1
|
Forozidou E, Syrnioti A, Laskou S, Poutoglidis A, Sapalidis K, Koletsa T. The expression of c-MYC, Cyclin D1 and Ki-67/MIB-1 in benign and malignant thyroid tissues: is there any diagnostic value? ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:267-271. [PMID: 39020541 PMCID: PMC11384854 DOI: 10.47162/rjme.65.2.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
AIM To investigate the immunohistochemical (IHC) expression and the diagnostic value of c-MYC, Cyclin D1, and Ki-67∕MIB-1 in follicular adenomas (FAs), follicular carcinomas (FCs), and anaplastic carcinomas (ACs) of the thyroid gland, as well as in their corresponding adjacent, non-neoplastic thyroid tissue (NNTT). MATERIALS AND METHODS We conducted a retrospective study of patients who were pathologically diagnosed with FA, FC, or AC after total thyroidectomy. Tissue microarrays with cores taken from neoplastic and adjacent NNTT were constructed. Immunohistochemistry for anti-c-MYC, anti-Cyclin D1, and anti-Ki-67∕MIB-1 antibodies was performed, and the positivity was evaluated. RESULTS Twenty-eight specimens were included. Nuclear c-MYC positivity was observed in 4∕11 FCs, and 3∕4 ACs, whereas cytoplasmic c-MYC positivity was found in 16∕24 NNTTs. Globally, there were statistically significant differences between neoplasms and NNTTs, with higher nuclear c-MYC and Cyclin D1 expression observed in neoplasms (p=0.017 and p=0.001, respectively). In contrast, cytoplasmic positivity was seen solely in NNTTs (p=0.001). Cyclin D1 positivity was noted in 11∕13 FAs, 7∕11 FCs, 2∕4 ATCs, and only in one NNTT. A statistically significant correlation was found between MIB1 and c-MYC nuclear positivity (p=0.040). CONCLUSIONS Our findings exhibit a clear difference in the IHC expression of c-MYC and Cyclin D1 between different types of thyroid tumors, as well as between the neoplastic and NNTT. Nuclear c-MYC positivity excludes the benign nature of a thyroid lesion, in contrast to cytoplasmic positivity, which demonstrates normal or hyperplastic nature.
Collapse
Affiliation(s)
- Evropi Forozidou
- Department of Anatomy and Surgical Anatomy, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece;
| | | | | | | | | | | |
Collapse
|
2
|
Satapathy S, Walker H, Brown J, Gambin Y, Wilson MR. The N-end rule pathway regulates ER stress-induced clusterin release to the cytosol where it directs misfolded proteins for degradation. Cell Rep 2023; 42:113059. [PMID: 37660295 DOI: 10.1016/j.celrep.2023.113059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/14/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
Previous work suggests that cell stress induces release of the normally secreted chaperone clusterin (CLU) into the cytosol. We analyzed the localization of CLU in healthy and stressed cells, the mechanism of its cytosolic release, and its interactions with cytosolic misfolded proteins. Key results of this study are the following: (1) full-length CLU is released to the cytosol during stress, (2) the CLU N-terminal D1 residue is recognized by the N-end rule pathway and together with the enzyme ATE1 is essential for cytosolic release, (3) CLU can form stable complexes with cytosolic misfolded proteins and direct them to the proteasome and autophagosomes, and (4) cytosolic CLU protects cells from hypoxic stress and the cytosolic overexpression of an aggregation-prone protein. Collectively, the results suggest that enhanced cytosolic release of CLU is a stress response that can inhibit the toxicity of misfolded proteins and facilitate their targeted degradation via both autophagy and the proteasome.
Collapse
Affiliation(s)
- Sandeep Satapathy
- The Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Holly Walker
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - James Brown
- EMBL Australia Node in Single Molecule Science, and School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yann Gambin
- EMBL Australia Node in Single Molecule Science, and School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mark R Wilson
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia.
| |
Collapse
|
3
|
Samimi H, Tavakoli R, Fallah P, Naderi Sohi A, Amini Shirkouhi M, Naderi M, Haghpanah V. BI-847325, a selective dual MEK and Aurora kinases inhibitor, reduces aggressive behavior of anaplastic thyroid carcinoma on an in vitro three-dimensional culture. Cancer Cell Int 2022; 22:388. [PMID: 36482411 PMCID: PMC9730667 DOI: 10.1186/s12935-022-02813-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is the most aggressive subtype of thyroid cancer. In this study, we used a three-dimensional in vitro system to evaluate the effect of a dual MEK/Aurora kinase inhibitor, BI-847325 anticancer drug, on several cellular and molecular processes involved in cancer progression. METHODS Human ATC cell lines, C643 and SW1736, were grown in alginate hydrogel and treated with IC50 values of BI-847325. The effect of BI-847325 on inhibition of kinases function of MEK1/2 and Aurora kinase B (AURKB) was evaluated via Western blot analysis of phospho-ERK1/2 and phospho-Histone H3 levels. Sodium/iodide symporter (NIS) and thyroglobulin (Tg), as two thyroid-specific differentiation markers, were measured by qRT-PCR as well as flow cytometry and immunoradiometric assay. Apoptosis was assessed by Annexin V/PI flow cytometry and BIM, NFκB1, and NFκB2 expressions. Cell cycle distribution and proliferation were determined via P16, AURKA, and AURKB expressions as well as PI and CFSE flow cytometry assays. Multidrug resistance was evaluated by examining the expression of MDR1 and MRP1. Angiogenesis and invasion were investigated by VEGF expression and F-actin labeling with Alexa Fluor 549 Phalloidin. RESULTS Western blot results showed that BI-847325 inhibits MEK1/2 and AURKB functions by decreasing phospho-ERK1/2 and phospho-Histone H3 levels. BI-847325 induced thyroid differentiation markers and apoptosis in ATC cell lines. Inversely, BI-847325 intervention decreased multidrug resistance, cell cycle progression, proliferation, angiogenesis, and invasion at the molecular and/or cellular levels. CONCLUSION The results of the present study suggest that BI-857,325 might be an effective multi-targeted anticancer drug for ATC treatment.
Collapse
Affiliation(s)
- Hilda Samimi
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran ,grid.411463.50000 0001 0706 2472Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Rezvan Tavakoli
- grid.420169.80000 0000 9562 2611Hepatitis and HIV Department, Pasteur Institute of Iran, Tehran, Iran
| | - Parviz Fallah
- grid.411705.60000 0001 0166 0922Department of Laboratory Science, Faculty of Allied Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Alireza Naderi Sohi
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Amini Shirkouhi
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Naderi
- grid.411705.60000 0001 0166 0922Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Haghpanah
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran ,grid.411705.60000 0001 0166 0922Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Rationale Efficacy and Safety Evidence of Lenvatinib and Pembrolizumab Association in Anaplastic Thyroid Carcinoma. Curr Oncol 2022; 29:7718-7731. [PMID: 36290887 PMCID: PMC9601195 DOI: 10.3390/curroncol29100610] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 01/13/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) are highly aggressive malignant tumors with poor overall prognosis despite multimodal therapy. As ATC are extremely rare, no randomized controlled study has been published for metastatic disease. Thyrosine kinase inhibitors, especially lenvatinib and immune checkpoint inhibitors such as pembrolizumab, are emerging drugs for ATC. Few studies have reported the efficacity of pembrolizumab and lenvatinib association, resulting in its frequent off-label use. In this review, we discuss rationale efficacy and safety evidence for the association of lenvatinib and pembrolizumab in ATC. First, we discuss preclinical rationale for pembrolizumab monotherapy, lenvatinib monotherapy and synergistic action of pembrolizumab and lenvatinib in the metastatic setting. We also discuss clinical evidence for immunotherapy and pembrolizumab in ATC through the analysis of studies evaluating immunotherapy, lenvatinib and pembrolizumab lenvatinib association in ATC. In addition, we discuss the safety of this association and potential predictive biomarkers of efficiency.
Collapse
|
5
|
OZİSİK H, OZDİL B, OZDEMİR M, SİPAHİ M, ERDOĞAN M, CETİNKALP S, OZGEN G, SAYGİLİ F, OKTAY G, AKTUG H. Anaplastik tiroid kanseri hücre hattının morfolojik analizi. EGE TIP DERGISI 2022. [DOI: 10.19161/etd.1168177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Aim: Thyroid follicular cell derived cancers are classified into three groups such as papillary thyroid cancer (85%), follicular thyroid cancer (12%) and anaplastic (undifferentiated) thyroid cancer (ATC) (3%). ATCs have very rapid course, poor treatment outcomes and they are very aggressive. The aim of current study was to assess the analysis of the morphological differences of ATC cell line with the normal thyroid cell line (NTC).
Materials and Methods: NTH and ATC cells were examined with haematoxylin and eosin, the nucleus: cytoplasm (N:C) ratios were detected, and cell cycles were investigated. These cell lines were compared according to their N:C ratio and their abundance in cell cycle phases.
Results: The N:C ratio was higher in ATC than NTC. Both cell groups were mostly found in G0/G1 phase (68.4; 82.8) and have statistical difference in both G0/G1 and S phases.
Conclusion: The rapid course and the rarity of ATC are significant barriers for clinical trials. Cultured cell lines are very important to explore the behaviour in the biology of ATC cells (such as the cell cycle), to understand the course of the disease, and to find an effective target for treatment.
Collapse
Affiliation(s)
- Hatice OZİSİK
- Ege University, Faculty of Medicine, Department of Endocrinology and Metabolism
| | - Berrin OZDİL
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, TEMEL TIP BİLİMLERİ BÖLÜMÜ, HİSTOLOJİ VE EMBRİYOLOJİ ANABİLİM DALI
| | - Merve OZDEMİR
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, TEMEL TIP BİLİMLERİ BÖLÜMÜ, HİSTOLOJİ VE EMBRİYOLOJİ ANABİLİM DALI
| | - Murat SİPAHİ
- DOKUZ EYLUL UNIVERSITY, INSTITUTE OF HEALTH SCIENCES, BIOCHEMISTRY (MEDICINE) (DR)
| | - Mehmet ERDOĞAN
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, DAHİLİ TIP BİLİMLERİ BÖLÜMÜ, İÇ HASTALIKLARI ANABİLİM DALI
| | - Sevki CETİNKALP
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, DAHİLİ TIP BİLİMLERİ BÖLÜMÜ, İÇ HASTALIKLARI ANABİLİM DALI
| | - Gokhan OZGEN
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, DAHİLİ TIP BİLİMLERİ BÖLÜMÜ, İÇ HASTALIKLARI ANABİLİM DALI
| | - Fusun SAYGİLİ
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, DAHİLİ TIP BİLİMLERİ BÖLÜMÜ, İÇ HASTALIKLARI ANABİLİM DALI
| | - Gulgun OKTAY
- DOKUZ EYLÜL ÜNİVERSİTESİ, TIP FAKÜLTESİ, TEMEL TIP BİLİMLERİ BÖLÜMÜ, TIBBİ BİYOKİMYA ANABİLİM DALI, KLİNİK BİYOKİMYA BİLİM DALI
| | - Huseyin AKTUG
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, TEMEL TIP BİLİMLERİ BÖLÜMÜ, HİSTOLOJİ VE EMBRİYOLOJİ ANABİLİM DALI
| |
Collapse
|
6
|
Wiggins RH, Hoffman JM, Fine GC, Covington MF, Salem AE, Koppula BR, Morton KA. PET-CT in Clinical Adult Oncology-V. Head and Neck and Neuro Oncology. Cancers (Basel) 2022; 14:cancers14112726. [PMID: 35681709 PMCID: PMC9179458 DOI: 10.3390/cancers14112726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Positron emission tomography (PET), typically combined with computed tomography (CT) has become a critical advanced imaging technique in oncology. With PET-CT, a radioactive molecule (radiotracer) is injected in the bloodstream and localizes to sites of tumor because of specific cellular features of the tumor that accumulate the targeting radiotracer. The CT scan, performed at the same time, provides information to facilitate attenuation correction, so that radioactivity from deep or dense structures can be better visualized, but with head and neck malignancies it is critical to provide correlating detailed anatomic imaging. PET-CT has a variety of applications in oncology, including staging, therapeutic response assessment, restaging, and surveillance. This series of six review articles provides an overview of the value, applications, and imaging and interpretive strategies of PET-CT in the more common adult malignancies. The fifth report in this series provides a review of PET-CT imaging in head and neck and neuro oncology. Abstract PET-CT is an advanced imaging modality with many oncologic applications, including staging, assessment of response to therapy, restaging, and longitudinal surveillance for recurrence. The goal of this series of six review articles is to provide practical information to providers and imaging professionals regarding the best use of PET-CT for specific oncologic indications, and the potential pitfalls and nuances that characterize these applications. In addition, key tumor-specific clinical information and representative PET-CT images are provided to outline the role that PET-CT plays in the management of oncology patients. Hundreds of different types of tumors exist, both pediatric and adult. A discussion of the role of FDG PET for all of these is beyond the scope of this review. Rather, this series of articles focuses on the most common adult malignancies that may be encountered in clinical practice. It also focuses on FDA-approved and clinically available radiopharmaceuticals, rather than research tracers or those requiring a local cyclotron. The fifth review article in this series focuses on PET-CT imaging in head and neck tumors, as well as brain tumors. Common normal variants, key anatomic features, and benign mimics of these tumors are reviewed. The goal of this review article is to provide the imaging professional with guidance in the interpretation of PET-CT for the more common head and neck malignancies and neuro oncology, and to inform the referring providers so that they can have realistic expectations of the value and limitations of PET-CT for the specific type of tumor being addressed.
Collapse
Affiliation(s)
- Richard H. Wiggins
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - John M. Hoffman
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Gabriel C. Fine
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Matthew F. Covington
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Ahmed Ebada Salem
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
- Department of Radiodiagnosis and Intervention, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Bhasker R. Koppula
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Kathryn A. Morton
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
- Intermountain Healthcare Hospitals, Summit Physician Specialists, Murray, UT 84123, USA
- Correspondence: ; Tel.: +1-801-581-7553
| |
Collapse
|
7
|
TROP-2, Nectin-4, GPNMB, and B7-H3 Are Potentially Therapeutic Targets for Anaplastic Thyroid Carcinoma. Cancers (Basel) 2022; 14:cancers14030579. [PMID: 35158847 PMCID: PMC8833363 DOI: 10.3390/cancers14030579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Anaplastic thyroid carcinoma is a highly aggressive thyroid tumor with a poor prognosis. There are limited choices for the effective treatment of this type of carcinoma. Whether the targets of antibody–drug conjugates are expressed in anaplastic thyroid carcinoma remains unclear. Therefore, we examined expression rates of the following antibody–drug conjugate targets using the tissue microarrays of anaplastic thyroid carcinomas: human epidermal growth factor receptor 2, nectin-4, trophoblast cell surface antigen 2, glycoprotein non-metastatic B, and B7-H3. We found that glycoprotein non-metastatic B and B7-H3 were expressed in most anaplastic thyroid carcinoma tissues. Trophoblast cell surface antigen 2 and nectin-4 were expressed in 65% and 59% of anaplastic thyroid carcinoma tissues, respectively. Trophoblast cell surface antigen 2 was high expressed in anaplastic thyroid carcinoma undifferentiated from papillary thyroid carcinoma. In contrast, nectin-4 expression was high in patients with de novo anaplastic thyroid carcinoma. These cell membrane proteins are potential therapeutic targets for anaplastic thyroid carcinoma. Abstract Background: Anaplastic thyroid carcinoma (ATC) is a highly aggressive thyroid tumor with a poor prognosis. However, there are limited choices for ATC treatment. Recently, the effectiveness of antibody–drug conjugates has been demonstrated in various carcinomas. Whether the targets of antibody–drug conjugates are expressed in anaplastic thyroid carcinoma remains unclear. Methods: Fifty-four patients with ATC were enrolled in this study. Tissue microarrays were constructed using the archives of formalin-fixed paraffin-embedded tissue blocks. All sections were stained with the following antibody–drug conjugate targets: human epidermal growth factor receptor 2 (HER2), nectin-4, trophoblast cell surface antigen 2 (TROP-2), glycoprotein non-metastatic B (GPNMB), and B7-H3. Results: HER2 was negative in all tissues, whereas GPNMB and B7-H3 were expressed in most ATC tissues. TROP-2 and nectin-4 were expressed in 65% and 59% of ATC tissues, respectively. TROP-2 was expressed at significantly higher levels in ATC undifferentiated from papillary thyroid carcinoma than in ATC undifferentiated from follicular thyroid carcinoma and de novo ATC. In contrast, nectin-4 expression was markedly higher in patients with de novo ATC than in those with papillary and follicular thyroid carcinoma. Conclusions: TROP-2 and nectin-4 are potential therapeutic targets for ATC undifferentiated from papillary thyroid carcinoma and de novo ATC, respectively. GPNMB and B7-H3 potential for treating all types of ATC.
Collapse
|
8
|
Smith SM. Molecular biology meets the endocrine pathologist: an appraisal of p27 in thyroid malignancy. DIAGNOSTIC HISTOPATHOLOGY 2020; 26:216-223. [DOI: 10.1016/j.mpdhp.2020.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Laetitia G, Sven S, Fabrice J. Combinatorial Therapies in Thyroid Cancer: An Overview of Preclinical and Clinical Progresses. Cells 2020; 9:E830. [PMID: 32235612 PMCID: PMC7226736 DOI: 10.3390/cells9040830] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Accounting for about 2% of cancers diagnosed worldwide, thyroid cancer has caused about 41,000 deaths in 2018. Despite significant progresses made in recent decades in the treatment of thyroid cancer, many resistances to current monotherapies are observed. In our complete review, we report all treatments that were tested in combination against thyroid cancer. Many preclinical studies investigating the effects of inhibitors of the MAPK and PI3K pathways highlighted the importance of mutations in such signaling pathways and their impacts on the subsequent efficacy of targeted therapies, thus reinforcing the need of more personalized therapeutic strategies. Our review also points out the multiple possibilities of combinatory strategies, particularly using therapies targeting proliferation, survival, angiogenesis, and in combination with conventional treatments such as chemotherapies. In any case, resistances to anticancer therapies always develop through the activation of alternative signaling pathways. Combinatory treatments aim to blockade such mechanisms, which are gradually decrypted, thus offering new perspectives for the future. The preclinical and clinical aspects of our review allow us to have a global opinion of the different therapeutic options currently evaluated in combination and to be aware about new perspectives of treatment of thyroid cancer.
Collapse
Affiliation(s)
- Gheysen Laetitia
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine, Mons University, Avenue du Champ de Mars, 8, B7000 Mons, Belgium; (S.S.); (J.F.)
| | | | | |
Collapse
|
10
|
A Comprehensive Bioinformatics Analysis of UBE2C in Cancers. Int J Mol Sci 2019; 20:ijms20092228. [PMID: 31067633 PMCID: PMC6539744 DOI: 10.3390/ijms20092228] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/24/2019] [Accepted: 05/03/2019] [Indexed: 12/22/2022] Open
Abstract
Ubiquitination is one of the main post-translational modification of proteins. It plays key roles in a broad range of cellular functions, including protein degradation, protein interactions, and subcellular location. In the ubiquitination system, different proteins are involved and their dysregulation can lead to various human diseases, including cancers. By using data available from the Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases, we here show that the ubiquitin conjugating enzyme, E2C (UBE2C), is overexpressed in all 27 cancers we investigated. UBE2C expression is significantly higher in late-stage tumors, which might indicate its involvement in tumor progression and invasion. This study also revealed that patients with higher UBE2C levels showed a shorter overall survival (OS) time and worse OS prognosis. Moreover, our data show that UBE2C higher-expression leads to worse disease-free survival prognosis (DFS), indicating that UBE2C overexpression correlates with poor clinical outcomes. We also identified genes with positive correlations with UBE2C in several cancers. We found a number of poorly studied genes (family with sequence similarity 72-member D, FAM72D; meiotic nuclear divisions 1, MND1; mitochondrial fission regulator 2, MTFR2; and POC1 centriolar protein A, POC1A) whose expression correlates with UBE2C. These genes might be considered as new targets for cancers therapies since they showed overexpression in several cancers and correlate with worse OS prognosis.
Collapse
|
11
|
Miyagawa Y, Araki K, Yamashita T, Tanaka S, Tanaka Y, Tomifuji M, Ueda Y, Yonemitsu Y, Shimada H, Shiotani A. Induction of cell fusion/apoptosis in anaplastic thyroid carcinoma in orthotopic mouse model by urokinase‐specific oncolytic Sendai virus. Head Neck 2019; 41:2873-2882. [DOI: 10.1002/hed.25769] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yoshihiro Miyagawa
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Koji Araki
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Taku Yamashita
- Department of Otolaryngology ‐ Head and Neck SurgeryKitasato University School of Medicine Sagamihara Japan
| | - Shingo Tanaka
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Yuya Tanaka
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Masayuki Tomifuji
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Yasuji Ueda
- Section of Gene Medicine, R&D CenterID Pharma Co., Ltd. Tokyo Japan
| | - Yoshikazu Yonemitsu
- R&D Laboratory for Innovative Biotherapeutics Science, Graduate School of Pharmaceutical SciencesKyushu University Fukuoka Japan
| | - Hideaki Shimada
- Department of SurgeryToho University School of Medicine Tokyo Japan
| | - Akihiro Shiotani
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| |
Collapse
|
12
|
Teshima M, Tokita K, Ryo E, Matsumoto F, Kondo M, Ikegami Y, Shinomiya H, Otsuki N, Hiraoka N, Nibu KI, Yoshimoto S, Mori T. Clinical impact of a cytological screening system using cyclin D1 immunostaining and genomic analysis for the diagnosis of thyroid nodules. BMC Cancer 2019; 19:245. [PMID: 30885146 PMCID: PMC6423761 DOI: 10.1186/s12885-019-5452-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 03/12/2019] [Indexed: 11/10/2022] Open
Abstract
Background Fine-needle aspiration (FNA) is the most reliable method for diagnosing thyroid nodules; however, some features such as atypia of undetermined significance or follicular lesion of undetermined significance can confound efforts to identify malignancies. Similar to BRAF, cyclin D1 may be a strong marker of cell proliferation. Methods One hundred two patients with thyroidal nodule were enrolled in this prospective study. Expression of cyclin D1 in thyroid nodules was determined by immunohistochemistry using both surgical specimens and their cytological specimens. The identification of the optimal cut off points for the diagnosis of malignancy were evaluated using the receiver operating characteristic (ROC) curves and the assessment of the area under the ROC curve (AUC). The specificity, sensitivity, positive predictive value (PPV) of markers were evaluated from crosstabs based on cut off points and significance were calculated. We also analyzed genetic variants by target NGS for thyroid nodule samples. Results The positive predictive value (PPV) and median stain ratio (MSR) of cyclin D1 nuclear staining was determined in papillary thyroid carcinoma (PPV = 91.5%, MSR = 48.5%), follicular adenoma (PPV = 66.7%, MSR = 13.1%), and adenomatous goiter and inflammation controls (MSR = 3.4%). In FNA samples, a threshold of 46% of immunolabelled cells allows to discriminate malignant lesions from benign ones (P < 0.0001), with 81% sensitivity and 100% specificity. A 46% cutoff value for positive cyclin D1 immunostaining in thyroid cells demonstrated 81% sensitivity and 100% specificity. In surgical specimens, ROC curve analysis showed a 5.8% cyclin D1 immunostaining score predicted thyroid neoplasms at 94.4% sensitivity and 92.3% specificity (P = 0.003), while a 15.7% score predicted malignancy at 86.4% sensitivity and 80.5% specificity (P < 0.0001). Finally, three tested clinico-pathological variables (extra thyroidal extension, intraglandular metastasis, and lymph node metastasis) were significant predictors of cyclin D1 immunostaining (P < 0.001). Conclusion Our cytological cyclin D1 screening system provides a simple, accurate, and convenient diagnostic method in precision medicine enabling ready determination of personalized treatment strategies for patients by next generation sequencing using cytological sample. Electronic supplementary material The online version of this article (10.1186/s12885-019-5452-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masanori Teshima
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan.,Department of Head and Neck Surgery, National Cancer Center Hospital, Tokyo, Japan.,Department of Otolaryngology - Head and Neck Surgery, Kobe University, School of Medicine, Kobe, Japan
| | - Kazuya Tokita
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Eijitsu Ryo
- Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan
| | - Fumihiko Matsumoto
- Department of Head and Neck Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Madoka Kondo
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Yota Ikegami
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Hirotaka Shinomiya
- Department of Otolaryngology - Head and Neck Surgery, Kobe University, School of Medicine, Kobe, Japan
| | - Naoki Otsuki
- Department of Otolaryngology - Head and Neck Surgery, Kobe University, School of Medicine, Kobe, Japan
| | - Nobuyoshi Hiraoka
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan.,Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan
| | - Ken-Ichi Nibu
- Department of Otolaryngology - Head and Neck Surgery, Kobe University, School of Medicine, Kobe, Japan
| | - Seiichi Yoshimoto
- Department of Head and Neck Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Taisuke Mori
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan. .,Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
13
|
Saini S, Tulla K, Maker AV, Burman KD, Prabhakar BS. Therapeutic advances in anaplastic thyroid cancer: a current perspective. Mol Cancer 2018; 17:154. [PMID: 30352606 PMCID: PMC6198524 DOI: 10.1186/s12943-018-0903-0] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/08/2018] [Indexed: 02/08/2023] Open
Abstract
Thyroid cancer incidence is increasing at an alarming rate, almost tripling every decade. In 2017, it was the fifth most common cancer in women. Although the majority of thyroid tumors are curable, about 2-3% of thyroid cancers are refractory to standard treatments. These undifferentiated, highly aggressive and mostly chemo-resistant tumors are phenotypically-termed anaplastic thyroid cancer (ATC). ATCs are resistant to standard therapies and are extremely difficult to manage. In this review, we provide the information related to current and recently emerged first-line systemic therapy (Dabrafenib and Trametinib) along with promising therapeutics which are in clinical trials and may be incorporated into clinical practice in the future. Different categories of promising therapeutics such as Aurora kinase inhibitors, multi-kinase inhibitors, epigenetic modulators, gene therapy using oncolytic viruses, apoptosis-inducing agents, and immunotherapy are reviewed. Combination treatment options that showed synergistic and antagonistic effects are also discussed. We highlight ongoing clinical trials in ATC and discuss how personalized medicine is crucial to design the second line of treatment. Besides using conventional combination therapy, embracing a personalized approach based on advanced genomics and proteomics assessment will be crucial to developing a tailored treatment plan to improve the chances of clinical success.
Collapse
Affiliation(s)
- Shikha Saini
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL USA
| | - Kiara Tulla
- Department of Surgery, Division of Surgical Oncology, University of Illinois-College of Medicine, Chicago, IL USA
| | - Ajay V. Maker
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL USA
- Department of Surgery, Division of Surgical Oncology, University of Illinois-College of Medicine, Chicago, IL USA
| | | | - Bellur S. Prabhakar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL USA
- Jesse Brown VA Medical Center, Chicago, IL USA
| |
Collapse
|
14
|
Sasanakietkul T, Murtha TD, Javid M, Korah R, Carling T. Epigenetic modifications in poorly differentiated and anaplastic thyroid cancer. Mol Cell Endocrinol 2018; 469:23-37. [PMID: 28552796 DOI: 10.1016/j.mce.2017.05.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/12/2017] [Accepted: 05/21/2017] [Indexed: 12/25/2022]
Abstract
Well-differentiated thyroid cancer accounts for the majority of endocrine malignancies and, in general, has an excellent prognosis. In contrast, the less common poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) are two of the most aggressive human malignancies. Recently, there has been an increased focus on the epigenetic alterations underlying thyroid carcinogenesis, including those that drive PDTC and ATC. Dysregulated epigenetic candidates identified include the Aurora group, KMT2D, PTEN, RASSF1A, multiple non-coding RNAs (ncRNA), and the SWI/SNF chromatin-remodeling complex. A deeper understanding of the signaling pathways affected by epigenetic dysregulation may improve prognostic testing and support the advancement of thyroid-specific epigenetic therapies. This review outlines the current understanding of epigenetic alterations observed in PDTC and ATC and explores the potential for exploiting this understanding in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Thanyawat Sasanakietkul
- Yale Endocrine Neoplasia Laboratory, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Surgery, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Timothy D Murtha
- Yale Endocrine Neoplasia Laboratory, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Surgery, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mahsa Javid
- Yale Endocrine Neoplasia Laboratory, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Surgery, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Reju Korah
- Yale Endocrine Neoplasia Laboratory, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Surgery, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Tobias Carling
- Yale Endocrine Neoplasia Laboratory, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Surgery, Section of Endocrine Surgery, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
15
|
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
|
16
|
Sakr HI, Chute DJ, Nasr C, Sturgis CD. cMYC expression in thyroid follicular cell-derived carcinomas: a role in thyroid tumorigenesis. Diagn Pathol 2017; 12:71. [PMID: 28974238 PMCID: PMC5627435 DOI: 10.1186/s13000-017-0661-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/25/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND cMYC regulates approximately 15% of human genes and is involved in up to 20% of all human cancers. Reports discussing cMYC protein expression in thyroid carcinomas are limited, with controversies pertaining to cMYC expression patterns noted in the literature. The aims of the current study were to clarify patterns and intensities of cMYC expression in follicular cell-derived thyroid carcinomas across a spectrum of cancer morphologies and disease aggressivities, to correlate cMYC with BRAFV600E expression, and to evaluate the potential role of cMYC in progression of well-differentiated thyroid carcinomas into less well-differentiated carcinomas. METHODS Immunohistochemical studies using specific monoclonal antibodies for cMYC and BRAFV600E were performed on tissue microarrays built from follicular cell-derived thyroid carcinomas (25 papillary, 24 follicular, 24 oncocytic variant of follicular, and 21 undifferentiated). In addition, cMYC IHC testing was also performed on whole tissue tumor sections from a subset of patients. Nodular hyperplasia cases were used as non-neoplastic controls. Appropriate positive and negative controls were included. RESULTS cMYC was expressed almost exclusively in a nuclear fashion in both thyroid carcinomas and nodular hyperplasias. cMYC expression was weakly positive in both nodular hyperplasias and well-differentiated carcinomas. The majority of undifferentiated carcinomas (UDCs) showed strong nuclear cMYC positivity. PTC cases that were positive for cMYC (6/25) harbored the BRAF V600E mutation. A correlation was confirmed between cMYC intensity and tumor size in UDCs. UDC cases that developed out of well-differentiated thyroid carcinomas showed frank overexpression of cMYC in the undifferentiated tumor components. CONCLUSIONS Our study suggests that nuclear overexpression of cMYC correlates with tumorigenesis / dedifferentiation in follicular cell derived thyroid carcinomas, a concept that has not been shown before on whole tissue sections.
Collapse
Affiliation(s)
- Hany I Sakr
- Cleveland Clinic, Department of Pathology and Laboratory Medicine, 9500 Euclid Avenue, L25, Cleveland, OH, 44195, USA
| | - Deborah J Chute
- Cleveland Clinic, Department of Pathology and Laboratory Medicine, 9500 Euclid Avenue, L25, Cleveland, OH, 44195, USA
| | - Christian Nasr
- Cleveland Clinic, Department of Endocrinology, Diabetes and Metabolism, Cleveland, USA
| | - Charles D Sturgis
- Cleveland Clinic, Department of Pathology and Laboratory Medicine, 9500 Euclid Avenue, L25, Cleveland, OH, 44195, USA.
| |
Collapse
|
17
|
Kotian S, Zhang L, Boufraqech M, Gaskins K, Gara SK, Quezado M, Nilubol N, Kebebew E. Dual Inhibition of HDAC and Tyrosine Kinase Signaling Pathways with CUDC-907 Inhibits Thyroid Cancer Growth and Metastases. Clin Cancer Res 2017; 23:5044-5054. [PMID: 28600475 DOI: 10.1158/1078-0432.ccr-17-1043] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/08/2017] [Accepted: 06/05/2017] [Indexed: 12/18/2022]
Abstract
Purpose: There is currently no standard therapy for anaplastic thyroid cancer (ATC) and poorly differentiated thyroid cancer (PDTC), which account for two-thirds of thyroid cancer-related deaths. Driver mutations in the PI3K/AKT and RAF/RAS/MEK/ERK pathways are common in ATC and PDTC. Histone deacetylases (HDAC) regulate cancer initiation and progression. Our aim was to determine the therapeutic efficacy of simultaneously targeting these pathways in thyroid cancer with a single agent and to evaluate biomarkers of treatment response.Experimental Design: CUDC-907 is a first-in-class compound, functioning as a dual inhibitor of HDACs and the PI3K/AKT pathway. We investigated its antiproliferative effect in vitro and in vivoResults: CUDC-907 significantly inhibited cellular proliferation in thyroid cancer cell lines, induced G2-M arrest with decreased levels of the checkpoint regulators cyclin B1, AURKA, AURKB, PLK1, and increased p21 and p27. Treatment induced apoptosis with increased caspase-3/7 activity and decreased survivin levels and decreased cellular migration and invasion. CUDC-907 treatment caused H3 hyperacetylation and decreased HDAC2 expression. HDAC2 was upregulated in ATC and other thyroid cancer histologic subtypes. CUDC-907 treatment reduced both p-AKT and p-ERK1/2 levels. Finally, CUDC-907 treatment, in a metastatic mouse model of thyroid cancer, showed significant inhibition of growth and metastases, and tumors from treated mice had decreased HDAC2 expression, suggesting that this may be a useful biomarker of response.Conclusions: Dual inhibition of HDAC and the tyrosine kinase signaling pathways with CUDC-907 is a promising treatment strategy for advanced, metastatic thyroid cancer. Clin Cancer Res; 23(17); 5044-54. ©2017 AACR.
Collapse
Affiliation(s)
- Shweta Kotian
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Lisa Zhang
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Myriem Boufraqech
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Kelli Gaskins
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Sudheer Kumar Gara
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Naris Nilubol
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Electron Kebebew
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland. .,Department of Surgery, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
18
|
Clusterin expression in medullary thyroid carcinoma is inversely correlated with the presence of lymph node metastases. Hum Pathol 2017; 64:37-43. [DOI: 10.1016/j.humpath.2017.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 03/06/2017] [Accepted: 03/19/2017] [Indexed: 12/21/2022]
|
19
|
Wang W, Zhou J, Zhao L, Chen S. Combination of SL327 and Sunitinib Malate leads to an additive anti-cancer effect in doxorubicin resistant thyroid carcinoma cells. Biomed Pharmacother 2017; 88:985-990. [PMID: 28178630 DOI: 10.1016/j.biopha.2017.01.135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 01/22/2017] [Accepted: 01/23/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Receptor tyrosine kinases (RTKs) play crucial roles in numerous cancer cell processes including cell survival, proliferation, and migration. MEK1/2 MAPK kinases are very important for cancer survival and development. Anaplastic thyroid carcinoma (ATC) is a deadly type of thyroid cancer and there are no very effective systemic treatment strategies for ATC so far. Also, ATC can easily become resistant to therapy of traditional therapeutic drugs for ATC, such as doxorubicin. Drug combination treatment could be a promising therapeutic strategy for ATC, especially for drug resistant ATC. METHODS We explored the combination effect between a MEK1/2 inhibitor SL327 and a multi-targeted RTK inhibitor Sunitinib Malate in doxorubicin resistant ATC cells using cell viability assay, cell migration assay, nuclei morphology and caspase-3 activity analysis, as well as in vivo tumor growth assay. RESULTS There is a significant additive effect between SL327 and Sunitinib Malate in reducing viability, increasing apoptosis, and suppressing migration of doxorubicin-resistant ATC cells. Importantly, combination of SL327 and Sunitinib Malate induced significant additive suppression of in vivo doxorubicin-resistant ATC tumor growth. CONCLUSIONS Our results suggest that the combination of MEK1/2 inhibitor and RTK inhibitor is promising for treatment of ATC especially doxorubicin-resistant ATC. The combination might not only enhance the anti-cancer efficacy, but also reduce the side effects and overcome drug resistance developed in ATC treatment. All these might provide useful information for clinical therapeutics of ATC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Endocrine, Affiliated Yantai Yuhuangding Hospital of Qingdao University Medical College, Shandong, 264000, PR China
| | - Jin Zhou
- Department of Endocrine, Affiliated Yantai Yuhuangding Hospital of Qingdao University Medical College, Shandong, 264000, PR China
| | - Lujie Zhao
- Department of Hemodialysis, Affiliated Yantai Yuhuangding Hospital of Qingdao University Medical College, Shandong, 264000, PR China
| | - Shulin Chen
- Department of Endocrine, Affiliated Yantai Yuhuangding Hospital of Qingdao University Medical College, Shandong, 264000, PR China.
| |
Collapse
|
20
|
Mesic A, Rogar M, Hudler P, Juvan R, Komel R. Association of the AURKA and AURKC gene polymorphisms with an increased risk of gastric cancer. IUBMB Life 2016; 68:634-44. [PMID: 27270838 DOI: 10.1002/iub.1521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/18/2016] [Indexed: 12/19/2022]
Abstract
Single nucleotide polymorphisms (SNPs) in mitotic checkpoint genes can contribute to susceptibility of human cancer, including gastric cancer (GC). We aimed to investigate the effects of Aurora kinase A (AURKA), Aurora kinase B (AURKB), and Aurora kinase C (AURKC) gene polymorphisms on GC risk in Slovenian population. We genotyped four SNPs in AURKA (rs2273535 and rs1047972), AURKB (rs2241909), and AURKC (rs758099) in a total of 128 GC patients and 372 healthy controls using TaqMan allelic discrimination assays to evaluate their effects on GC risk. Our results showed that genotype frequencies between cases and controls were significantly different for rs1047972 and rs758099 (P < 0.05). Our study demonstrated that AURKA rs1047972 TT and (CC + CT) genotypes were significantly associated with an increased risk of gastric cancer. Our results additionally revealed that AURKC rs758099 TT and (CC + CT) genotypes were also associated with increased GC risk. In stratified analysis, genotypes TT and (CC + CT) of AURKA rs1047972 SNP were associated with increased risk of both, intestinal and diffuse, types of GC. In addition, AURKC rs758099 TT and (CC + CT) genotypes were positively associated with increased intestinal type GC risk, but not with an increased diffuse type GC risk. Based on these results, we can conclude that AURKA rs1047972 and AURKC rs758099 polymorphisms could affect the risk of GC development. Further larger studies are needed to confirm these findings. © 2016 IUBMB Life, 68(8):634-644, 2016.
Collapse
Affiliation(s)
- Aner Mesic
- Department of Biology, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Marija Rogar
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Hudler
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Juvan
- Clinical Department for Abdominal Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Radovan Komel
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Deregulation of EGFR/PI3K and activation of PTEN by photodynamic therapy combined with carboplatin in human anaplastic thyroid cancer cells and xenograft tumors in nude mice. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 148:118-127. [DOI: 10.1016/j.jphotobiol.2015.03.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/16/2015] [Accepted: 03/16/2015] [Indexed: 12/22/2022]
|
24
|
Onoda N, Nakamura M, Aomatsu N, Noda S, Kashiwagi S, Kurata K, Uchino S, Hirakawa K. Significant cytostatic effect of everolimus on a gefitinib-resistant anaplastic thyroid cancer cell line harboring PI3KCA gene mutation. Mol Clin Oncol 2015; 3:522-526. [PMID: 26137260 DOI: 10.3892/mco.2015.496] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/14/2015] [Indexed: 01/16/2023] Open
Abstract
We previously demonstrated the efficacy of gefitinib, a tyrosine kinase inhibitor of the epidermal growth factor receptor (EGFR), on an anaplastic thyroid cancer (ATC) cell line. We also observed that gefitinib was not effective in regulating cell growth in a different ATC cell line that exhibited an altered EGFR-initiated signal transduction pathway. In the present study, we attempted to regulate the downstream effector of EGFR-Akt-mammalian target of rapamycin (mTOR) pathway by an mTOR inhibitor, everolimus. A total of 8 ATC cell lines were employed, 7 of which were established in our institute. OCUT-2 was known to carry a mutation in the phosphoinositide-3-kinase, catalytic, α polypeptide gene (PI3KCA) and to be gefitinib-resistant, whereas ACT-1 exhibited a remarkable growth arrest by gefitinib. All the cell lines were tested for the cytotoxic effect of everolimus. The mechanisms of cellular toxicity were investigated by EGFR stimulation, cell cycle and concurrent exposure to paclitaxel. In OCUT-2, but not in any of the other cell lines, everolimus achieved a significant growth inhibition (inhibition of 30 and 50% was achieved by concentrations of 0.8 and 5 nM, respectively). The growth in OCUT-2 was inhibited by everolimus, even with concordant EGFR stimulation. This effect was demonstrated by a G2M cell cycle arrest. An additive effect of everolimus onto the cytotoxic effect of paclitaxel was demonstrated at a dose of 1-2 nM. A significant growth inhibitory effect of everolimus on the gefitinib-resistant ATC cell line was demonstrated, suggesting a possible correlation between the efficacy of everolimus and PI3KCA gene mutation and the significance of molecular-targeted therapy in the management of ATC.
Collapse
Affiliation(s)
- Naoyoshi Onoda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Masanori Nakamura
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Naoki Aomatsu
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Satoru Noda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Shinichiro Kashiwagi
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Kento Kurata
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | | | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| |
Collapse
|
25
|
Selemetjev SA, Savin SB, Paunovic IR, Tatic SB, Cvejic D. Changes in the expression pattern of apoptotic molecules (galectin-3, Bcl-2, Bax, survivin) during progression of thyroid malignancy and their clinical significance. Wien Klin Wochenschr 2014; 127:337-44. [DOI: 10.1007/s00508-014-0674-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 11/02/2014] [Indexed: 12/22/2022]
|
26
|
Zhang P, Wu SK, Wang Y, Fan ZX, Li CR, Feng M, Xu P, Wang WD, Lang JY. p53, MDM2, eIF4E and EGFR expression in nasopharyngeal carcinoma and their correlation with clinicopathological characteristics and prognosis: A retrospective study. Oncol Lett 2014; 9:113-118. [PMID: 25435943 PMCID: PMC4246848 DOI: 10.3892/ol.2014.2631] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 08/22/2014] [Indexed: 12/19/2022] Open
Abstract
In the present study, the expression of p53, mouse double minute 2 homolog (MDM2), eukaryotic translation initiation factor 4E (eIF4E), and epidermal growth factor receptor (EGFR) were investigated in nasopharyngeal carcinoma (NPC), and the correlation between their expression and clinicopathological characteristics and prognosis was analyzed. The medical records of 96 NPC patients who had undergone biopsy prior to radical radiotherapy and chemotherapy between 2005 and 2009 were reviewed, retrospectively. All patients received intensity-modulated radiotherapy with concurrent platinum-based chemotherapy. Patients were followed-up for three years. Streptavidin-peroxidase immunohistochemistry was used to evaluate the expression of p53, MDM2, eIF4E and EGFR in NPC biopsy specimens, and the association between their expression and clinical parameters and survival was analyzed. The p53, MDM2, eIF4E and EGFR expression rates were 65.6% (63/96), 79.16% (76/96), 77.08% (74/96) and 89.5% (86/96), respectively. p53 (χ2,20.322; P=0.001) and EGFR (χ2,8.337; P=0.005) expression were found to correlate with T stage, whereas MDM2 (χ2,16.361; P=0.001) expression was found to correlate with lymph node metastasis. p53 expression was found to inversely correlate with MDM2 expression (r, −3.24; P<0.05). Three-year survival rates were lower in p53-positive (76.2%) patients when compared with p53-negative (93.9%) patients. In addition, three-year survival rates were lower in EGFR-positive (75.8%) patients than in EGFR-negative patients (91.2%). The Cox proportional-hazards regression model revealed that p53 (β,−0.455; χ2,5.491; P=0.019) and EGFR (β, 3.93; χ2, 11.95; P=0.001) expression were independent prognostic factors. Thus, it was hypothesized that p53 and EGFR expression present potential unfavorable prognostic markers for patients with NPC.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Radiation Oncology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan, 610041, P.R. China
| | - Song-Ke Wu
- Department of Oncology, Cangxi People's Hospital, Guangyuan, Sichuan, 618400, P.R. China
| | - Ying Wang
- Department of Pathology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - Zi-Xuan Fan
- Department of Radiation Oncology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan, 610041, P.R. China
| | - Chu-Rong Li
- Department of Radiation Oncology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan, 610041, P.R. China
| | - Mei Feng
- Department of Radiation Oncology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan, 610041, P.R. China
| | - Peng Xu
- Department of Radiation Oncology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan, 610041, P.R. China
| | - Wei-Dong Wang
- Department of Radiation Oncology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan, 610041, P.R. China
| | - Jin-Yi Lang
- Department of Radiation Oncology, Sichuan Provincial Cancer Hospital, Chengdu, Sichuan, 610041, P.R. China
| |
Collapse
|
27
|
Baldini E, Tuccilli C, Prinzi N, Sorrenti S, Antonelli A, Gnessi L, Morrone S, Moretti C, Bononi M, Arlot-Bonnemains Y, D'Armiento M, Ulisse S. Effects of selective inhibitors of Aurora kinases on anaplastic thyroid carcinoma cell lines. Endocr Relat Cancer 2014; 21:797-811. [PMID: 25074669 DOI: 10.1530/erc-14-0299] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Aurora kinases are serine/threonine kinases that play an essential role in cell division. Their aberrant expression and/or function induce severe mitotic abnormalities, resulting in either cell death or aneuploidy. Overexpression of Aurora kinases is often found in several malignancies, among which is anaplastic thyroid carcinoma (ATC). We have previously demonstrated the in vitro efficacy of Aurora kinase inhibitors in restraining cell growth and survival of different ATC cell lines. In this study, we sought to establish which Aurora might represent the preferential drug target for ATC. To this end, the effects of two selective inhibitors of Aurora-A (MLN8237) and Aurora-B (AZD1152) on four human ATC cell lines (CAL-62, BHT-101, 8305C, and 8505C) were analysed. Both inhibitors reduced cell proliferation in a time- and dose-dependent manner, with IC50 ranges of 44.3-134.2 nM for MLN8237 and of 9.2-461.3 nM for AZD1152. Immunofluorescence experiments and time-lapse videomicroscopy yielded evidence that each inhibitor induced distinct mitotic phenotypes, but both of them prevented the completion of cytokinesis. As a result, poliploidy increased in all AZD1152-treated cells, and in two out of four cell lines treated with MLN8237. Apoptosis was induced in all the cells by MLN8237, and in BHT-101, 8305C, and 8505C by AZD1152, while CAL-62 exposed to AZD1152 died through necrosis after multiple rounds of endoreplication. Both inhibitors were capable of blocking anchorage-independent cell growth. In conclusion, we demonstrated that either Aurora-A or Aurora-B might represent therapeutic targets for the ATC treatment, but inhibition of Aurora-A appears more effective for suppressing ATC cell proliferation and for inducing the apoptotic pathway.
Collapse
Affiliation(s)
- Enke Baldini
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Chiara Tuccilli
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Natalie Prinzi
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Salvatore Sorrenti
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Alessandro Antonelli
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Lucio Gnessi
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Stefania Morrone
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Costanzo Moretti
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Marco Bononi
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Yannick Arlot-Bonnemains
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Massimino D'Armiento
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Salvatore Ulisse
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| |
Collapse
|
28
|
Che HY, Guo HY, Si XW, You QY, Lou WY. PP121, a dual inhibitor of tyrosine and phosphoinositide kinases, inhibits anaplastic thyroid carcinoma cell proliferation and migration. Tumour Biol 2014; 35:8659-64. [PMID: 24867098 DOI: 10.1007/s13277-014-2118-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/19/2014] [Indexed: 01/04/2023] Open
Abstract
The tyrosine and phosphoinositide kinases play crucial roles in the regulation of many cancer cell processes including cell survival and cell motility. Anaplastic thyroid carcinoma (ATC) is a rare and deadly type of thyroid cancer, and so far, there are no effective therapeutic compounds for ATC. Herein, we investigate the anticancer activities of PP121, a dual inhibitor of tyrosine and phosphoinositide kinases, in ATC therapy. We found that PP121 is effective at suppressing cell viability, inducing cell apoptosis, and inhibiting cell migration and invasion. The potential anticancer mechanism for PP121 might be its inhibitory effects on phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways in ATC cells. Furthermore, PP121 is effective at suppressing ATC tumor growth in vivo. In summary, our studies suggest that PP121 might be a promising therapeutic compound for ATC treatment, which might shed new light on ATC therapy.
Collapse
Affiliation(s)
- Huan-Yong Che
- Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568 Zhongxing North Road, Shaoxing, Zhejiang, 312000, People's Republic of China
| | | | | | | | | |
Collapse
|
29
|
Efficacy of combined treatment for anaplastic thyroid carcinoma: results of a multinstitutional retrospective analysis. Int J Surg 2014; 12 Suppl 1:S178-82. [PMID: 24866070 DOI: 10.1016/j.ijsu.2014.05.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 05/03/2014] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Anaplastic thyroid carcinoma (ATC) is a killer tumor, characterized by local invasiveness, risk of recurrence and very poor prognosis. Due to its rarity, clinical case studies concerning management are lacking. METHODS We retrospectively reviewed a multinstitutional clinical series of 114 consecutive patients treated between 1996 and 2012 for ATC. The outcomes of a combined treatment were analyzed considering the impact of surgery and radiotherapy on survival. RESULTS Patients were divided in groups A and B considering tumor size (tumor lesser and larger than 5 cm). Surgery was carried out in 71 patients, radiotherapy in 89 patients. Tracheostomy and endoprothesis were used respectively in 48.7% and in 25.6% of patients. The mean survival was 5.35 (±3.2) months with no significant difference in group A vs group B. A better survival was observed in both groups of patients undergone a surgical treatment compared to no treated patients (p = 0.001 and p = 0.0001) or to patients undergone radiotherapy alone (p = 0.047 and p = 0.0001). Combination of surgery and radiotherapy significantly improved outcome (p = 0.017). DISCUSSION Despite disappointing results from single therapeutic approach, multimodal strategy has progressively become the treatment of choice in ATC, with surgery being the cornerstone of the management. CONCLUSION Although dismal prognosis, the combined treatment might significantly improves locoregional disease control, achieving acceptable survival in selected patients and adequate palliation of the symptoms.
Collapse
|
30
|
Okada T, Nakamura T, Watanabe T, Onoda N, Ashida A, Okuyama R, Ito KI. Coexpression of EpCAM, CD44 variant isoforms and claudin-7 in anaplastic thyroid carcinoma. PLoS One 2014; 9:e94487. [PMID: 24727741 PMCID: PMC3984167 DOI: 10.1371/journal.pone.0094487] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/16/2014] [Indexed: 12/14/2022] Open
Abstract
Background Anaplastic thyroid cancer is considered to be one of the most aggressive human malignancies, and the mean survival time after diagnosis is approximately six months, regardless of treatments. This study aimed to examine how EpCAM and its related molecules are involved in the characteristics of anaplastic thyroid carcinoma. Methodology/Principal Findings Two differentiated thyroid cancer cell lines (TPC-1 and FTC-133), and two anaplastic thyroid cancer cell lines (FRO, ACT-1) were analyzed for expression of CD44 standard isoform (CD44s), CD44 variant isoforms, and EpCAM, and human aldehyde dehydrogenase-1 (ALDH1) enzymatic activity using flow cytometry. CD44s expression was higher in TPC-1 and FTC-133 than in the FRO and ACT-1, whereas ALDH1 activities were higher in FRO and ACT-1 than in TPC-1 and FTC-133. An inverse correlation between CD44s expression and ALDH1 activity was observed in all thyroid cancer cell lines. As for the expressions of CD44 variant isoforms, ACT-1 showed higher and FRO showed moderate CD44v6 expressions, whereas either TPC-1 or FTC-133 showed negative CD44v6 expression. EpCAM expressions in FRO and ACT-1 were higher than those in TPC-1 and FTC-133, and EpCAM expressions inversely correlated with those of CD44s. A positive correlation was observed between EpCAM expression and ALDH1 activity in thyroid cancer cell lines. In the RT-PCR analysis, the expression levels of EpCAM, caludin-7 and ALDH1 in FRO and ATC-1 cells were significantly higher than those in TPC-1 and FTC-133 cells. In clinical specimens of thyroid cancers, nuclear expression of EpCAM and high expression of CD44v6 were detected significantly more frequently in anaplastic carcinomas. Conclusions/Significance Our study suggests the possibility that EpCAM, together with CD44v6 and claudin-7 as well as ALDH1, may be involved in the development of the aggressive phenotype of anaplastic thyroid carcinoma. Our findings may suggest a novel therapeutic strategy for treatment of anaplastic thyroid carcinoma.
Collapse
MESH Headings
- Aldehyde Dehydrogenase 1 Family
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Differentiation
- Cell Line, Tumor
- Claudins/genetics
- Claudins/metabolism
- Epithelial Cell Adhesion Molecule
- Gene Expression Regulation, Neoplastic
- Humans
- Hyaluronan Receptors/metabolism
- Immunohistochemistry
- Isoenzymes/metabolism
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Retinal Dehydrogenase/metabolism
- Thyroid Carcinoma, Anaplastic/enzymology
- Thyroid Carcinoma, Anaplastic/genetics
- Thyroid Carcinoma, Anaplastic/metabolism
- Thyroid Carcinoma, Anaplastic/pathology
Collapse
Affiliation(s)
- Toshihiro Okada
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Teruo Nakamura
- Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takayuki Watanabe
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Naoyoshi Onoda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Atsuko Ashida
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Ryuhei Okuyama
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Ken-ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- * E-mail:
| |
Collapse
|
31
|
Antonelli A, Bocci G, Fallahi P, La Motta C, Ferrari SM, Mancusi C, Fioravanti A, Di Desidero T, Sartini S, Corti A, Piaggi S, Materazzi G, Spinelli C, Fontanini G, Danesi R, Da Settimo F, Miccoli P. CLM3, a multitarget tyrosine kinase inhibitor with antiangiogenic properties, is active against primary anaplastic thyroid cancer in vitro and in vivo. J Clin Endocrinol Metab 2014; 99:E572-81. [PMID: 24423321 DOI: 10.1210/jc.2013-2321] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CONTEXT AND OBJECTIVE We have studied the antitumor activity of a pyrazolo[3,4-d]pyrimidine compound (CLM3) proposed for a multiple signal transduction inhibition [including the RET tyrosine kinase, epidermal growth factor receptor, and vascular endothelial growth factor (VEGF) receptor and with antiangiogenic activity] in primary anaplastic thyroid cancer (ATC) cells, in the human cell line 8305C (undifferentiated thyroid cancer), and in an ATC-cell line (AF). DESIGN AND MAIN OUTCOME MEASURES CLM3 was tested in primary ATC cells at the concentrations of 5, 10, 30, and 50 μM; in 8305C cells, in AF cells, at 1, 5, 10, 30, 50, or 100 μM; and in AF cells in CD nu/nu mice. RESULTS CLM3 significantly inhibited the proliferation of 8305C and AF cells, also inducing apoptosis. A significant reduction of proliferation with CLM3 in ATC cells (P < .01, ANOVA) was shown. CLM3 increased the percentage of apoptotic ATC cells dose dependently (P < .001, ANOVA) and inhibited migration (P < .01) and invasion (P < .001). The AF cell line was injected sc in CD nu/nu mice, and tumor masses became detectable 15 days later. CLM3 (50 mg/kg per die) significantly inhibited tumor growth (starting 16 d after the beginning of treatment). CLM3 significantly decreased the VEGF-A expression and microvessel density in AF tumor tissues. Furthermore, CLM3 inhibited epidermal growth factor receptor, AKT, and ERK1/2 phosphorylation and down-regulated cyclin D1 in 8305C and AF cells. CONCLUSIONS The antitumor and antiangiogenic activity of a pyrazolo[3,4-d]pyrimidine compound (CLM3) is very promising in anaplastic thyroid cancer, opening the way to a future clinical evaluation.
Collapse
Affiliation(s)
- Alessandro Antonelli
- Departments of Clinical and Experimental Medicine (A.A., G.B., P.F., S.M.F., C.M., A.F., T.D.D., R.D.), Pharmacy (C.L.M., S.S., F.D.S.), Translational Research and New Technologies in Medicine and Surgery (A.C., S.P.), and Surgical, Medical, Molecular Pathology, and Critical Area (G.M., C.S., G.F., P.M.), University of Pisa, 56126 Pisa, Italy; and Istituto Toscano Tumori (G.B.), 50139 Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Che HY, Guo HY, Si XW, You QY, Lou WY. Additive effect by combination of Akt inhibitor, MK-2206, and PDGFR inhibitor, tyrphostin AG 1296, in suppressing anaplastic thyroid carcinoma cell viability and motility. Onco Targets Ther 2014; 7:425-32. [PMID: 24665203 PMCID: PMC3961587 DOI: 10.2147/ott.s57324] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The phosphatidylinositol-3-kinase/Akt pathway and receptor tyrosine kinases regulate many tumorigenesis related cellular processes including cell metabolism, cell survival, cell motility, and angiogenesis. Anaplastic thyroid carcinoma (ATC) is a rare type of thyroid cancer with no effective systemic therapy. It has been shown that Akt activation is associated with tumor progression in ATC. Here we observed the additive effect between an Akt inhibitor (MK-2206) and a novel platelet-derived growth factor receptor inhibitor (tyrphostin AG 1296) in ATC therapy. We found an additive effect between MK-2206 and tyrphostin AG 1296 in suppressing ATC cell viability. The combination of MK-2206 and tyrphostin AG 1296 induces additive apoptosis, additive suppression of the Akt signaling pathway, as well as additive inhibition of cell migration and invasion of ATC cells. Furthermore, the combination of MK-2206 and tyrphostin AG 1296 induced additive suppression of ATC tumor growth in vivo. In summary, our studies suggest that the combination of Akt and receptor tyrosine kinase inhibitors may be an efficient therapeutic strategy for ATC treatment, which might shed new light on ATC therapy.
Collapse
Affiliation(s)
- Huan-Yong Che
- Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang, People's Republic of China
| | - Hang-Yuan Guo
- Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang, People's Republic of China
| | - Xu-Wei Si
- Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang, People's Republic of China
| | - Qiao-Ying You
- Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang, People's Republic of China
| | - Wei-Ying Lou
- Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang, People's Republic of China
| |
Collapse
|
33
|
Overexpression of Aurora-C interferes with the spindle checkpoint by promoting the degradation of Aurora-B. Cell Death Dis 2014; 5:e1106. [PMID: 24603334 PMCID: PMC3973241 DOI: 10.1038/cddis.2014.37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/16/2014] [Accepted: 01/16/2014] [Indexed: 12/24/2022]
Abstract
The chromosomal passenger complex (CPC) plays a pivotal role in controlling accurate chromosome segregation and cytokinesis during cell division. Aurora-B, one of the chromosomal passenger proteins, is important for the mitotic spindle assembly checkpoint (SAC). Previous reports noted that Aurora-C is predominantly expressed in male germ cells and has the same subcellular localization as Aurora-B. Increasing evidence indicates that Aurora-C is overexpressed in many somatic cancers, although its function is uncertain. Our previous study showed that the aberrant expression of Aurora-C increases the tumorigenicity of cancer cells. Here, we demonstrate that overexpressed Aurora-C displaces the centromeric localization of CPCs, including INCENP, survivin, and Aurora-B. When cells were treated with nocodazole to turn on SAC, both the Aurora-B protein stability and kinase activity were affected by overexpressed Aurora-C. As a result, the activation of spindle checkpoint protein, BubR1, and phosphorylation of histone H3 and MCAK were also eliminated in Aurora-C-overexpressing cells. Thus, our results suggest that aberrantly expressed Aurora-C in somatic cancer cells may impair SAC by displacing the centromeric localization of CPCs.
Collapse
|
34
|
Chiu CG, St-Pierre P, Nabi IR, Wiseman SM. Autocrine motility factor receptor: a clinical review. Expert Rev Anticancer Ther 2014; 8:207-17. [DOI: 10.1586/14737140.8.2.207] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
35
|
Bossi P, Locati LD, Licitra L. Biological agents in head and neck cancer. Expert Rev Anticancer Ther 2014; 7:1643-50. [DOI: 10.1586/14737140.7.11.1643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
36
|
Parenti R, Salvatorelli L, Magro G. Anaplastic Thyroid Carcinoma: Current Treatments and Potential New Therapeutic Options with Emphasis on TfR1/CD71. Int J Endocrinol 2014; 2014:685396. [PMID: 25097549 PMCID: PMC4102021 DOI: 10.1155/2014/685396] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/15/2014] [Accepted: 06/17/2014] [Indexed: 12/24/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is one of the most aggressive human cancers. Actually, ATC is refractory to conventional therapies, including surgery, chemotherapy, radiotherapy, and radioiodine ((131)I) therapy. Accordingly, genetic and molecular characterizations of ATC have been frequently and periodically reviewed in order to identify potential biological markers exploitable for target therapy. This review briefly focuses on main molecular events that characterize ATC and provides an update about preclinical studies. In addition, the overexpression of transferrin receptor 1 (TfR1/CD71) by neoplastic cells of ATC is emphasized in that it could represent a potential therapeutic target. In this regard, new therapeutic approaches based on the use of monoclonal or recombinant antibodies, or transferrin-gallium-TfR1/CD71 molecular complexes, or lastly small interfering RNAs (siRNAs) are proposed.
Collapse
Affiliation(s)
- Rosalba Parenti
- Department of Bio-Medical Sciences, Physiology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- *Rosalba Parenti:
| | - Lucia Salvatorelli
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
| | - Gaetano Magro
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
| |
Collapse
|
37
|
Baldini E, D'Armiento M, Ulisse S. A new aurora in anaplastic thyroid cancer therapy. Int J Endocrinol 2014; 2014:816430. [PMID: 25097550 PMCID: PMC4106108 DOI: 10.1155/2014/816430] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/11/2014] [Indexed: 01/08/2023] Open
Abstract
Anaplastic thyroid cancers (ATC) are among the most aggressive human neoplasms with a dire prognosis and a median survival time of few months from the diagnosis. The complete absence of effective therapies for ATC renders the identification of novel therapeutic approaches sorely needed. Chromosomal instability, a feature of all human cancers, is thought to represent a major driving force in thyroid cancer progression and a number of mitotic kinases showing a deregulated expression in malignant thyroid tissues are now held responsible for thyroid tumor aneuploidy. These include the three members of the Aurora family (Aurora-A, Aurora-B, and Aurora-C), serine/threonine kinases that regulate multiple aspects of chromosome segregation and cytokinesis. Over the last few years, several small molecule inhibitors targeting Aurora kinases were developed, which showed promising antitumor effects against a variety of human cancers, including ATC, in preclinical studies. Several of these molecules are now being evaluated in phase I/II clinical trials against advanced solid and hematological malignancies. In the present review we will describe the structure, expression, and mitotic functions of the Aurora kinases, their implications in human cancer progression, with particular regard to ATC, and the effects of their functional inhibition on malignant cell proliferation.
Collapse
Affiliation(s)
- Enke Baldini
- Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Massimino D'Armiento
- Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Salvatore Ulisse
- Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- *Salvatore Ulisse:
| |
Collapse
|
38
|
Mochizuki Y, Ito KI, Izumi H, Kohno K, Amano J. Expression of polypeptide N-acetylgalactosaminyl transferase-3 and its association with clinicopathological factors in thyroid carcinomas. Thyroid 2013; 23:1553-60. [PMID: 23659732 DOI: 10.1089/thy.2012.0613] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Polypeptide N-acetylgalactosaminyl transferase-3 (GalNAc-T3) has been reportedly expressed in several human adenocarcinomas and is associated with clinicopathological features of tumors. We investigated the clinicopathological significance of GalNAc-T3 in thyroid cancer. METHODS We evaluated the expression of GalNAc-T3 in 167 patients with thyroid cancer using a specific antibody and analyzed the association between its expression and clinicopathological features. RESULTS GalNAc-T3 was expressed in 85.8% of normal follicular epithelial cells. In papillary carcinomas, positive staining was observed in 101 (73.7%) cases. Well-differentiated components (papillary and follicular) of papillary carcinomas were significantly more frequently positive than poorly differentiated components (trabecular and solid) (p<0.01), and GalNAc-T3 was highly expressed in papillary carcinomas that had invaded beyond the thyroid capsule (p=0.026). GalNAc-T3 was expressed in 40% and 20% of well and poorly differentiated components of follicular carcinomas, respectively. Thirteen of 15 anaplastic carcinomas were negative for GalNAc-T3 and thyroglobulin. Positive staining for GalNAc-T3 was not observed in any of the medullary carcinomas. CONCLUSIONS Our data suggest that GalNAc-T3 expression may be a useful indicator of tumor differentiation in thyroid carcinomas.
Collapse
Affiliation(s)
- Yasuhiro Mochizuki
- 1 Division of Breast and Endocrine Surgery, Shinshu University School of Medicine , Matsumoto, Nagano, Japan
| | | | | | | | | |
Collapse
|
39
|
Isham CR, Bossou AR, Negron V, Fisher KE, Kumar R, Marlow L, Lingle WL, Smallridge RC, Sherman EJ, Suman VJ, Copland JA, Bible KC. Pazopanib enhances paclitaxel-induced mitotic catastrophe in anaplastic thyroid cancer. Sci Transl Med 2013; 5:166ra3. [PMID: 23283368 DOI: 10.1126/scitranslmed.3004358] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Anaplastic thyroid cancer (ATC) has perhaps the worst prognosis of any cancer, with a median survival of only about 5 months regardless of stage. Pazopanib monotherapy has promising clinical activity in differentiated thyroid cancers (generally attributed to vascular endothelial growth factor receptor inhibition), yet has less effective single-agent activity in ATC. We now report that combining pazopanib with microtubule inhibitors such as paclitaxel produced heightened and synergistic antitumor effects in ATC cells and xenografts that were associated with potentiated mitotic catastrophe. We hypothesized that combined effects may reflect enhanced paclitaxel-induced cytotoxicity mediated by cell cycle regulatory kinase inhibition by pazopanib. Indeed, pazopanib potently inhibited aurora A, with pazopanib/paclitaxel synergy recapitulated by aurora A short hairpin RNA knockdown or by specific aurora A pharmacological inhibition. Pazopanib/paclitaxel synergy was reversed by aurora A knockdown. Moreover, aurora A (but not B or C) message and protein levels were significantly increased in patient ATCs, and durable benefit resulted from pilot clinical translation of pazopanib/paclitaxel therapy in a patient with metastatic ATC. Collectively, these results suggest that the pazopanib/paclitaxel combination is a promising candidate therapeutic approach in ATC and that aurora A may represent a potentially viable therapeutic molecular target in ATC.
Collapse
Affiliation(s)
- Crescent R Isham
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Fisher KE, Jani JC, Fisher SB, Foulks C, Hill CE, Weber CJ, Cohen C, Sharma J. Epidermal growth factor receptor overexpression is a marker for adverse pathologic features in papillary thyroid carcinoma. J Surg Res 2013; 185:217-24. [PMID: 23746767 DOI: 10.1016/j.jss.2013.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 04/28/2013] [Accepted: 05/01/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) overexpression (EGFR-H) is implicated in thyroid carcinoma disease progression; however, the clinicopathologic significance of EGFR-H in tumors that harbor EGFR and/or v-Raf murine sarcoma viral oncogene homolog B1 (BRAF)(V600E) mutations is unknown. METHODS Tissue microarrays from 81 patients who had undergone thyroidectomy for carcinoma from 2002-2011 were scored for EGFR expression using immunohistochemistry. Somatic mutations in EGFR exons 19 and 21 and BRAF were analyzed. Correlations between the EGFR immunohistochemistry, EGFR, and BRAF(V600E) mutations and the clinicopathologic features were assessed. RESULTS EGFR-H was detected in 39.5% of carcinomas (n = 32) from patients with papillary (PTC, 46.2%, n = 18), follicular (29.6%, n = 8), and anaplastic (100.0%, n = 6) but not medullary (0.0%, n = 9) thyroid carcinoma. BRAF(V600E) mutations were identified in 22.2% of the carcinoma cases (n = 18, 15 PTCs and 3 anaplastic thyroid carcinomas). No somatic EGFR mutations were detected in any subtype. On PTC univariate analysis, EGFR-H correlated with increasing stage, extrathyroid extension, tumor capsule invasion, adverse pathologic features (any demonstration of extrathyroid extension, tumor capsule invasion, lymphovascular invasion, lymph node metastasis, and/or distant metastasis), and BRAF(V600E) mutations. On multivariate analysis, EGFR-H correlated with BRAF(V600E) mutations. In BRAF wild-type PTCs, the correlation between EGFR-H and adverse pathologic features approached statistical significance (P = 0.065). CONCLUSIONS EGFR-H could be an important biomarker for aggressive PTCs, particularly in BRAF wild-type PTCs. Despite EGFR-H in PTC, follicular thyroid carcinoma, and anaplastic thyroid carcinoma by immunohistochemistry, somatic EGFR mutations were absent. Therefore, future investigations of EGFR should consider histologic and immunohistochemical methods, in addition to molecular profiling of thyroid carcinomas. This multimodal approach is particularly important for future clinical trials testing anti-EGFR therapy.
Collapse
Affiliation(s)
- Kevin E Fisher
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
BACKGROUND With our growing understanding of the immune system and mechanisms employed by tumors to evade destruction, the field of cancer immunotherapy has been revitalized. Concurrent inflammation has long been associated with follicular cell-derived thyroid cancer (FDTC). In the last decade, much research has focused on characterizing the tumor-associated immune response in patients with FDTC. SUMMARY Mast cells, natural killer cells, macrophages, dendritic cells, B cells, and T cells have been identified within FDTC-associated immune infiltrate. Collectively, these findings suggest that the immune response to FDTC is compromised and may even promote tumor progression. A more thorough characterization of the tumor-associated immune response in FDTC may lead to the development of immune-based adjuvant therapies for patients with aggressive disease. CONCLUSIONS Immune-based therapies could provide essential alternatives to patients that cannot be treated surgically, those with recurrent or persistent lymph node metastases, and those with anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Jena D French
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado , Aurora, CO 80045, USA.
| |
Collapse
|
42
|
Stickel JS, Mayer F, Vollmer JP, Zürn CS, Gawaz M, Kanz L, Bauer A. Ventricular thrombus and thrombocytopenia as first presentation of anaplastic thyroid carcinoma. J Clin Oncol 2013; 31:e194-6. [PMID: 23478056 DOI: 10.1200/jco.2012.43.4894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
43
|
Baquero P, Sánchez-Hernández I, Jiménez-Mora E, Orgaz JL, Jiménez B, Chiloeches A. (V600E)BRAF promotes invasiveness of thyroid cancer cells by decreasing E-cadherin expression through a Snail-dependent mechanism. Cancer Lett 2013; 335:232-41. [PMID: 23435375 DOI: 10.1016/j.canlet.2013.02.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/12/2013] [Indexed: 01/07/2023]
Abstract
BRAF is a main oncogene in human thyroid cancer. Here, we show that BRAF depletion by siRNA or inhibition of its activity by treatment with BRAF inhibitor PLX4720 decreases migration and invasion in thyroid cancer cells expressing oncogenic (V600E)BRAF through a MEK/ERK-dependent mechanism, since treatment with the MEK inhibitor U0126 exerts the same effect. Moreover, over-expression of (V600E)BRAF increases migration and invasion of wild-type BRAF thyroid cells. Using the same strategies, we demonstrate that these effects are mediated by upregulation of the transcriptional repressor Snail with a concomitant decrease of its target E-cadherin, both hallmarks of EMT. These results reveal a novel (V600E)BRAF-induced mechanism in thyroid tumours progression and provides a rationale for using the PLX4720 inhibitor to target (V600E)BRAF signalling to effectively control progression of thyroid cancer.
Collapse
Affiliation(s)
- Pablo Baquero
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
44
|
Bellelli R, Castellone MD, Garcia-Rostan G, Ugolini C, Nucera C, Sadow PM, Nappi TC, Salerno P, Cantisani MC, Basolo F, Gago TA, Salvatore G, Santoro M. FOXM1 is a molecular determinant of the mitogenic and invasive phenotype of anaplastic thyroid carcinoma. Endocr Relat Cancer 2012; 19:695-710. [PMID: 22919068 PMCID: PMC3637951 DOI: 10.1530/erc-12-0031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Anaplastic thyroid carcinoma (ATC) is a very aggressive thyroid cancer. forkhead box protein M1 (FOXM1) is a member of the forkhead box family of transcription factors involved in control of cell proliferation, chromosomal stability, angiogenesis, and invasion. Here, we show that FOXM1 is significantly increased in ATCs compared with normal thyroid, well-differentiated thyroid carcinomas (papillary and/or follicular), and poorly differentiated thyroid carcinomas (P=0.000002). Upregulation of FOXM1 levels in ATC cells was mechanistically linked to loss-of-function of p53 and to the hyperactivation of the phosphatidylinositol-3-kinase/AKT/FOXO3a pathway. Knockdown of FOXM1 by RNA interference inhibited cell proliferation by arresting cells in G2/M and reduced cell invasion and motility. This phenotype was associated with decreased expression of FOXM1 target genes, like cyclin B1 (CCNB1), polo-like kinase 1 (PLK1), Aurora B (AURKB), S-phase kinase-associated protein 2 (SKP2), and plasminogen activator, urokinase: uPA (PLAU). Pharmacological inhibition of FOXM1 in an orthotopic mouse model of ATC reduced tumor burden and metastasization. All together, these findings suggest that FOXM1 represents an important player in thyroid cancer progression to the anaplastic phenotype and a potential therapeutic target for this fatal cancer.
Collapse
Affiliation(s)
- Roberto Bellelli
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, 'L. Califano', Universita' Federico II, Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Soares P, Lima J, Preto A, Castro P, Vinagre J, Celestino R, Couto JP, Prazeres H, Eloy C, Máximo V, Sobrinho-Simões M. Genetic alterations in poorly differentiated and undifferentiated thyroid carcinomas. Curr Genomics 2012; 12:609-17. [PMID: 22654560 PMCID: PMC3271313 DOI: 10.2174/138920211798120853] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 08/29/2011] [Accepted: 08/30/2011] [Indexed: 12/20/2022] Open
Abstract
Thyroid gland presents a wide spectrum of tumours derived from follicular cells that range from well differentiated, papillary and follicular carcinoma (PTC and FTC, respectively), usually carrying a good prognosis, to the clinically aggressive, poorly differentiated (PDTC) and undifferentiated thyroid carcinoma (UTC).It is usually accepted that PDTC and UTC occur either de novo or progress from a pre-existing well differentiated carcinoma through a multistep process of genetic and epigenetic changes that lead to clonal expansion and neoplastic development. Mutations and epigenetic alterations in PDTC and UTC are far from being totally clarified. Assuming that PDTC and UTC may derive from well differentiated thyroid carcinomas (WDTC), it is expected that some PDTC and UTC would harbour genetic alterations that are typical of PTC and FTC. This is the case for some molecular markers (BRAF and NRAS) that are present in WDTC, PDTC and UTC. Other genes, namely P53, are almost exclusively detected in less differentiated and undifferentiated thyroid tumours, supporting a diagnosis of PDTC or, much more often, UTC. Thyroid-specific rearrangements RET/PTC and PAX8/PPARγ, on the other hand, are rarely found in PDTC and UTC, suggesting that these genetic alterations do not predispose cells to dedifferentiation. In the present review we have summarized the molecular changes associated with the two most aggressive types of thyroid cancer.
Collapse
Affiliation(s)
- Paula Soares
- Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), 4200-465 Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Smallridge RC. Approach to the patient with anaplastic thyroid carcinoma. J Clin Endocrinol Metab 2012; 97:2566-72. [PMID: 22869844 PMCID: PMC3410281 DOI: 10.1210/jc.2012-1314] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 04/18/2012] [Indexed: 11/19/2022]
Abstract
Anaplastic thyroid carcinoma is the least common but most lethal of thyroid cancers. All patients are classified as stage IV, with the primary lesion restricted to the thyroid gland in stage IVA; locoregional lymph nodes may exist in IVA/IVB; and IVC disease is defined by distant metastases. Prognosis is highly dependent on disease extent at presentation, and staging and establishing a plan of care must be accomplished quickly. Although almost all studies are biased due to their retrospective nature, the most important factors associated with longer survival are completeness of surgical resection (achievable in only a minority of patients) and high-dose (>40 Gy) external beam radiotherapy (preferably intensity modulated radiation therapy). Recent reports suggest that a multimodal approach (surgery, radiation, and chemotherapy) is beneficial. Given the high lethality even with apparent local disease, combination systemic therapy (cytotoxics and/or targeted agents) may improve outcomes in stage IVA/IVB patients. Newer, more effective drug combinations are urgently needed for IVC patients who want aggressive therapy. A candid discussion of the prognosis and management options, including palliative care/hospice, should be held with the patient and caregiver as soon as possible after diagnosis to clarify the patient's preference and expectations. Prospective multicenter clinical trials, incorporating molecular analyses of tumors, are required if we are to improve survival in anaplastic thyroid carcinoma.
Collapse
Affiliation(s)
- Robert C Smallridge
- Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida 32224, USA.
| |
Collapse
|
47
|
Kojic KL, Kojic SL, Wiseman SM. Differentiated thyroid cancers: a comprehensive review of novel targeted therapies. Expert Rev Anticancer Ther 2012; 12:345-57. [PMID: 22369326 DOI: 10.1586/era.12.8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Differentiated thyroid carcinoma (DTC) accounts for more than 90% of new thyroid cancer diagnoses, and includes papillary, follicular and Hürthle cell carcinoma. The prognosis for the vast majority of individuals diagnosed with DTC is excellent, with current treatment that includes surgery, radioactive iodine ablation and postoperative thyroid-stimulating hormone suppression. Unfortunately, the small proportion of individuals who develop radioactive iodine-resistant recurrent disease have few treatment options, and the vast majority will eventually die from their disease. Recently, several novel targets for anticancer agents have been identified and offer new hope for thyroid cancer patients diagnosed with progressive disease. In addition to targeting genes commonly altered in thyroid cancer, which include mutations in BRAF, RAS and RET, proangiogenic growth factor receptors and the sodium-iodide symporter have also been targeted. Several clinical trials evaluating tyrosine kinase and angiogenesis inhibitors for treatment of individuals diagnosed with metastatic or treatment-refractory DTC are currently underway. The objective of this review is to evaluate recent clinical trials that have studied novel targeted drugs for treatment of DTC.
Collapse
Affiliation(s)
- Katarina L Kojic
- St Paul's Hospital, Department of Surgery, University of British Columbia, C303-1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada
| | | | | |
Collapse
|
48
|
Shankar J, Wiseman SM, Meng F, Kasaian K, Strugnell S, Mofid A, Gown A, Jones SJM, Nabi IR. Coordinated expression of galectin-3 and caveolin-1 in thyroid cancer. J Pathol 2012; 228:56-66. [PMID: 22513979 DOI: 10.1002/path.4041] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 03/13/2012] [Accepted: 04/07/2012] [Indexed: 01/06/2023]
Abstract
Galectin-3 (Gal3) is the single most accurate marker for the diagnosis of differentiated thyroid cancer (DTC). Gal3 overrides the tumour suppressor activity of caveolin-1 (Cav1) and functions in concert with Cav1 to promote focal adhesion turnover and tumour cell migration and invasion. To study their coordinated role in progression of a human cancer, we investigated the expression of Gal3 and Cav1 in specimens of human benign thyroid lesions, DTC and anaplastic thyroid cancer (ATC). Gal3 and Cav1 expression is significantly associated with DTC and ATC, but not benign nodules. Essentially all Cav1-positive DTC cancers express Gal3, supporting the synergistic activity of these two proteins in DTC progression. Similarly, coordinated elevated Gal3/Cav1 expression was observed in three DTC-derived cell lines (papillary TCP1 and KTC1 and follicular FTC133) but only one (ACT1) of five ATC-derived cell lines. Using siRNA knockdown, Gal3 and Cav1 were shown to be required for RhoA GTPase activation, stabilization of focal adhesion kinase (FAK; a measure of focal adhesion signalling and turnover) and increased migration of the DTC cell lines studied, but not the ATC cell lines, including ACT1, which expresses elevated levels of Gal3 and Cav1. Co-expression of Gal3 and Cav1 in the T238 anaplastic cell line stabilized FAK-GFP in focal adhesions. Gal3 and Cav1 therefore function synergistically to promote focal adhesion signalling, migration and progression of DTC.
Collapse
Affiliation(s)
- Jay Shankar
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Evans JJ, Crist HS, Durvesh S, Bruggeman RD, Goldenberg D. A comparative study of cell cycle mediator protein expression patterns in anaplastic and papillary thyroid carcinoma. Cancer Biol Ther 2012; 13:776-81. [PMID: 22688732 DOI: 10.4161/cbt.20560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is an extremely aggressive and rapidly fatal neoplasm. The aim of this study was to identify a limited cell cycle associated protein expression pattern unique to ATC and to correlate that pattern with clinical outcome. This represents one of the largest tissue micro-array projects comparing the cell cycle protein expression data of ATC to other well-differentiated tumors in the literature. Tissue microarrays were created from 21 patients with ATC and an age and gender matched cohort of patients with papillary thyroid carcinoma (PTC). Expression of epidermal growth factor receptor, cyclin D1, cyclin E, p53, p21, p16, aurora kinase A, opioid growth factor (OGF), OGF-receptor, thyroglobulin and Ki-67 was evaluated in a semi-quantitative fashion. Differences in protein expression between the cohorts were evaluated using chi-square tests with Bonferroni adjustments. Survival time and presence of metastasis at presentation were collected. The ATC cohort showed a statistically significant decrease (p < 0.05) in thyroglobulin expression and statistically significant increases (p < 0.05) in Ki-67 and p53 expression as compared with the PTC cohort. A trend toward loss of p16 and p21 expression was noted in the ATC cohort. A trend toward decreased survival was noted with p21 expression. These data indicate disruption of the normal cell cycle with aberrant expression of multiple protein markers suggesting increased proliferative activity and loss of control of cell cycle progression to G₁ phase. These findings support the assertion that ATC may represent the furthest end of a continuum of thyroid carcinoma dedifferentiation.
Collapse
Affiliation(s)
- Juanita J Evans
- Department of Pathology, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | | | | | | | | |
Collapse
|
50
|
Lim SM, Shin SJ, Chung WY, Park CS, Nam KH, Kang SW, Keum KC, Kim JH, Cho JY, Hong YK, Cho BC. Treatment outcome of patients with anaplastic thyroid cancer: a single center experience. Yonsei Med J 2012; 53:352-7. [PMID: 22318823 PMCID: PMC3282954 DOI: 10.3349/ymj.2012.53.2.352] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Anaplastic thyroid cancer is known to have a poor prognosis due to its aggressive and rapid metastasis with median survival of less than 6 months. Multimodal treatment involving surgery and chemoradiotherapy has been used to improve the survival of patients. Here, we retrospectively review of treatment outcome of 13 consecutive patients who were treated at a single center. MATERIALS AND METHODS We retrospectively reviewed medical records of 13 anaplastic thyroid cancer patients who received multidisciplinary treatment between 2006 and 2010. Kaplan-Meier survival curve was used to analyze progression-free survival and overall survival of patients. RESULTS The median patient age at diagnosis was 69 years, and six patients had stage IVc diseases. Eight patients received primary surgery followed by radiotherapy or concurrent chemoradiotherapy (CCRT). Five patients received weekly doxorubicin-based definitive CCRT, but only one patient's condition remained stable, while the rest experienced rapid disease progression. The median progression-free survival was 2.8 months (95% CI, 1.2-4.4 months), and the median overall survival was 3.8 months (95% CI, 3.0-4.6 months). CONCLUSION Patients with anaplastic thyroid cancer showed poor prognosis despite multimodality treatment. Therefore, identification of novel therapeutic targets is warranted to take an effective mode of treatment.
Collapse
Affiliation(s)
- Sun Min Lim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sang-Joon Shin
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Woong Youn Chung
- Department of General Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Cheong Soo Park
- Department of General Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Kee-Hyun Nam
- Department of General Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Sang-Wook Kang
- Department of General Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Ki Chang Keum
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Joo Hang Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Yong Cho
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Department of Medical Oncology, Gangnam Severance Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | | | - Byoung Chul Cho
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|