologyEndocrine Malignancy Working GroupMayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Robert C Smallridge
- Departments of Cancer BiologyInternal MedicineDivision of EndocrinologyEndocrine Malignancy Working GroupMayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA Departments of Cancer BiologyInternal MedicineDivision of EndocrinologyEndocrine Malignancy Working GroupMayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA Departments of Cancer BiologyInternal MedicineDivision of EndocrinologyEndocrine Malignancy Working GroupMayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - John A Copland
- Departments of Cancer BiologyInternal MedicineDivision of EndocrinologyEndocrine Malignancy Working GroupMayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA Departments of Cancer BiologyInternal MedicineDivision of EndocrinologyEndocrine Malignancy Working GroupMayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| |
Collapse
|
147
|
Haddad RI, Lydiatt WM, Ball DW, Busaidy NL, Byrd D, Callender G, Dickson P, Duh QY, Ehya H, Haymart M, Hoh C, Hunt JP, Iagaru A, Kandeel F, Kopp P, Lamonica DM, McCaffrey JC, Moley JF, Parks L, Raeburn CD, Ridge JA, Ringel MD, Scheri RP, Shah JP, Smallridge RC, Sturgeon C, Wang TN, Wirth LJ, Hoffmann KG, Hughes M. Anaplastic Thyroid Carcinoma, Version 2.2015. J Natl Compr Canc Netw 2015; 13:1140-50. [PMID: 26358798 PMCID: PMC4986600 DOI: 10.6004/jnccn.2015.0139] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
This selection from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Thyroid Carcinoma focuses on anaplastic carcinoma because substantial changes were made to the systemic therapy recommendations for the 2015 update. Dosages and frequency of administration are now provided, docetaxel/doxorubicin regimens were added, and single-agent cisplatin was deleted because it is not recommended for patients with advanced or metastatic anaplastic thyroid cancer.
Collapse
|
148
|
Haghpanah V, Fallah P, Tavakoli R, Naderi M, Samimi H, Soleimani M, Larijani B. Antisense-miR-21 enhances differentiation/apoptosis and reduces cancer stemness state on anaplastic thyroid cancer. Tumour Biol 2015; 37:1299-308. [PMID: 26289851 DOI: 10.1007/s13277-015-3923-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is the most aggressive malignancy in thyroid cancers. Resistance to current therapies is still a challenge. MicroRNAs are a class of small non-coding RNAs, regulating gene expression. MiR-21 is an oncomiR that is overexpressed in nearly all cancers including ATC. Accumulating evidence suggested that miR-21 has a role in cancer stemness state, apoptosis, cell cycle progression, and differentiation. Therefore, we evaluated the application of Off-miR-21 to sequester the microRNA for therapeutic purposes on ATC cell lines. In this study, C643 and SW1736 were transducted by hsa-miR-21 antagomir (Off-miR-21). PTEN gene expression was performed as a known target of miR-21. Stemness state in cancer stem cells (CSCs) was evaluated by the changes of CSC biomarkers including Oct-4 and ABCG2. Apoptosis was assessed by PDCD4 and Mcl-1 gene expression and flow cytometry. Sodium/iodide symporter (NIS) and thyroglobulin (TG) were measured as ATC differentiation markers. In addition, cell cycle progression was investigated via the alterations of p21 gene expression and flow cytometry. Specific downregulation of miR-21 induced the differentiation and apoptosis in C643 and SW1736. Inversely, the treatment inhibited stemness state and cell cycle progression. Knockdown of miR-21 significantly increased the expression of PDCD4, p21, NIS, and TG while leading to decreased expression of Oct-4, ABCG2, and Mcl-1.Taken together, the results suggest that miR-21, as an oncomiR, has a role not only in stemness state but also in tumor growth, differentiation, and apoptosis. Hence, suppression of miR-21 could pave the way for ATC therapy.
Collapse
Affiliation(s)
- Vahid Haghpanah
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Dr. Shariati Hospital, North Kargar Ave., Tehran, 14114, Iran
| | - Parviz Fallah
- Department of Laboratory Science, Faculty of Allied Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Rezvan Tavakoli
- Department of Molecular Biology and Genetic Engineering, Stem Cell Technology Research Center, Tehran, Iran
| | - Mahmood Naderi
- Department of Molecular Biology and Genetic Engineering, Stem Cell Technology Research Center, Tehran, Iran
- Liver and Pancreatobiliary Diseases Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hilda Samimi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Dr. Shariati Hospital, North Kargar Ave., Tehran, 14114, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Dr. Shariati Hospital, North Kargar Ave., Tehran, 14114, Iran.
| |
Collapse
|
149
|
Broecker-Preuss M, Viehof J, Jastrow H, Becher-Boveleth N, Fuhrer D, Mann K. Cell death induction by the BH3 mimetic GX15-070 in thyroid carcinoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015. [PMID: 26198850 PMCID: PMC4510903 DOI: 10.1186/s13046-015-0186-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background The evasion of cell death is one of the hallmarks of cancer, contributing to both tumor progression and resistance to therapy. Dedifferentiated and anaplastic thyroid carcinomas that do not take up radioiodine are resistant to conventional anticancer treatments and patients with these tumors are difficult to treat. BH3 mimetics are a new class of drugs that target anti-apoptotic proteins of the BCL-2 family and promote cell death. The purpose of this study was to analyze the molecular effects of the BH3 mimetic GX15-070 on thyroid carcinoma cell lines and to characterize cell death induced by GX15-070. Methods A total of 17 cell lines derived from follicular, papillary, and anaplastic thyroid carcinomas were treated with GX15-070. Cell viability was measured with MTT assay while cell cycle phase distribution and subG1 peaks were determined after propidium iodide staining. We assessed cell death via the caspase 3/7 activity, caspase cleavage products, lactate dehydrogenase (LDH) liberation assays, and a LC3 analysis by western blot. Ultrastructural changes were analysed by electron microscopy of GX15-070-treated cells. Results After GX15-070 treatment, the number of viable cells was decreased in all cell lines examined, with IC50 values ranging from 48nM to 3.25 μM. We observed biochemical markers of autophagic cell death and necrosis like LC3 conversion and LDH release after the GX15-070 treatment. Electron microscopy revealed several common characteristic ultrastructural changes like swelling of mitochondria, dilatation of rough endoplasmic reticulum, membrane blebbing and formation of vacuoles. GX15-070 treatment induced DNA fragmentation detected by subG1-peak induction and an arrest in G1 phase of the cell cycle. Caspase activation after GX15-070 incubation was detected but had no effect on viability of cells. Conclusions With these experiments we demonstrated the efficacy of the BH3 mimetic drug GX15-070 acting against dedifferentiated thyroid carcinoma cells of various histological origins by the induction of cell death. GX15-070-treated cells underwent non-classical cell death with signs of apoptosis, autophagy and necrosis in parallel. GX15-07 and related compounds thus may be a new therapeutic option for dedifferentiated thyroid carcinoma of various histological subtypes.
Collapse
Affiliation(s)
- Martina Broecker-Preuss
- Department of Endocrinology and Metabolism, and Division of Laboratory Research, University Hospital Essen, Hufelandstrasse 55, Essen, Germany. .,Present address: Department of Clinical Chemistry, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| | - Jan Viehof
- Department of Endocrinology and Metabolism, and Division of Laboratory Research, University Hospital Essen, Hufelandstrasse 55, Essen, Germany. .,Present address: Ruhrlandklinik, University Hospital Essen, Tüschener Weg 40, 45239, Essen, Germany.
| | - Holger Jastrow
- Institute of Anatomy, University Hospital Essen, Hufelandstrasse 55, Essen, Germany.
| | - Nina Becher-Boveleth
- Department of Endocrinology and Metabolism, and Division of Laboratory Research, University Hospital Essen, Hufelandstrasse 55, Essen, Germany. .,Present address: Clinic of Nuclear Medicine, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany.
| | - Dagmar Fuhrer
- Department of Endocrinology and Metabolism, and Division of Laboratory Research, University Hospital Essen, Hufelandstrasse 55, Essen, Germany.
| | - Klaus Mann
- Department of Endocrinology and Metabolism, and Division of Laboratory Research, University Hospital Essen, Hufelandstrasse 55, Essen, Germany. .,Present address: Center of Endocrinology Alter Hof München, Dienerstr. 12, 80331, Munich, Germany.
| |
Collapse
|
150
|
Granata R, Locati LD, Licitra L. Fosbretabulin for the treatment of anaplastic thyroid cancer. Future Oncol 2015; 10:2015-21. [PMID: 25396774 DOI: 10.2217/fon.14.154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fosbretabulin tromethamine is a vascular disrupting agent, which is a type of drug that is designed to damage the vasculature (blood vessels) of cancer tumors, causing central necrosis. This drug showed activity against anaplastic thyroid cancer that was demonstrated in orthotopic xenograft models as well as in Phase I/II trials with or without carboplatin and paclitaxel combination therapy. In all of these studies, fosbretabulin was well tolerated.
Collapse
Affiliation(s)
- Roberta Granata
- Head & Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | |
Collapse
|