51
|
Chen C, Liu J. Histone acetylation modifications: A potential targets for the diagnosis and treatment of papillary thyroid cancer. Front Oncol 2022; 12:1053618. [DOI: 10.3389/fonc.2022.1053618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/10/2022] [Indexed: 11/30/2022] Open
Abstract
Thyroid cancer is a common malignancy of the endocrine system, with papillary thyroid cancer (PTC) being the most common type of pathology. The incidence of PTC is increasing every year. Histone acetylation modification is an important part of epigenetics, regulating histone acetylation levels through histone acetylases and histone deacetylases, which alters the proliferation and differentiation of PTC cells and affects the treatment and prognosis of PTC patients. Histone deacetylase inhibitors induce histone acetylation, resulting in the relaxation of chromatin structure and activation of gene transcription, thereby promoting differentiation, apoptosis, and growth arrest of PTC cells.
Collapse
|
52
|
Oh JM, Gangadaran P, Rajendran RL, Hong CM, Lee J, Ahn BC. Different Expression of Thyroid-Specific Proteins in Thyroid Cancer Cells between 2-Dimensional (2D) and 3-Dimensional (3D) Culture Environment. Cells 2022; 11:3559. [PMID: 36428988 PMCID: PMC9688357 DOI: 10.3390/cells11223559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
The two-dimensional (2D) monolayer culture as a conventional method has been widely applied in molecular biology fields, but it has limited capability to recapitulate real cell environments, being prone to misinterpretation with poor prediction of in vivo behavior. Recently, the three-dimensional (3D) spheroid culture has been studied extensively. Spheroids are self-assembled cell aggregates that have biomimicry capabilities. The behavior of thyroid cancer under the 3D spheroid culture environment has been studied; however, there are no reports regarding differences in the degree of thyroid cancer cell differentiation under 2D and 3D culture conditions. This study investigated the expression of thyroid differentiation proteins related to iodide-metabolizing mechanisms in thyroid cancer cells under different culture conditions. Four thyroid cancer cell lines and one thyroid follicular epithelial cell line were grown in adherent 2D cell culture and 3D spheroid culture with agarose-coated plates. We observed changes in proliferation, hypoxia, extracellular matrix (ECM), cytoskeleton, thyroid-specific proteins, and thyroid transcription factors. All cell lines were successfully established in the spheroid following cell aggregation. Proliferation considerably decreased, while hypoxia-inducible factor 1-α(HIF1-α) was promoted in 3D spheroids; moreover, 3D spheroids with thyroid cancers showed diminished thyroid differentiation markers, but thyroid follicular epithelial cells revealed either a maintenance or weak decline of protein expression. We verified that the 3D spheroid culture environment can be similar to in vivo conditions because of its alterations in numerous cellular and functional activities, including morphology, cellular proliferation, viability, hypoxia, ECM, cytoskeleton, and thyroid differentiation, compared to the conventional 2D monolayer culture environment. An in vitro experimental study using 3D spheroid culture is ideal for the faster discovery of new drugs.
Collapse
Affiliation(s)
- Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea
| |
Collapse
|
53
|
Wang M, Li L, Xu Y, Du J, Ling C. Roles of hepatic stellate cells in NAFLD: From the perspective of inflammation and fibrosis. Front Pharmacol 2022; 13:958428. [PMID: 36313291 PMCID: PMC9606692 DOI: 10.3389/fphar.2022.958428] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become one of the most common diseases and severe problems worldwide because of the global increase in obesity, dyslipidemia, hypertension, and type 2 diabetes mellitus. NAFLD includes a wide spectrum of liver diseases, the histological forms of which range from non-alcoholic fatty liver (NAFL), which is generally nonprogressive, to non-alcoholic steatohepatitis (NASH), which can progress to chronic hepatitis, liver cirrhosis (LC), and sometimes hepatocellular carcinoma (HCC). Unlike NAFL, as the progressive form of NAFLD, NASH is characterized by the presence of inflammation with or without fibrosis in addition to hepatic steatosis. Although it is widely known and proved that persistent hepatic injury and chronic inflammation in the liver activate quiescent hepatic stellate cells (HSCs) and lead to hepatic fibrosis, the three-step process of “inflammation-fibrosis-carcinoma” in NAFLD has not been investigated and clarified clearly. In this process, the initiation of inflammation in the liver and the function of various liver inflammatory cells have been discussed regularly, while the activated HSCs, which constitute the principal cells responsible for fibrosis and their cross-talk with inflammation, seem not to be investigated specifically and frequently. Also, accumulated evidence suggests that HSCs can not only be activated by inflammation but also participate in the regulation of liver inflammation. Therefore, it is necessary to investigate the unique roles of HSCs in NAFLD from the perspective of inflammation and fibrosis. Here, we review the pivotal effects and mechanisms of HSCs and highlight the potential value of HSC-targeted treatment methods in NAFLD.
Collapse
Affiliation(s)
- Man Wang
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Lei Li
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yannan Xu
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Juan Du
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Changquan Ling
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
54
|
Zhang J, Zhao A, Jia X, Li X, Liang Y, Liu Y, Xie X, Qu X, Wang Q, Zhang Y, Gao R, Yu Y, Yang A. Sinomenine Hydrochloride Promotes TSHR-Dependent Redifferentiation in Papillary Thyroid Cancer. Int J Mol Sci 2022; 23:10709. [PMID: 36142613 PMCID: PMC9500915 DOI: 10.3390/ijms231810709] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Radioactive iodine (RAI) plays an important role in the diagnosis and treatment of papillary thyroid cancer (PTC). The curative effects of RAI therapy are not only related to radiosensitivity but also closely related to the accumulation of radionuclides in the lesion in PTC. Sinomenine hydrochloride (SH) can suppress tumor growth and increase radiosensitivity in several tumor cells, including PTC. The aim of this research was to investigate the therapeutic potential of SH on PTC cell redifferentiation. In this study, we treated BCPAP and TPC-1 cells with SH and tested the expression of thyroid differentiation-related genes. RAI uptake caused by SH-pretreatment was also evaluated. The results indicate that 4 mM SH significantly inhibited proliferation and increased the expression of the thyroid iodine-handling gene compared with the control group (p < 0.005), including the sodium/iodide symporter (NIS). Furthermore, SH also upregulated the membrane localization of NIS and RAI uptake. We further verified that upregulation of NIS was associated with the activation of the thyroid-stimulating hormone receptor (TSHR)/cyclic adenosine monophosphate (cAMP) signaling pathway. In conclusion, SH can inhibit proliferation, induce apoptosis, promote redifferentiation, and then increase the efficacy of RAI therapy in PTC cells. Thus, our results suggest that SH could be useful as an adjuvant therapy in combination with RAI therapy in PTC.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Aomei Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Xi Jia
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Xinru Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yiqian Liang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Xin Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Xijie Qu
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Qi Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yuemin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Rui Gao
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yan Yu
- Department of Public Health, Health Science Center of Xi’an Jiaotong University, Xi’an 710061, China
| | - Aimin Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
55
|
Li J, Zhang Y, Sun F, Xing L, Sun X. Towards an era of precise diagnosis and treatment: Role of novel molecular modification-based imaging and therapy for dedifferentiated thyroid cancer. Front Endocrinol (Lausanne) 2022; 13:980582. [PMID: 36157447 PMCID: PMC9493193 DOI: 10.3389/fendo.2022.980582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/04/2022] [Indexed: 12/07/2022] Open
Abstract
Dedifferentiated thyroid cancer is the major cause of mortality in thyroid cancer and is difficult to treat. Hence, the essential molecular mechanisms involved in dedifferentiation should be thoroughly investigated. Several studies have explored the biomolecular modifications of dedifferentiated thyroid cancer such as DNA methylation, protein phosphorylation, acetylation, ubiquitination, and glycosylation and the new targets for radiological imaging and therapy in recent years. Novel radionuclide tracers and drugs have shown attractive potential in the early diagnosis and treatment of dedifferentiated thyroid cancer. We summarized the updated molecular mechanisms of dedifferentiation combined with early detection by molecular modification-based imaging to provide more accurate diagnosis and novel therapeutics in the management of dedifferentiated thyroid cancer.
Collapse
Affiliation(s)
- Jing Li
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yingjie Zhang
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fenghao Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
56
|
A Potential Four-Gene Signature and Nomogram for Predicting the Overall Survival of Papillary Thyroid Cancer. DISEASE MARKERS 2022; 2022:8735551. [PMID: 36193505 PMCID: PMC9526076 DOI: 10.1155/2022/8735551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/14/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Background. Although the prognosis of papillary thyroid cancer (PTC) is relatively good, some patients experience recurrence or distant metastasis after thyroidectomy and progress to radioactive iodine refractory stage. Therefore, accurate prediction of clinical outlook can aid to screen out the minority of patients with poorer prognosis and avoid excessive treatment in low-risk patients. Methods. The RNA-seq and clinical data of PTC patients was downloaded from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) databases. Multivariate and Lasso Cox regression analyses were used to construct a prognostic nomogram to predict overall survival (OS). Thereafter, quantitative RT-PCR and Human Protein Atlas (HPA) database were employed to verify the expression of key genes. Results. A four-gene risk score comprising ABI3BP, DPT, MRO, and TENM1 was exhibited strong prognostic value. Moreover, an integrated nomogram was established based on the risk score, age, AJCC (American Joint Commission on Cancer) stage, tumor size, extrathyroidal extension, and history of neoadjuvant treatment, which exhibited significantly better predictive performance than TNM stage system (
). GSEA (Gene Set Enrichment Analysis) and GSVA (Gene Set Variation Analysis) revealed that the different tumor-associated hallmarks, biological processes, and pathways were substantially enriched in the poor-prognosis group. In addition, a ceRNA network was constructed by including the four genes (ABI3BP, DPT, MRO, and TENM1), 54 lncRNAs, and 10 miRNAs. Finally, both the relative mRNA and protein expression of ABI3BP, DPT, MRO, and TENM1 were validated. Conclusion. The present study identified a four-gene risk signature and developed a novel nomogram, which could be regarded as a reliable prognostic model for PTC patients. The findings also revealed preliminary potential mechanisms that may influence the prognosis outcome. These results can be conducive to design personalized treatment and prognosis management in affected patients.
Collapse
|
57
|
Sparano C, Moog S, Hadoux J, Dupuy C, Al Ghuzlan A, Breuskin I, Guerlain J, Hartl D, Baudin E, Lamartina L. Strategies for Radioiodine Treatment: What’s New. Cancers (Basel) 2022; 14:cancers14153800. [PMID: 35954463 PMCID: PMC9367259 DOI: 10.3390/cancers14153800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Radioiodine treatment (RAI) represents the most widespread and effective therapy for differentiated thyroid cancer (DTC). RAI goals encompass ablative (destruction of thyroid remnants, to enhance thyroglobulin predictive value), adjuvant (destruction of microscopic disease to reduce recurrences), and therapeutic (in case of macroscopic iodine avid lesions) purposes, but its use has evolved over time. Randomized trial results have enabled the refinement of RAI indications, moving from a standardized practice to a tailored approach. In most cases, low-risk patients may safely avoid RAI, but where necessary, a simplified protocol, based on lower iodine activities and human recombinant TSH preparation, proved to be just as effective, reducing overtreatment or useless impairment of quality of life. In pediatric DTC, RAI treatments may allow tumor healing even at the advanced stages. Finally, new challenges have arisen with the advancement in redifferentiation protocols, through which RAI still represents a leading therapy, even in former iodine refractory cases. RAI therapy is usually well-tolerated at low activities rates, but some concerns exist concerning higher cumulative doses and long-term outcomes. Despite these achievements, several issues still need to be addressed in terms of RAI indications and protocols, heading toward the RAI strategy of the future.
Collapse
Affiliation(s)
- Clotilde Sparano
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Service d’oncologie Endocrinienne, Département d’Imagerie Médicale, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Sophie Moog
- Service d’oncologie Endocrinienne, Département d’Imagerie Médicale, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Julien Hadoux
- Service d’oncologie Endocrinienne, Département d’Imagerie Médicale, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Corinne Dupuy
- UMR 9019 CNRS, Université Paris-Saclay, Gustave Roussy, 94800 Villejuif, France
| | - Abir Al Ghuzlan
- Département de Biologie et Pathologie Médicales, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Ingrid Breuskin
- Département Anesthésie Chirurgie et Interventionnel, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Joanne Guerlain
- Département Anesthésie Chirurgie et Interventionnel, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Dana Hartl
- Département Anesthésie Chirurgie et Interventionnel, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Eric Baudin
- Service d’oncologie Endocrinienne, Département d’Imagerie Médicale, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
| | - Livia Lamartina
- Service d’oncologie Endocrinienne, Département d’Imagerie Médicale, Gustave Roussy, 112 rue Edouard Vaillant, 94805 Villejuif, France
- Correspondence:
| |
Collapse
|
58
|
Liu L, Shi Y, Lai Q, Huang Y, Jiang X, Liu Q, Huang Y, Xia Y, Xu D, Jiang Z, Tu W. Construction of a Signature Model to Predict the Radioactive Iodine Response of Papillary Thyroid Cancer. Front Endocrinol (Lausanne) 2022; 13:865909. [PMID: 35634509 PMCID: PMC9132198 DOI: 10.3389/fendo.2022.865909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/28/2022] [Indexed: 12/04/2022] Open
Abstract
Papillary thyroid cancer (PTC) accounts for about 90% of thyroid cancer. There are approximately 20%-30% of PTC patients showing disease persistence/recurrence and resistance to radioactive iodine (RAI) treatment. For these PTC patients with RAI refractoriness, the prognosis is poor. In this study, we aimed to establish a comprehensive prognostic model covering multiple signatures to increase the predictive accuracy for progression-free survival (PFS) of PTC patients with RAI treatment. The expression profiles of mRNAs and miRNAs as well as the clinical information of PTC patients were extracted from TCGA and GEO databases. A series of bioinformatics methods were successfully applied to filtrate a two-RNA model (IPCEF1 and hsa-mir-486-5p) associated with the prognosis of RAI-therapy. Finally, the RNA-based risk score was calculated based on the Cox coefficient of the individual RNA, which achieved good performances by the time-dependent receiver operating characteristic (tROC) curve and PFS analyses. Furthermore, the predictive power of the nomogram, integrated with the risk score and clinical parameters (age at diagnosis and tumor stage), was assessed by tROC curves. Collectively, our study demonstrated high precision in predicting the RAI response of PTC patients.
Collapse
Affiliation(s)
- Lina Liu
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Yuhong Shi
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Qian Lai
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Yuan Huang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Xue Jiang
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Qian Liu
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Ying Huang
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Yuxiao Xia
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Dongkun Xu
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Zhiqiang Jiang
- Department of General Surgery, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Wenling Tu
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
59
|
Li C, Yuan Q, Xu G, Yang Q, Hou J, Zheng L, Wu G. A seven-autophagy-related gene signature for predicting the prognosis of differentiated thyroid carcinoma. World J Surg Oncol 2022; 20:129. [PMID: 35459137 PMCID: PMC9034603 DOI: 10.1186/s12957-022-02590-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/07/2022] [Indexed: 12/20/2022] Open
Abstract
Background Numerous studies have implicated autophagy in the pathogenesis of thyroid carcinoma. This investigation aimed to establish an autophagy-related gene model and nomogram that can help predict the overall survival (OS) of patients with differentiated thyroid carcinoma (DTHCA). Methods Clinical characteristics and RNA-seq expression data from TCGA (The Cancer Genome Atlas) were used in the study. We also downloaded autophagy-related genes (ARGs) from the Gene Set Enrichment Analysis website and the Human Autophagy Database. First, we assigned patients into training and testing groups. R software was applied to identify differentially expressed ARGs for further construction of a protein-protein interaction (PPI) network for gene functional analyses. A risk score-based prognostic risk model was subsequently developed using univariate Cox regression and LASSO-penalized Cox regression analyses. The model’s performance was verified using Kaplan-Meier (KM) survival analysis and ROC curve. Finally, a nomogram was constructed for clinical application in evaluating the patients with DTHCA. Finally, a 7-gene prognostic risk model was developed based on gene set enrichment analysis. Results Overall, we identified 54 differentially expressed ARGs in patients with DTHCA. A new gene risk model based on 7-ARGs (CDKN2A, FGF7, CTSB, HAP1, DAPK2, DNAJB1, and ITPR1) was developed in the training group and validated in the testing group. The predictive accuracy of the model was reflected by the area under the ROC curve (AUC) values. Univariate and multivariate Cox regression analysis indicated that the model could independently predict the prognosis of patients with THCA. The constrained nomogram derived from the risk score and age also showed high prediction accuracy. Conclusions Here, we developed a 7-ARG prognostic risk model and nomogram for differentiated thyroid carcinoma patients that can guide clinical decisions and individualized therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-022-02590-6.
Collapse
Affiliation(s)
- Chengxin Li
- Department of Breast & Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qianqian Yuan
- Department of Breast & Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Gaoran Xu
- Department of Breast & Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qian Yang
- Department of Breast & Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinxuan Hou
- Department of Breast & Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lewei Zheng
- Department of Breast & Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Gaosong Wu
- Department of Breast & Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
60
|
Transcription Factor CREB3L1 Regulates the Expression of the Sodium/Iodide Symporter (NIS) in Rat Thyroid Follicular Cells. Cells 2022; 11:cells11081314. [PMID: 35455992 PMCID: PMC9029047 DOI: 10.3390/cells11081314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023] Open
Abstract
The transcription factor CREB3L1 is expressed in a wide variety of tissues including cartilage, pancreas, and bone. It is located in the endoplasmic reticulum and upon stimulation is transported to the Golgi where is proteolytically cleaved. Then, the N-terminal domain translocates to the nucleus to activate gene expression. In thyroid follicular cells, CREB3L1 is a downstream effector of thyrotropin (TSH), promoting the expression of proteins of the secretory pathway along with an expansion of the Golgi volume. Here, we analyzed the role of CREB3L1 as a TSH-dependent transcriptional regulator of the expression of the sodium/iodide symporter (NIS), a major thyroid protein that mediates iodide uptake. We show that overexpression and inhibition of CREB3L1 induce an increase and decrease in the NIS protein and mRNA levels, respectively. This, in turn, impacts on NIS-mediated iodide uptake. Furthermore, CREB3L1 knockdown hampers the increase the TSH-induced NIS expression levels. Finally, the ability of CREB3L1 to regulate the promoter activity of the NIS-coding gene (Slc5a5) was confirmed. Taken together, our findings highlight the role of CREB3L1 in maintaining the homeostasis of thyroid follicular cells, regulating the adaptation of the secretory pathway as well as the synthesis of thyroid-specific proteins in response to TSH stimulation.
Collapse
|
61
|
Personalized Diagnosis in Differentiated Thyroid Cancers by Molecular and Functional Imaging Biomarkers: Present and Future. Diagnostics (Basel) 2022; 12:diagnostics12040944. [PMID: 35453992 PMCID: PMC9030409 DOI: 10.3390/diagnostics12040944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Personalized diagnosis can save unnecessary thyroid surgeries, in cases of indeterminate thyroid nodules, when clinicians tend to aggressively treat all these patients. Personalized diagnosis benefits from a combination of imagery and molecular biomarkers, as well as artificial intelligence algorithms, which are used more and more in our timeline. Functional imaging diagnosis such as SPECT, PET, or fused images (SPECT/CT, PET/CT, PET/MRI), is exploited at maximum in thyroid nodules, with a long history in the past and a bright future with many suitable radiotracers that could properly contribute to diagnosing malignancy in thyroid nodules. In this way, patients will be spared surgery complications, and apparently more expensive diagnostic workouts will financially compensate each patient and also the healthcare system. In this review we will summarize essential available diagnostic tools for malignant and benignant thyroid nodules, beginning with functional imaging, molecular analysis, and combinations of these two and other future strategies, including AI or NIS targeted gene therapy for thyroid carcinoma diagnosis and treatment as well.
Collapse
|
62
|
Oh JM, Rajendran RL, Gangadaran P, Hong CM, Jeong JH, Lee J, Ahn BC. Targeting GLI1 Transcription Factor for Restoring Iodine Avidity with Redifferentiation in Radioactive-Iodine Refractory Thyroid Cancers. Cancers (Basel) 2022; 14:1782. [PMID: 35406554 PMCID: PMC8997411 DOI: 10.3390/cancers14071782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022] Open
Abstract
Radioactive-iodine (RAI) therapy is the mainstay for patients with recurrent and metastatic thyroid cancer. However, many patients exhibit dedifferentiation characteristics along with lack of sodium iodide symporter (NIS) functionality, low expression of thyroid-specific proteins, and poor RAI uptake, leading to poor prognosis. Previous studies have demonstrated the effect of GLI family zinc finger 1 (GLI1) inhibition on tumor growth and apoptosis. In this study, we investigated the role of GLI1 in the context of redifferentiation and improvement in the efficacy of RAI therapy for thyroid cancer. We evaluated GLI1 expression in several thyroid cancer cell lines and selected TPC-1 and SW1736 cell lines showing the high expression of GLI. We performed GLI1 knockdown and evaluated the changes of thyroid-specific proteins expression, RAI uptake and I-131-mediated cytotoxicity. The effect of GANT61 (GLI1 inhibitor) on endogenous NIS expression was also assessed. Endogenous NIS expression upregulated by inhibiting GLI1, in addition, increased expression level in plasma membrane. Also, GLI1 knockdown increased expression of thyroid-specific proteins. Restoration of thyroid-specific proteins increased RAI uptake and I-131-mediated cytotoxic effect. Treatment with GANT61 also increased expression of endogenous NIS. Targeting GLI1 can be a potential strategy with redifferentiation for restoring RAI avidity in dedifferentiated thyroid cancers.
Collapse
Affiliation(s)
- Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Ju Hye Jeong
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| |
Collapse
|
63
|
Propensity Score-Matched Analysis to Identify Pathways Associated with Loss of Sodium Iodide Symporter in Papillary Thyroid Cancer. Curr Issues Mol Biol 2022; 44:1488-1496. [PMID: 35723359 PMCID: PMC9164071 DOI: 10.3390/cimb44040101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/19/2022] [Accepted: 03/22/2022] [Indexed: 01/02/2023] Open
Abstract
Sodium iodide symporter (NIS) expression in thyroid follicular cells plays an important role in normal physiology and radioactive iodine therapy for thyroid cancer. Loss of NIS expression is often seen in thyroid cancers and may lead to radioiodine refractoriness. To explore novel mechanisms of NIS repression beyond oncogenic drivers, clinical and RNA-seq data from the thyroid cancer dataset of The Cancer Genome Atlas were analyzed. Propensity score matching was used to control for various genetic background factors. We found that tumoral NIS expression was negatively correlated with tumor size. Additionally, low NIS expression was the only factor associated with recurrence-free survival in a Cox multivariate regression analysis. After matching for clinicopathologic profiles and driver mutations, the principal component analysis revealed distinct gene expressions between the high and low NIS groups. Gene set enrichment analysis suggested the downregulation of hedgehog signaling, immune networks, and cell adhesions. Positively enriched pathways included DNA replication, nucleotide excision repair, MYC, and Wnt/β-catenin pathways. In summary, we identified several potential targets which could be exploited to rescue the loss of NIS expression and develop redifferentiation strategies to facilitate radioactive iodine therapy for thyroid cancer.
Collapse
|
64
|
Wang W, Bai N, Li X. Comprehensive Analysis of the Prognosis and Drug Sensitivity of Differentiation-Related lncRNAs in Papillary Thyroid Cancer. Cancers (Basel) 2022; 14:1353. [PMID: 35267662 PMCID: PMC8909347 DOI: 10.3390/cancers14051353] [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: 12/21/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
Dedifferentiation is the main concern associated with radioactive iodine (RAI) refractoriness in patients with papillary thyroid cancer (PTC), and the underlying mechanisms of PTC dedifferentiation remain unclear. The present work aimed to identify a useful signature to indicate dedifferentiation and further explore its role in prognosis and susceptibility to chemotherapy drugs. A total of five prognostic-related DR-lncRNAs were selected to establish a prognostic-predicting model, and corresponding risk scores were closely associated with the infiltration of immune cells and immune checkpoint blockade. Moreover, we built an integrated nomogram based on DR-lncRNAs and age that showed a strong ability to predict the 3- and 5-year overall survival. Interestingly, drug sensitivity analysis revealed that the low-risk group was more sensitive to Bendamustine and TAS-6417 than the high-risk group. In addition, knockdown of DR-lncRNAs (DPH6-DT) strongly promoted cell proliferation, invasion, and migration via PI3K-AKT signal pathway in vitro. Furthermore, DPH6-DT downregulation also increased the expression of vimentin and N-cadherin during epithelial-mesenchymal transition. This study firstly confirms that DR-lncRNAs play a vital role in the prognosis and immune cells infiltration in patients with PTC, as well as a predictor of the drugs' chemosensitivity. Based on our results, DR-lncRNAs can serve as a promising prognostic biomarkers and treatment targets.
Collapse
Affiliation(s)
- Wenlong Wang
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ning Bai
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Xinying Li
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
65
|
Giovanella L, Deandreis D, Vrachimis A, Campenni A, Petranovic Ovcaricek P. Molecular Imaging and Theragnostics of Thyroid Cancers. Cancers (Basel) 2022; 14:1272. [PMID: 35267580 PMCID: PMC8909041 DOI: 10.3390/cancers14051272] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging plays an important role in the evaluation and management of different thyroid cancer histotypes. The existing risk stratification models can be refined, by incorporation of tumor-specific molecular markers that have theranostic power, to optimize patient-specific (individualized) treatment decisions. Molecular imaging with varying radioisotopes of iodine (i.e., 131I, 123I, 124I) is an indispensable component of dynamic and theragnostic risk stratification of differentiated carcinoma (DTC) while [18F]F-fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) helps in addressing disease aggressiveness, detects distant metastases, and risk-stratifies patients with radioiodine-refractory DTC, poorly differentiated and anaplastic thyroid cancers. For medullary thyroid cancer (MTC), a neuroendocrine tumor derived from thyroid C-cells, [18F]F-dihydroxyphenylalanine (6-[18F]FDOPA) PET/CT and/or [18F]FDG PET/CT can be used dependent on serum markers levels and kinetics. In addition to radioiodine therapy for DTC, some theragnostic approaches are promising for metastatic MTC as well. Moreover, new redifferentiation strategies are now available to restore uptake in radioiodine-refractory DTC while new theragnostic approaches showed promising preliminary results for advanced and aggressive forms of follicular-cell derived thyroid cancers (i.e., peptide receptor radiotherapy). In order to help clinicians put the role of molecular imaging into perspective, the appropriate role and emerging opportunities for molecular imaging and theragnostics in thyroid cancer are discussed in our present review.
Collapse
Affiliation(s)
- Luca Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Desiree’ Deandreis
- Division of Nuclear Medicine, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10126 Turin, Italy;
| | - Alexis Vrachimis
- Department of Nuclear Medicine, German Oncology Center, University Hospital of the European University, Limassol 4108, Cyprus;
| | - Alfredo Campenni
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98121 Messina, Italy;
| | - Petra Petranovic Ovcaricek
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, 10000 Zagreb, Croatia;
| |
Collapse
|
66
|
Zhang Y, Zou W, Zhu X, Jiang L, Gui C, Fan Q, Tu Y, Chen J. UPDATED UNDERSTANDING OF THE MOLECULAR TARGETS OF RADIOIODINE IN DIFFERENTIATED THYROID CANCER. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2022; 18:86-92. [PMID: 35975265 PMCID: PMC9365402 DOI: 10.4183/aeb.2022.86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Radioactive iodine (RAI) therapy is a mainstay adjuvant treatment for thyroid cancer. Administration of RAI therapy after total or near-total thyroidectomy has shown a survival advantage in numerous properly selected patients. However, the role of RAI therapy after reoperation for persistent or recurrent differentiated thyroid carcinomas (DTCs) is unclear. One reason may be the possible downregulation of the I- transport system after primary surgery. RAI is transported by the sodium iodide symporter (NIS), PENDRIN, anoctamin 1 (ANO1) and cystic fibrosis transmembrane conductance regulator (CFTR) and emits β particles that destroy follicular cells. The identification of pathways of iodide (I-) transport has allowed use of the transport system to render tumours susceptible to RAI treatment via gene therapy. This review focuses on the effect of RAI therapy in follicular cell-derived thyroid cancers and offers potential novel targets that enable improved radioiodine uptake and thus an improved prognosis of thyroid cancer.
Collapse
Affiliation(s)
- Y. Zhang
- Hubei Cancer Hospital - Department of Head and Neck Surgery, Wuhan, China
| | - W. Zou
- First People’s Hospital of Yichang - Department of General Surgery II, Yichang, Hubei, China
| | - X. Zhu
- Hubei Cancer Hospital - Department of Head and Neck Surgery, Wuhan, China
| | - L. Jiang
- Hubei Cancer Hospital - Department of Head and Neck Surgery, Wuhan, China
| | - C. Gui
- Hubei Cancer Hospital - Department of Head and Neck Surgery, Wuhan, China
| | - Q. Fan
- Hubei Cancer Hospital - Department of Head and Neck Surgery, Wuhan, China
| | - Y. Tu
- Liuzhou Traditional Chinese Medical Hospital - Department of Otolaryngology & Head and Neck Surgery, Liuzhou, Guangxi, China
| | - J. Chen
- Hubei Cancer Hospital - Department of Head and Neck Surgery, Wuhan, China
| |
Collapse
|
67
|
Huang S, Qi M, Tian T, Dai H, Tang Y, Huang R. Positive BRAFV600E mutation of primary tumor influences radioiodine avidity but not prognosis of papillary thyroid cancer with lung metastases. Front Endocrinol (Lausanne) 2022; 13:959089. [PMID: 36407316 PMCID: PMC9666419 DOI: 10.3389/fendo.2022.959089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
PURPOSE This study investigated the relationship between BRAFV600E mutation of the primary tumor and radioiodine avidity in lung metastases (LMs) and then further evaluated the impact of BRAFV600E mutation and radioiodine avidity status on the prognosis of papillary thyroid cancer (PTC) with LMs. METHODS Ninety-four PTC patients with LMs after total thyroidectomy and cervical lymph node dissection between January 2012 and September 2021 were retrospectively included. All patients received BRAFV600E mutation examination of primary tumors and radioactive iodine (RAI) therapy. The therapeutic response was evaluated by Response Evaluation Criteria in Solid Tumors (RECIST) assessments (version 1.1). For patients with target lesions, the response was divided into complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD); for patients without target lesions, the response was divided into CR, non-CR/non-PD, and PD. In therapeutic response, PR and SD were classified as non-CR/non-PD for analysis. The chi-square test and logistic regression were used to analyze the impact factor on PD and mortality. Progression-free survival (PFS) and overall survival (OS) curves were constructed by the Kaplan-Meier method. RESULTS It was found that 21.2% (7/33) of patients with positive BRAFV600E mutation and 62.3% (38/61) of patients with negative BRAFV600E mutation had radioiodine-avid LMs (χ2 = 14.484, p = 0.000). Patients with positive BRAFV600E mutation are more likely to lose radioiodine avidity; the odds ratios (ORs) were 5.323 (95% CI: 1.953-14.514, p = 0.001). Finally, 25 patients had PD, and six patients died; loss of radioiodine avidity was the independent predictor for PD, and the ORs were 10.207 (95% CI: 2.629-39.643, p = 0.001); BRAFV600E mutation status was not correlated with PD (p = 0.602), whether in the radioiodine avidity group (p = 1.000) or the non-radioiodine avidity group (p = 0.867). Similarly, BRAFV600E mutation status was not correlated with mortality; only loss of radioiodine avidity was the unfavorable factor associated with mortality in univariate analyses (p = 0.030). CONCLUSION Patients with LMs of PTC were more likely to lose radioiodine avidity when their primary tumor had positive BRAFV600E mutation; however, only radioiodine avidity and not BRAFV600E mutation status affected the clinical outcome of patients with lung metastatic PTC.
Collapse
Affiliation(s)
- Shuhui Huang
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Mengfang Qi
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Tian Tian
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Hongyuan Dai
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yuan Tang
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Rui Huang, ; Yuan Tang,
| | - Rui Huang
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Rui Huang, ; Yuan Tang,
| |
Collapse
|
68
|
Zhao H, Gong Y. Radioactive iodine in low- to intermediate-risk papillary thyroid cancer. Front Endocrinol (Lausanne) 2022; 13:960682. [PMID: 36034423 PMCID: PMC9402902 DOI: 10.3389/fendo.2022.960682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/21/2022] [Indexed: 11/22/2022] Open
Abstract
It remains controversial whether papillary thyroid cancer (PTC) patients with low- to intermediate-risk disease should receive radioactive iodine (RAI) after total thyroidectomy (TT). We aim to identify those who might benefit from RAI treatment in PTC patients with cervical nodal metastasis after TT. Patients were divided into TT and TT+RAI groups from the Surveillance, Epidemiology, and End Results (SEER) database (2004-2018). Overall survival (OS) and cancer-specific survival (CSS) were compared, and propensity score matching (PSM) was performed between groups. A total of 15,179 patients were enrolled, including 3,387 (22.3%) who underwent TT and 11,792 (77.7%) who received TT+RAI. The following characteristics were more likely to present in the TT+RAI group: multifocality, capsular extension, T3, N1b, and more metastatic cervical lymph nodes. RAI was associated with better OS in low- to intermediate-risk PTC patients in the multivariate Cox regression model. The subgroup analysis showed that RAI predicted better OS in patients ≥55 years, American Joint Committee on Cancer (AJCC) stage II, and capsular extension with a hazard ratio (HR) (95% CI) of 0.57 (0.45-0.72), 0.57 (0.45-0.72), and 0.68 (0.51-0.91), respectively. However, RAI failed to improve the prognoses of patients with age <55 years, AJCC stage I, PTC ≤1 cm, and capsular invasion. In the PSM cohort with 3,385 paired patients, TT+RAI treatment predicted better OS compared with TT alone. In addition, TT+RAI predicted better OS in patients with metastatic cervical lymph nodes ≥2, multifocality, extracapsular extension, and American Thyroid Association (ATA) intermediate risk. In conclusion, RAI was associated with better OS in low- to intermediate-risk PTC patients with age ≥55 years, multifocality, extrathyroidal extension, and ATA intermediate risk. However, the survival benefit from RAI may be limited in patients with AJCC stage I, PTC ≤1 cm, unifocality, capsular invasion, and ATA low-risk diseases; these patients even showed pathological cervical lymph node metastasis.
Collapse
Affiliation(s)
| | - Yiping Gong
- *Correspondence: Hengqiang Zhao, ; Yiping Gong,
| |
Collapse
|
69
|
Sa R, Liang R, Qiu X, He Z, Liu Z, Chen L. IGF2BP2-dependent activation of ERBB2 signaling contributes to acquired resistance to tyrosine kinase inhibitor in differentiation therapy of radioiodine-refractory papillary thyroid cancer. Cancer Lett 2021; 527:10-23. [PMID: 34896211 DOI: 10.1016/j.canlet.2021.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/25/2021] [Accepted: 12/05/2021] [Indexed: 02/08/2023]
Abstract
Acquired drug resistance represents a major obstacle to tyrosine kinase inhibitor (TKI)-induced differentiation therapy of radioiodine-refractory papillary thyroid cancer (RR-PTC); thus, there is an urgent need to elucidate the underlying mechanisms. Here, selumetinib-resistant PTC (PTCSR) cell lines, which were characterized by loss of sodium/iodide symporter expression, enhanced insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2), and activated V-Erb-B2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2) signaling, were initially established using a dose escalation method. Upon knockdown of IGF2BP2 in PTCSR cells, ERBB2 signaling was inhibited, and the acquired drug resistance was partially reversed. Mechanistically, the luciferase activity assay showed that IGF2BP2 bound to the N6-methyladenosine-binding site in the coding sequence of ERBB2 mRNA, yielding an increased ERBB2 translation efficacy revealed by polysome profiling. Inhibition of ERBB2 and IGF2BP2 by lapatinib robustly rescued the PTCSR cells from acquired dedifferentiation. Our study demonstrated that IGF2BP2-dependent ERBB2 signaling activation contributes to acquired resistance to TKI, which may be a promising differentiation strategy for RR-PTC by targeting IGF2BP2.
Collapse
Affiliation(s)
- Ri Sa
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Rd, Shanghai, 200233, People's Republic of China; Department of Nuclear Medicine, The First Hospital of Jilin University, 1# Xinmin St, Changchun, 130021, People's Republic of China.
| | - Rui Liang
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, People's Republic of China.
| | - Xian Qiu
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Rd, Shanghai, 200233, People's Republic of China.
| | - Ziyan He
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Rd, Shanghai, 200233, People's Republic of China.
| | - Zhiyan Liu
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Rd, Shanghai, 200233, People's Republic of China.
| | - Libo Chen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Rd, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
70
|
MAPK Inhibition Requires Active RAC1 Signaling to Effectively Improve Iodide Uptake by Thyroid Follicular Cells. Cancers (Basel) 2021; 13:cancers13225861. [PMID: 34831012 PMCID: PMC8616057 DOI: 10.3390/cancers13225861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/19/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The Sodium/Iodide Simulator (NIS) is responsible for the uptake of iodide in the thyroid follicular cells. NIS is present in most differentiated thyroid carcinomas (DTC), allowing radioactive iodine (RAI) to be used to destroy malignant cells. However, a significant proportion of DTCs stop picking up iodide and become resistant to RAI therapy. This is mainly due to the symporter no longer being produced or not being placed correctly at the cell’s membrane. This has been associated with mechanisms linked to malignant transformation, namely the overactivation of the so-called MAPK pathway. Thus, several drugs have been developed to inhibit this pathway, attempting to increase NIS levels and iodide uptake. However, MAPK inhibitors have had only partial success in restoring NIS expression. We found that the activity of another protein, the small GTPase RAC1, has an important role in this process, determining the outcome of MAPK inhibitors. Thus, our findings open new opportunities to find effective therapeutic alternatives for DTC resistant to RAI. Abstract The Sodium/Iodide Symporter (NIS) is responsible for the active transport of iodide into thyroid follicular cells. Differentiated thyroid carcinomas (DTCs) usually preserve the functional expression of NIS, allowing the use of radioactive iodine (RAI) as the treatment of choice for metastatic disease. However, a significant proportion of patients with advanced forms of TC become refractory to RAI therapy and no effective therapeutic alternatives are available. Impaired iodide uptake is mainly caused by the defective functional expression of NIS, and this has been associated with several pathways linked to malignant transformation. MAPK signaling has emerged as one of the main pathways implicated in thyroid tumorigenesis, and its overactivation has been associated with the downregulation of NIS expression. Thus, several strategies have been developed to target the MAPK pathway attempting to increase iodide uptake in refractory DTC. However, MAPK inhibitors have had only partial success in restoring NIS expression and, in most cases, it remained insufficient to allow effective treatment with RAI. In a previous work, we have shown that the activity of the small GTPase RAC1 has a positive impact on TSH-induced NIS expression and iodide uptake in thyroid cells. RAC1 is a downstream effector of NRAS, but not of BRAF. Therefore, we hypothesized that the positive regulation induced by RAC1 on NIS could be a relevant signaling cue in the mechanism underlying the differential response to MEK inhibitors, observed between NRAS- and BRAF-mutant tumors. In the present study, we found that the recovery of NIS expression induced through MAPK pathway inhibition can be enhanced by potentiating RAC1 activity in thyroid cell systems. The negative impact on NIS expression induced by the MAPK-activating alterations, NRAS Q61R and BRAF V600E, was partially reversed by the presence of the MEK 1/2 inhibitors AZD6244 and CH5126766. Notably, the inhibition of RAC1 signaling partially blocked the positive impact of MEK inhibition on NIS expression in NRAS Q61R cells. Conversely, the presence of active RAC1 considerably improved the rescue of NIS expression in BRAF V600E thyroid cells treated with MEK inhibitors. Overall, our data support an important role for RAC1 signaling in enhancing MAPK inhibition in the context of RAI therapy in DTC, opening new opportunities for therapeutic intervention.
Collapse
|
71
|
Chang A, Ling J, Ye H, Zhao H, Zhuo X. Enhancement of nanoparticle-mediated double suicide gene expression driven by 'E9-hTERT promoter' switch in dedifferentiated thyroid cancer cells. Bioengineered 2021; 12:6572-6578. [PMID: 34506254 PMCID: PMC8806866 DOI: 10.1080/21655979.2021.1974648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Differentiated thyroid cancer (DTC), such as papillary thyroid cancer, has a good prognosis after routine treatment. However, in the course of treatment, 5% to 20% of cases may dedifferentiate and can be transformed into dedifferentiated DTC (deDTC) or anaplastic thyroid cancer, leading to treatment failure. To date, several drugs have been used effectively for dedifferentiated thyroid cancer, whereas gene therapy may be a potential method. Literature reported that double suicide genes driven by human telomerase reverse transcriptase promoter (hTERTp) can specifically express in cancer cells and kill them. However, the weak activity of hTERTp limits its further research. To overcome this weakness, we constructed a novel chitosan nanocarrier containing double suicide genes driven by a ‘gene switch’ (a cascade of radiation enhancer E9 and a hTERTp). The vector was labeled with iodine-131 (131I). On one hand, E9 can significantly enhance the activity of hTERTp under the weak radiation of 131I, thereby increasing the expression of double suicide genes in deDTC cells. On the other hand, 131I also plays a certain killing role when it enters host cells. The proposed nanocarrier has good specificity for deDTC cells and thus deserves further study.
Collapse
Affiliation(s)
- Aoshuang Chang
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Junjun Ling
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Department of Oncology, Chongqing Institute of Traditional Chinese Medicine, Chongqing, China
| | - Huiping Ye
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Houyu Zhao
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xianlu Zhuo
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
72
|
Early Predictive Response to Multi-Tyrosine Kinase Inhibitors in Advanced Refractory Radioactive-Iodine Differentiated Thyroid Cancer: A New Challenge for [ 18F]FDG PET/CT. Diagnostics (Basel) 2021; 11:diagnostics11081417. [PMID: 34441351 PMCID: PMC8392185 DOI: 10.3390/diagnostics11081417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 11/22/2022] Open
Abstract
Differentiated thyroid cancer (DTC) represents the most common thyroid cancer histotype. Generally, it exhibits a good prognosis after conventional treatments; nevertheless, about 20% of patients can develop a local recurrence and/or distant metastasis. In one-third of advanced DTC, the metastatic lesions lose the ability to take up iodine and become radioactive iodine-refractory (RAI-R) DTC. In this set of patients, the possibility to perform localized treatments should always be taken into consideration before the initiation of systemic therapy. In the last decade, some multi-tyrosine kinase inhibitor (MKI) drugs were approved for advanced DTC, impacting on patient’s survival rate, but at the same time, these therapies have been associated with several adverse events. In this clinical context, the role of 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography/computed tomography ([18F]FDG PET/CT) in the early treatment response to these innovative therapies was investigated, in order to assess the potentiality of this diagnostic tool in the early recognition of non-responders, avoiding unnecessary therapy. Herein, we aimed to present a critical overview about the reliability of [18F]FDG PET/CT in the early predictive response to MKIs in advanced differentiated thyroid cancer.
Collapse
|
73
|
Jin Y, Liu B, Younis MH, Huang G, Liu J, Cai W, Wei W. Next-Generation Molecular Imaging of Thyroid Cancer. Cancers (Basel) 2021; 13:3188. [PMID: 34202358 PMCID: PMC8268517 DOI: 10.3390/cancers13133188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
An essential aspect of thyroid cancer (TC) management is personalized and precision medicine. Functional imaging of TC with radioiodine and [18F]FDG has been frequently used in disease evaluation for several decades now. Recently, advances in molecular imaging have led to the development of novel tracers based on aptamer, peptide, antibody, nanobody, antibody fragment, and nanoparticle platforms. The emerging targets-including HER2, CD54, SHP2, CD33, and more-are promising targets for clinical translation soon. The significance of these tracers may be realized by outlining the way they support the management of TC. The provided examples focus on where preclinical investigations can be translated. Furthermore, advances in the molecular imaging of TC may inspire the development of novel therapeutic or theranostic tracers. In this review, we summarize TC-targeting probes which include transporter-based and immuno-based imaging moieties. We summarize the most recent evidence in this field and outline how these emerging strategies may potentially optimize clinical practice.
Collapse
Affiliation(s)
- Yuchen Jin
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, China
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Beibei Liu
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliatede to Shanghai Jiao Tong University, Shanghai 200233, China;
| | - Muhsin H. Younis
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| |
Collapse
|