1
|
Wan P, Zhang D, Liu X, Lu W. Arid4a Suppresses Breast Tumor Metastasis by Enhancing MTSS1 Expression via mRNA Stability. Cancer Med 2025; 14:e70732. [PMID: 40066676 PMCID: PMC11894439 DOI: 10.1002/cam4.70732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Tumor metastasis is one of the main causes of death in cancer patients; however, the mechanism controlling metastasis is unclear. The posttranscriptional regulation of metastasis-related genes mediated by AT-rich interactive domain-containing protein 4A (Arid4a), an RNA-binding protein (RBP), has not been elucidated. METHODS Bioinformatic analysis, qRT-PCR, immunohistochemistry, and immunoblotting were employed to determine the expression of Arid4a in breast tumor tissues and its association with the survival of cancer patients. In vitro and in vivo cellular experiments were used to assess the function of Arid4a in breast tumor metastasis. PCR array, RNA immunoprecipitation (RIP), luciferase, mRNA stability, RIP-ChIP, and EMSA were conducted to elucidate the potential mechanism of Arid4a. RESULTS Reduced expression of Arid4a in breast tumor samples was detected via bioinformatic analyses and experimental methods. Low Arid4a expression was significantly correlated with poor prognosis in breast cancer patients. Gain-of-function and silencing experiments confirmed the inhibitory effect of Arid4a on tumor metastasis in vitro and in vivo. Mechanistically, Arid4a preferentially stabilizes metastasis-suppressing transcripts, including metastasis suppressor 1 (MTSS1), tissue inhibitor of metalloproteinase 2 (TIMP2), retinoblastoma 1 (Rb1), and phosphatase and tensin homolog (PTEN), through binding to a conserved structural RNA element localized in the 3' untranslated region (3'UTR). The Arid domain of Arid4a is required for its mRNA stabilization and metastasis inhibition. Notably, the expression of Arid4a and metastasis-suppressing genes was positively correlated in human breast tumor tissues. CONCLUSIONS Arid4a was confirmed to suppress breast tumor metastasis progression by stabilizing the transcripts of tumor metastasis-suppressing genes, suggesting that Arid4a might be a potential therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Pengfei Wan
- Department of Geriatric MedicineQilu Hospital of Shandong UniversityJinanShandongChina
| | - Dandan Zhang
- Department of Geriatric MedicineQilu Hospital of Shandong UniversityJinanShandongChina
| | - Xueting Liu
- Institute of MicrocirculationChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Wenbao Lu
- Institute of MicrocirculationChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Lin YB, Hu HW, Chung AK, Lu JY, Wu WC, Chiu IH, Chu I, Lin CC, Lee JH, Nien FJ, Chen KY, Wu MH, Chen CN, Wang CW, Kuo TC, Lin CH, Cheng MF, Chiu WY, Kuo SW, Hsih WH, Wang CY, Yang WS, Chen PL, Shih SR. Genomic Differences in Thyroid Cancers From Primary Sites Versus Distant Metastases in Individual Patients: A Clinical Perspective and Preliminary Report. Head Neck 2025. [PMID: 39936351 DOI: 10.1002/hed.28100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/27/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Distant metastasis is a leading cause of thyroid cancer (TC)-related deaths. Genetic profiling is typically limited to one sample per patient due to cost and sampling-risk concerns. Differences between samples from thyroid and distant metastasis within individual patients are unclear. METHODS Patients with TC and distant metastasis were recruited for genetic analysis. RESULTS Using a TC-specific NGS panel, 66 specimens from 29 patients were analyzed, identifying 16 mutations and 4 fusions, including two novel fusions (FGFR2-SHTN1 and RFTN1-BRAF). Genetic alterations differed between primary and metastatic sites in nine patients (31%), predominantly in additional oncogenic alterations (89%). More genetic alterations were found at the primary site in three patients and metastatic sites in four. Distinct mutations were found in two patients. A longer time interval between specimen acquisitions was significantly associated with genetic discrepancies (p = 0.032). CONCLUSION Patterns of genetic discrepancies between primary and metastatic TC vary, offering valuable insights for clinical practice.
Collapse
Affiliation(s)
- Yen-Bo Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Hsiang-Wei Hu
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - An-Ko Chung
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jin-Ying Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Chen Wu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - I-Hsuan Chiu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - I Chu
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jih-Hsiang Lee
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, Hsin-Chu Branch, Hsinchu, Taiwan
| | - Feng-Jung Nien
- Department of Internal Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Kuen-Yuan Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Hsun Wu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Nan Chen
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Wei Wang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Hung Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Fang Cheng
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Yih Chiu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuenn-Wen Kuo
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Hui Hsih
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Yun-Lin Branch, Douliu City, Taiwan
| | - Chih-Yuan Wang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wei-Shiung Yang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shyang-Rong Shih
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
3
|
Li S, Hu G, Chen Y, Sang Y, Tang Q, Liu R. TERT upstream promoter methylation regulates TERT expression and acts as a therapeutic target in TERT promoter mutation-negative thyroid cancer. Cancer Cell Int 2024; 24:271. [PMID: 39097722 PMCID: PMC11297792 DOI: 10.1186/s12935-024-03459-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND DNA hypermethylation and hotspot mutations were frequently observed in the upstream and core promoter of telomerase reverse transcriptase (TERT), respectively, and they were associated with increased TERT expression and adverse clinical outcomes in thyroid cancer. In TERT promoter mutant cancer cells, the hypomethylated TERT mutant allele was active and the hypermethylated TERT wild-type allele was silenced. However, whether and how the upstream promoter methylation regulates TERT expression in TERT mutation-negative cells were largely unknown. METHODS DNA demethylating agents 5-azacytidine and decitabine and a genomic locus-specific demethylation system based on dCas9-TET1 were used to assess the effects of TERT upstream promoter methylation on TERT expression, cell growth and apoptosis of thyroid cancer cells. Regulatory proteins binding to TERT promoter were identified by CRISPR affinity purification in situ of regulatory elements (CAPTURE) combined with mass spectrometry. The enrichments of selected regulatory proteins and histone modifications were evaluated by chromatin immunoprecipitation. RESULTS The level of DNA methylation at TERT upstream promoter and expression of TERT were significantly decreased after treatment with 5-azacytidine or decitabine in TERT promoter wild-type thyroid cancer cells. Genomic locus-specific demethylation of TERT upstream promoter induced TERT downregulation, along with cell apoptosis and growth inhibition. Consistently, demethylating agents sharply inhibited the growth of thyroid cancer cells harboring hypermethylated TERT but had little effect on cells with TERT hypomethylation. Moreover, we identified that the chromatin remodeling protein CHD4 binds to methylated TERT upstream promoter and promotes its transcription by suppressing the enrichment of H3K9me3 and H3K27me3 at TERT promoter. CONCLUSIONS This study uncovered the mechanism of promoter methylation mediated TERT activation in TERT promoter mutation-negative thyroid cancer cells and indicated TERT upstream promoter methylation as a therapeutic target for thyroid cancer.
Collapse
Affiliation(s)
- Shiyong Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510080, China
| | - Guanghui Hu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510080, China
| | - Yulu Chen
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510080, China
| | - Ye Sang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510080, China
| | - Qin Tang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510080, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
4
|
Zheng W, Pu M, Li X, Du Z, Jin S, Li X, Zhou J, Zhang Y. Deep learning model accurately classifies metastatic tumors from primary tumors based on mutational signatures. Sci Rep 2023; 13:8752. [PMID: 37253775 PMCID: PMC10229594 DOI: 10.1038/s41598-023-35842-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 05/24/2023] [Indexed: 06/01/2023] Open
Abstract
Metastatic propagation is the leading cause of death for most cancers. Prediction and elucidation of metastatic process is crucial for the treatment of cancer. Even though somatic mutations have been linked to tumorigenesis and metastasis, it is less explored whether metastatic events can be identified through genomic mutational signatures, which are concise descriptions of the mutational processes. Here, we developed MetaWise, a Deep Neural Network (DNN) model, by applying mutational signatures as input features calculated from Whole-Exome Sequencing (WES) data of TCGA and other metastatic cohorts. This model can accurately classify metastatic tumors from primary tumors and outperform traditional machine learning (ML) models and a deep learning (DL) model, DiaDeL. Signatures of non-coding mutations also have a major impact on the model's performance. SHapley Additive exPlanations (SHAP) and Local Surrogate (LIME) analyses identify several mutational signatures which are directly correlated to metastatic spread in cancers, including APOBEC-mutagenesis, UV-induced signatures, and DNA damage response deficiency signatures.
Collapse
Affiliation(s)
- Weisheng Zheng
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
| | - Mengchen Pu
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
| | - Xiaorong Li
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
- Minzu University of China, Beijing, China
| | - Zhaolan Du
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
- Beijing University of Technology, Beijing, China
| | - Sutong Jin
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
- Harbin Institute of Technology, Weihai, Shandong, China
| | - Xingshuai Li
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
| | - Jielong Zhou
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
| | - Yingsheng Zhang
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China.
| |
Collapse
|
5
|
Wang J, Zhong F, Li J, Yue H, Li W, Lu X. The epigenetic factor CHD4 contributes to metastasis by regulating the EZH2/β-catenin axis and acts as a therapeutic target in ovarian cancer. J Transl Med 2023; 21:38. [PMID: 36681835 PMCID: PMC9862813 DOI: 10.1186/s12967-022-03854-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/26/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The overall survival rate of patients with advanced ovarian cancer (OC) has remained static for several decades. Advanced ovarian cancer is known for its poor prognosis due to extensive metastasis. Epigenetic alterations contribute to tumour progression and therefore are of interest for potential therapeutic strategies. METHODS Following our previous study, we identified that CHD4, a chromatin remodelling factor, plays a strong role in ovarian cancer cell metastasis. We investigated the clinical significance of CHD4 through TCGA and GEO database analyses and explored the effect of CHD4 expression modulation and romidepsin treatment on the biological behaviour of ovarian cancer through CCK-8 and transwell assays. Bioluminescence imaging of tumours in xenografted mice was applied to determine the therapeutic effect of romidepsin. GSEA and western blotting were used to screen the regulatory mechanism of CHD4. RESULTS In ovarian cancer patient specimens, high CHD4 expression was associated with a poor prognosis. Loss of function of CHD4 in ovarian cancer cells induced suppression of migration and invasion. Mechanistically, CHD4 knockdown suppressed the expression of EZH2 and the nuclear accumulation of β-catenin. CHD4 also suppressed the metastasis of ovarian cancer cells and prevented disease progression in a mouse model. To inhibit the functions of CHD4 that are mediated by histone deacetylase, we evaluated the effect of the HDAC1/2 selective inhibitor romidepsin. Our findings indicated that treatment with romidepsin suppressed the progression of metastases in vitro and in vivo. CONCLUSIONS Collectively, our results uncovered an oncogenic function of CHD4 in ovarian cancer and provide a rationale for clinical trials of romidepsin in ovarian cancer patients.
Collapse
Affiliation(s)
- Jieyu Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200090, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, 200090, China
| | - Fangfang Zhong
- Department of Pathology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200090, China
| | - Jun Li
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200090, China
| | - Huiran Yue
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200090, China
| | - Wenzhi Li
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200090, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, 200090, China
| | - Xin Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200090, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, 200090, China.
| |
Collapse
|
6
|
Thyroglobulin expression, Ki-67 index, and lymph node ratio in the prognostic assessment of papillary thyroid cancer. Sci Rep 2023; 13:1070. [PMID: 36658256 PMCID: PMC9852547 DOI: 10.1038/s41598-023-27684-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
The clinical significance of thyroglobulin (Tg) expression in papillary thyroid cancer (PTC) has not been systematically explored in relation to the Ki-67 index, lymph node ratio (LNR), or other conventional prognostic predictors. In this retrospective study of 327 patients with PTC, we investigated the immunohistochemical expression of Tg in both primary tumors and their matching lymph node metastases in relation to the Ki-67 index, LNR, and clinical data. Tumoral Tg immunoreactivity was inversely correlated to the Ki-67 index and tumor recurrence. The Ki-67 index was higher in lymph node metastases (mean 4%) than in the primary tumors (mean 3%). Reduced Tg expression, estimated as 0-25% Tg positive tumor cells, was more common in lymph node metastases compared to primary tumors. In addition to advanced metastatic burden (defined as N1b stage and LNR ≥ 21%), low Tg expression (0-25% positive tumor cells) in lymph node metastases had a significant prognostic impact with shorter recurrence-free survival. These findings support the potential value of histopathological assessment of Tg expression and Ki-67 index in lymph node metastases as complementary predictors to anticipate the prognosis of PTC patients better.
Collapse
|
7
|
Alamri AM, Alkhilaiwi FA, Khan NU, Tasleem M. In silico Screening and Validation of Achyranthes aspera as a Potential Inhibitor of BRAF and NRAS in Controlling Thyroid Cancer. Anticancer Agents Med Chem 2023; 23:2111-2126. [PMID: 37287303 DOI: 10.2174/1871520623666230607125258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Thyroid carcinoma (THCA) is one of the most prevalent endocrine tumors, accounting for 3.4% of all cancers diagnosed annually. Single Nucleotide Polymorphisms (SNPs) are the most prevalent genetic variation associated with thyroid cancer. Understanding thyroid cancer genetics will enhance diagnosis, prognosis, and treatment. METHODS This TCGA-based study analyzes thyroid cancer-associated highly mutated genes through highly robust in silico techniques. Pathway, gene expression, and survival studies were performed on the top 10 highly mutated genes (BRAF, NRAS, TG, TTN, HRAS, MUC16, ZFHX3, CSMD2, EIFIAX, SPTA1). Novel natural compounds from Achyranthes aspera Linn were discovered to target two highly mutated genes. The natural compounds and synthetic drugs used to treat thyroid cancer were subjected to comparative molecular docking against BRAF and NRAS targets. The ADME characteristics of Achyranthes aspera Linn compounds were also investigated. RESULTS The gene expression analysis revealed that the expression of ZFHX3, MCU16, EIF1AX, HRAS, and NRAS was up-regulated in tumor cells while BRAF, TTN, TG, CSMD2, and SPTA1 were down-regulated in tumor cells. In addition, the protein-protein interaction network demonstrated that HRAS, BRAF, NRAS, SPTA1, and TG proteins have strong interactions with each other as compared to other genes. The ADMET analysis shows that seven compounds have druglike properties. These compounds were further studied for molecular docking studies. The compounds MPHY012847, IMPHY005295, and IMPHY000939 show higher binding affinity with BRAF than pimasertib. In addition, IMPHY000939, IMPHY000303, IMPHY012847, and IMPHY005295 showed a better binding affinity with NRAS than Guanosine Triphosphate. CONCLUSION The outcomes of docking experiments conducted on BRAF and NRAS provide insight into natural compounds with pharmacological characteristics. These findings indicate that natural compounds derived from plants as a more promising cancer treatment option. Thus, the results of docking investigations conducted on BRAF and NRAS substantiate the conclusions that the molecule possesses the most suited drug-like qualities. Compared to other compounds, natural compounds are superior, and they are also druggable. This demonstrates that natural plant compounds can be an excellent source of potential anti-cancer agents. The preclinical research will pave the road for a possible anti-cancer agent.
Collapse
Affiliation(s)
- Ahmad M Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
- Cancer Research Unit, King Khalid University, Abha, 61413, Saudi Arabia
| | - Faris A Alkhilaiwi
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Najeeb U Khan
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, 25130, Pakistan
| | - Munazzah Tasleem
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| |
Collapse
|
8
|
Schiantarelli J, Pappa T, Conway J, Crowdis J, Reardon B, Dietlein F, Huang J, Stanizzi D, Carey E, Bosma-Moody A, Imamovic A, Han S, Camp S, Kofman E, Shannon E, Barletta JA, He MX, Liu D, Park J, Lorch JH, Van Allen EM. Mutational Footprint of Platinum Chemotherapy in a Secondary Thyroid Cancer. JCO Precis Oncol 2022; 6:e2200183. [PMID: 36075011 PMCID: PMC9489159 DOI: 10.1200/po.22.00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Julia Schiantarelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Theodora Pappa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Jake Conway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Jett Crowdis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Brendan Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Felix Dietlein
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | | | - Darren Stanizzi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Evan Carey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Alice Bosma-Moody
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Alma Imamovic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Seunghun Han
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Biological and Biomedical Sciences, Harvard Medical School, Boston, MA
| | - Sabrina Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Eric Kofman
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Justine A. Barletta
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
- Harvard Graduate Program in Biophysics, Boston, MA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Jochen H. Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
9
|
Siraj S, Masoodi T, Siraj AK, Azam S, Qadri Z, Parvathareddy SK, Bu R, Siddiqui KS, Al-Sobhi SS, AlDawish M, Al-Kuraya KS. APOBEC SBS13 Mutational Signature-A Novel Predictor of Radioactive Iodine Refractory Papillary Thyroid Carcinoma. Cancers (Basel) 2022; 14:1584. [PMID: 35326735 PMCID: PMC8946015 DOI: 10.3390/cancers14061584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/03/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Standard surgery followed by radioactive iodine (131I, RAI) therapy are not curative for 5−20% of papillary thyroid carcinoma (PTC) patients with RAI refractory disease. Early predictors indicating therapeutic response to RAI therapy in PTC are yet to be elucidated. Whole-exome sequencing was performed (at median depth 198x) on 66 RAI-refractory and 92 RAI-avid PTCs with patient-matched germline. RAI-refractory tumors were significantly associated with distinct aggressive clinicopathological features, including positive surgical margins (p = 0.016) and the presence of lymph node metastases at primary diagnosis (p = 0.012); higher nonsilent tumor mutation burden (p = 0.011); TERT promoter (TERTp) mutation (p < 0.0001); and the enrichment of the APOBEC-related single-base substitution (SBS) COSMIC mutational signatures 2 (p = 0.030) and 13 (p < 0.001). Notably, SBS13 (odds ratio [OR] 30.4, 95% confidence intervals [CI] 1.43−647.22) and TERTp mutation (OR 41.3, 95% CI 4.35−391.60) were revealed to be independent predictors of RAI refractoriness in PTC (p = 0.029 and 0.001, respectively). Although SBS13 and TERTp mutations alone highly predicted RAI refractoriness, when combined, they significantly increased the likelihood of predicting RAI refractoriness in PTC. This study highlights the APOBEC SBS13 mutational signature as a novel independent predictor of RAI refractoriness in a distinct subgroup of PTC.
Collapse
Affiliation(s)
- Sarah Siraj
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| | - Tariq Masoodi
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| | - Abdul K. Siraj
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| | - Saud Azam
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| | - Zeeshan Qadri
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| | - Sandeep K. Parvathareddy
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| | - Rong Bu
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| | - Khawar S. Siddiqui
- Department of Pediatric Hematology-Oncology, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia;
| | - Saif S. Al-Sobhi
- Department of Surgery, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia;
| | - Mohammed AlDawish
- Department of Endocrinology and Diabetes, Prince Sultan Military Medical City, P.O. Box 261370, Riyadh 11342, Saudi Arabia;
| | - Khawla S. Al-Kuraya
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.S.); (T.M.); (A.K.S.); (S.A.); (Z.Q.); (S.K.P.); (R.B.)
| |
Collapse
|
10
|
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
|
11
|
Oskouie AA, Ahmadi MS, Taherkhani A. Identification of Prognostic Biomarkers in Papillary Thyroid Cancer and Developing Non-Invasive Diagnostic Models Through Integrated Bioinformatics Analysis. Microrna 2022; 11:73-87. [PMID: 35068400 DOI: 10.2174/2211536611666220124115445] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/21/2021] [Accepted: 12/31/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Papillary thyroid cancer (PTC) is the most frequent subtype of thyroid carcinoma, mainly detected in patients with benign thyroid nodules (BTN). Due to the invasiveness of accurate diagnostic tests, there is a need to discover applicable biomarkers for PTC. So, in this study, we aimed to identify the genes associated with prognosis in PTC. Besides, we performed a machine learning tool to develop a non-invasive diagnostic approach for PTC. METHODS For the study purposes, the miRNA dataset GSE130512 was downloaded from the GEO database and then analyzed to identify the common differentially expressed miRNAs in patients with non-metastatic PTC (nm-PTC)/metastatic PTC (m-PTC) compared with BTNs. The SVM was also applied to differentiate patients with PTC from those patients with BTN using the common DEMs. A protein-protein interaction network was also constructed based on the targets of the common DEMs. Next, functional analysis was performed, the hub genes were determined, and survival analysis was then executed. RESULTS A total of three common miRNAs were found to be differentially expressed among patients with nm-PTC/m-PTC compared with BTNs. In addition, it was established that the autophagosome maturation, ciliary basal body-plasma membrane docking, antigen processing as ubiquitination & proteasome degradation, and class I MHC mediated antigen processing & presentation are associated with the pathogenesis of PTC. Furthermore, it was illustrated that RPS6KB1, CCNT1, SP1, and CHD4 might serve as new potential biomarkers for PTC prognosis. CONCLUSION RPS6KB1, CCNT1, SP1, and CHD4 may be considered new potential biomarkers used for prognostic aims in PTC. However, performing validation tests is inevitable in the future.
Collapse
Affiliation(s)
- Afsaneh Arefi Oskouie
- Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Saeed Ahmadi
- Department of Otorhinolaryngology, Besat Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
12
|
Kim M, Kwon CH, Jang MH, Kim JM, Kim EH, Jeon YK, Kim SS, Choi KU, Kim IJ, Park M, Kim BH. Whole-Exome Sequencing in Papillary Microcarcinoma: Potential Early Biomarkers of Lateral Lymph Node Metastasis. Endocrinol Metab (Seoul) 2021; 36:1086-1094. [PMID: 34731936 PMCID: PMC8566127 DOI: 10.3803/enm.2021.1132] [Citation(s) in RCA: 4] [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: 06/02/2021] [Revised: 08/09/2021] [Accepted: 09/01/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Early identification of patients with high-risk papillary thyroid microcarcinoma (PTMC) that is likely to progress has become a critical challenge. We aimed to identify somatic mutations associated with lateral neck lymph node (LN) metastasis (N1b) in patients with PTMC. METHODS Whole-exome sequencing (WES) of 14 PTMCs with no LN metastasis (N0) and 13 N1b PTMCs was performed using primary tumors and matched normal thyroid tissues. RESULTS The mutational burden was comparable in N0 and N1b tumors, as the median number of mutations was 23 (range, 12 to 46) in N0 and 24 (range, 12 to 50) in N1b PTMC (P=0.918). The most frequent mutations were detected in PGS1, SLC4A8, DAAM2, and HELZ in N1b PTMCs alone, and the K158Q mutation in PGS1 (four patients, Fisher's exact test P=0.041) was significantly enriched in N1b PTMCs. Based on pathway analysis, somatic mutations belonging to the receptor tyrosine kinase-RAS and NOTCH pathways were most frequently affected in N1b PTMCs. We identified four mutations that are predicted to be pathogenic in four genes based on Clinvar and Combined Annotation-Dependent Depletion score: BRAF, USH2A, CFTR, and PHIP. A missense mutation in CFTR and a nonsense mutation in PHIP were detected in N1b PTMCs only, although in one case each. BRAF mutation was detected in both N0 and N1b PTMCs. CONCLUSION This first comprehensive WES analysis of the mutational landscape of N0 and N1b PTMCs identified pathogenic genes that affect biological functions associated with the aggressive phenotype of PTMC.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Chae Hwa Kwon
- Biomedical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Min Hee Jang
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Jeong Mi Kim
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Eun Heui Kim
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Yun Kyung Jeon
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Sang Soo Kim
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Kyung-Un Choi
- Department of Pathology, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - In Joo Kim
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Meeyoung Park
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Bo Hyun Kim
- Department of Internal Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| |
Collapse
|
13
|
Hescheler DA, Riemann B, Hartmann MJM, Michel M, Faust M, Bruns CJ, Alakus H, Chiapponi C. Targeted Therapy of Papillary Thyroid Cancer: A Comprehensive Genomic Analysis. Front Endocrinol (Lausanne) 2021; 12:748941. [PMID: 34630336 PMCID: PMC8498581 DOI: 10.3389/fendo.2021.748941] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Background A limited number of targeted therapy options exist for papillary thyroid cancer (PTC) to date. Based on genetic alterations reported by the "The Cancer Genome Atlas (TCGA)", we explored whether PTC shows alterations that may be targetable by drugs approved by the FDA for other solid cancers. Methods Databases of the National Cancer Institute and MyCancerGenome were screened to identify FDA-approved drugs for targeted therapy. Target genes were identified using Drugbank. Genetic alterations were classified into conferring drug sensitivity or resistance using MyCancerGenome, CiViC, TARGET, and OncoKB. Genomic data for PTC were extracted from TCGA and mined for alterations predicting drug response. Results A total of 129 FDA-approved drugs with 128 targetable genes were identified. One hundred ninety-six (70%) of 282 classic, 21 (25%) of 84 follicular, and all 30 tall-cell variant PTCs harbored druggable alterations: 259 occurred in 29, 39 in 19, and 31 in 2 targetable genes, respectively. The BRAF V600 mutation was seen in 68% of classic, 16% of follicular variant, and 93% of tall-cell variant PTCs. The RET gene fusion was seen in 8% of classic PTCs, NTRK1 and 3 gene fusions in 3%, and other alterations in <2% of classic variant PTCs. Ninety-nine of 128 (77%) FDA-approved targetable genes did not show any genetic alteration in PTC. Beside selective and non-selective BRAF-inhibitors, no other FDA-approved drug showed any frequent predicted drug sensitivity (<10%). Conclusion Treatment strategies need to focus on resistance mechanisms to BRAF inhibition and on genetic alteration-independent alternatives rather than on current targeted drugs.
Collapse
Affiliation(s)
- Daniel A. Hescheler
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Burkhard Riemann
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Milan J. M. Hartmann
- Department of General, Visceral, Tumor and Transplant Surgery, University Hospital Cologne, Cologne, Germany
| | - Maximilian Michel
- Institute of Zoology, University of Cologne Germany, Cologne, Germany
| | - Michael Faust
- Policlinic for Prevention, Diabetes and Endocrinology, University of Cologne, Cologne, Germany
| | - Christiane J. Bruns
- Department of General, Visceral, Tumor and Transplant Surgery, University Hospital Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral, Tumor and Transplant Surgery, University Hospital Cologne, Cologne, Germany
| | - Costanza Chiapponi
- Department of General, Visceral, Tumor and Transplant Surgery, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
14
|
Wang W, Shen C, Zhao Y, Sun B, Bai N, Li X. Identification and validation of potential novel biomarkers to predict distant metastasis in differentiated thyroid cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1053. [PMID: 34422965 PMCID: PMC8339873 DOI: 10.21037/atm-21-383] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/16/2021] [Indexed: 12/18/2022]
Abstract
Background Distant metastasis (DM) is not common in differentiated thyroid cancer (DTC). However, it is associated with a significantly poor prognosis. Early detection of high-risk DTC patients is difficult, and the molecular mechanism is still unclear. Therefore, the present study aims to establish a novel predictive model based on clinicopathological parameters and DM-related gene signatures to provide guidelines for clinicians in decision making. Methods Weighted gene co-expression network analysis (WGCNA) was performed to discover co-expressed gene modules and hub genes associated with DM. Univariate and multivariate analyses were carried out to identify independent clinicopathological risk factors based on The Cancer Genome Atlas (TCGA) database. An integrated nomogram prediction model was established. Finally, real hub genes were validated using the GSE60542 database and various thyroid cell lines. Results The midnightblue module was most significantly positively correlated with DM (R=0.56, P=9e-06) by as per WGCNA. DLX5 (AUC: 0.769), COX6B2 (AUC: 0.764), and LYPD1 (AUC: 0.760) were determined to be the real hub genes that play a crucial role in predicting DM. Meanwhile, univariate and multivariate analyses demonstrated that T-stage (OR, 15.03; 95% CI, 1.75-319.40; and P=0.024), histologic subtype (OR, 0.17; 95% CI, 0.03-0.92; and P=0.042) were the independent predictors of DM. Subsequently, a nomogram model was constructed based on gene signatures and independent clinical risk factors exhibited good performance. Additionally, the mRNA expressions of real hub genes in the GSE60542 dataset were consistent with TCGA. Conclusions The present study has provided a reliable model to predict DM in patients with DTC. This model is likely to serve as an individual risk assessment tool in therapeutic decision-making.
Collapse
Affiliation(s)
- Wenlong Wang
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Shen
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha, China
| | - Yunzhe Zhao
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha, China
| | - Botao Sun
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Bai
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Thyroid Surgery Department, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Pre-Therapeutic Measurements of Iodine Avidity in Papillary and Poorly Differentiated Thyroid Cancer Reveal Associations with Thyroglobulin Expression, Histological Variants and Ki-67 Index. Cancers (Basel) 2021; 13:cancers13143627. [PMID: 34298840 PMCID: PMC8307105 DOI: 10.3390/cancers13143627] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/29/2022] Open
Abstract
Papillary thyroid cancer (PTC) and poorly differentiated thyroid cancer (PDTC) are treated with radioiodine to reduce recurrence and to treat the spread of disease. Adequate iodine accumulation in cancer tissue, iodine avidity, is important for treatment effect. This study investigated which clinical and histological tumour characteristics correlate with avidity. To quantify avidity in cancer tissue, tracer amounts of iodine-131 were given to 45 patients with cytologically confirmed thyroid cancer. At pathology grossing, representative samples of tumour and lymph nodes were taken and subjected to radioactivity quantification ex vivo to determine avidity. Afterwards, samples underwent extended pathology work-up and analysis. We found that tumoural Tg expression and Ki-67 index were correlated with avidity, whereas tumour size and pT stage were not. The histological variant of thyroid cancer was also correlated with iodine avidity. Variants associated with worse clinical prognoses displayed lower avidity than variants with better prognoses. This work provides new information on which tumours have low iodine avidity. Lower avidity in aggressive histological PTC variants may explain their overall poorer prognoses. Our findings also suggest that radioiodine dosage could be adapted to Tg expression, Ki-67 index or histological variant instead of pT stage, potentially improving the efficacy of radioiodine therapy.
Collapse
|
16
|
Hu MI, Waguespack SG, Dosiou C, Ladenson PW, Livhits MJ, Wirth LJ, Sadow PM, Krane JF, Stack BC, Zafereo ME, Ali SZ, Weitzman SP, Hao Y, Babiarz JE, Kennedy GC, Kloos RT. Afirma Genomic Sequencing Classifier and Xpression Atlas Molecular Findings in Consecutive Bethesda III-VI Thyroid Nodules. J Clin Endocrinol Metab 2021; 106:2198-2207. [PMID: 34009369 PMCID: PMC8277199 DOI: 10.1210/clinem/dgab304] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Indexed: 12/13/2022]
Abstract
CONTEXT Broad genomic analyses among thyroid histologies have been described from relatively small cohorts. OBJECTIVE Investigate the molecular findings across a large, real-world cohort of thyroid fine-needle aspiration (FNA) samples. DESIGN Retrospective analysis of RNA sequencing data files. SETTING Clinical Laboratory Improvement Amendments laboratory performing Afirma Genomic Sequencing Classifier (GSC) and Xpression Atlas (XA) testing. PARTICIPANTS A total of 50 644 consecutive Bethesda III-VI nodules. INTERVENTION None. MAIN OUTCOME MEASURES Molecular test results. RESULTS Of 48 952 Bethesda III/IV FNAs studied, 66% were benign by Afirma GSC. The prevalence of BRAF V600E was 2% among all Bethesda III/IV FNAs and 76% among Bethesda VI FNAs. Fusions involving NTRK, RET, BRAF, and ALK were most prevalent in Bethesda V (10%), and 130 different gene partners were identified. Among small consecutive Bethesda III/IV sample cohorts with one of these fusions and available surgical pathology excision data, the positive predictive value of an NTRK or RET fusion for carcinoma or noninvasive follicular thyroid neoplasm with papillary-like nuclear features was >95%, whereas for BRAF and ALK fusions it was 81% and 67%, respectively. At least 1 genomic alteration was identified by the expanded Afirma XA panel in 70% of medullary thyroid carcinoma classifier-positive FNAs, 44% of Bethesda III or IV Afirma GSC suspicious FNAs, 64% of Bethesda V FNAs, and 87% of Bethesda VI FNAs. CONCLUSIONS This large study demonstrates that almost one-half of Bethesda III/IV Afirma GSC suspicious and most Bethesda V/VI nodules had at least 1 genomic variant or fusion identified, which may optimize personalized treatment decisions.
Collapse
Affiliation(s)
- Mimi I Hu
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: Mimi I. Hu, MD, Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1461, Houston, TX 77030, USA.
| | - Steven G Waguespack
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chrysoula Dosiou
- Department of Medicine, Division of Endocrinology, Gerontology, & Metabolism, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul W Ladenson
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Masha J Livhits
- Department of Surgery, Section of Endocrine Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Lori J Wirth
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Peter M Sadow
- Department of Pathology, Head and Neck Pathology Subspecialty, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey F Krane
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Brendan C Stack
- Department of Otolaryngology-Head and Neck Surgery, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Mark E Zafereo
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Syed Z Ali
- Departments of Pathology and Radiology, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Steven P Weitzman
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yangyang Hao
- Research and Development, Veracyte, South San Francisco, CA 94080, USA
| | - Joshua E Babiarz
- Research and Development, Veracyte, South San Francisco, CA 94080, USA
| | - Giulia C Kennedy
- Departments of Clinical Affairs, Medical Affairs, Research and Development, Veracyte, Inc., South San Francisco, CA 94080, USA
| | - Richard T Kloos
- Department of Medical Affairs, Veracyte, Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
17
|
Luo H, Liao X, Qin Y, Hou Q, Xue Z, Liu Y, Shen F, Wang Y, Jiang Y, Song L, Chen H, Zhang L, Wei T, Dai L, Yang L, Zhang W, Li Z, Xu H, Zhu J, Shu Y. Longitudinal Genomic Evolution of Conventional Papillary Thyroid Cancer With Brain Metastasis. Front Oncol 2021; 11:620924. [PMID: 34249677 PMCID: PMC8260944 DOI: 10.3389/fonc.2021.620924] [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: 10/24/2020] [Accepted: 06/04/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Brain metastasis is extremely rare but predicts dismal prognosis in papillary thyroid cancer (PTC). Dynamic evaluation of stepwise metastatic lesions was barely conducted to identify the longitudinal genomic evolution of brain metastasis in PTC. METHOD Chronologically resected specimen was analyzed by whole exome sequencing, including four metastatic lymph nodes (lyn 1-4) and brain metastasis lesion (BM). Phylogenetic tree was reconstructed to infer the metastatic pattern and the potential functional mutations. RESULTS Contrasting with lyn1, ipsilateral metastatic lesions (lyn2-4 and BM) with shared biallelic mutations of TSC2 indicated different genetic originations from multifocal tumors. Lyn 3/4, particularly lyn4 exhibited high genetic similarity with BM. Besides the similar mutational compositions and signatures, shared functional mutations (CDK4 R24C , TP53R342*) were observed in lyn3/4 and BM. Frequencies of these mutations gradually increase along with the metastasis progression. Consistently, TP53 knockout and CDK4 R24C introduction in PTC cells significantly decreased radioiodine uptake and increased metastatic ability. CONCLUSION Genomic mutations in CDK4 and TP53 during the tumor evolution may contribute to the lymph node and brain metastasis of PTC.
Collapse
Affiliation(s)
- Han Luo
- Department of Thyroid and Parathyroid Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Qin
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Qianqian Hou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhinan Xue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Thyroid and Parathyroid Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Feiyang Shen
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yuelan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Linlin Song
- Department of Thyroid and Parathyroid Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Haining Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyun Zhang
- Department of Thyroid and Parathyroid Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Wei
- Department of Thyroid and Parathyroid Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhihui Li
- Department of Thyroid and Parathyroid Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Heng Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiang Zhu
- Department of Thyroid and Parathyroid Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Muzza M, Pogliaghi G, Persani L, Fugazzola L, Colombo C. Combined Mutational and Clonality Analyses Support the Existence of Intra-Tumor Heterogeneity in Papillary Thyroid Cancer. J Clin Med 2021; 10:jcm10122645. [PMID: 34208446 PMCID: PMC8234519 DOI: 10.3390/jcm10122645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/27/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Despite its potential clinical impact, intra-tumor genetic heterogeneity (ITH) has been scantly investigated in papillary thyroid cancer (PTC). We studied ITH in PTC by combining, for the first time, data derived from the evaluation of the normalized allelic frequencies (NAF) of the mutation/s, using a customized MassARRAY panel, and those obtained by the HUMARA clonality assay. Among tumors with a single mutation, 80% of cases with NAF 50 ± 5% were clonal, consistent with the presence of a single mutated clone, while 20% of cases showed a polyclonal pattern, suggesting the presence of the same mutation in two or more clones. Differently, all cases with NAF < 45% were polyclonal. Among tumors with double mutation, cases with both mutations showing NAF 50 ± 5% were monoclonal, consistent with the presence of a single clone harboring both mutations. On the other hand, all cases with double mutation at NAF < 45% were polyclonal, indicating the presence of two clones with different mutations. Finally, no significant differences in the clinico-pathological characteristics were found between monoclonal and polyclonal tumors. In conclusion, the present study adds insights into the concept of ITH in PTC, which warrants attention because the occurrence of this phenomenon is likely to affect the response to targeted drugs.
Collapse
Affiliation(s)
- Marina Muzza
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (M.M.); (G.P.)
| | - Gabriele Pogliaghi
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (M.M.); (G.P.)
| | - Luca Persani
- Division of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (L.P.); (C.C.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Laura Fugazzola
- Division of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (L.P.); (C.C.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
- Correspondence:
| | - Carla Colombo
- Division of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (L.P.); (C.C.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| |
Collapse
|
19
|
Stenman A, Backman S, Johansson K, Paulsson JO, Stålberg P, Zedenius J, Juhlin CC. Pan-genomic characterization of high-risk pediatric papillary thyroid carcinoma. Endocr Relat Cancer 2021; 28:337-351. [PMID: 33827048 PMCID: PMC8111328 DOI: 10.1530/erc-20-0464] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/06/2021] [Indexed: 12/20/2022]
Abstract
Pediatric papillary thyroid carcinomas (pPTCs) are often indolent tumors with excellent long-term outcome, although subsets of cases are clinically troublesome and recur. Although it is generally thought to exhibit similar molecular aberrancies as their counterpart tumors in adults, the pan-genomic landscape of clinically aggressive pPTCs has not been previously described. In this study, five pairs of primary and synchronously metastatic pPTC from patients with high-risk phenotypes were characterized using parallel whole-genome and -transcriptome sequencing. Primary tumors and their metastatic components displayed an exceedingly low number of coding somatic mutations and gross chromosomal alterations overall, with surprisingly few shared mutational events. Two cases exhibited one established gene fusion event each (SQSTM1-NTRK3 and NCOA4-RET) in both primary and metastatic tissues, and one case each was positive for a BRAF V600E mutation and a germline truncating CHEK2 mutation, respectively. One single case was without apparent driver events and was considered as a genetic orphan. Non-coding mutations in cancer-associated regions were generally not present. By expressional analyses, fusion-driven primary and metastatic pPTC clustered separately from the mutation-driven cases and the sole genetic orphan. We conclude that pPTCs are genetically indolent tumors with exceedingly stable genomes. Several mutations found exclusively in the metastatic samples which may represent novel genetic events that drive the metastatic behavior, and the differences in mutational compositions suggest early clonal divergence between primary tumors and metastases. Moreover, an overrepresentation of mutational and expressional dysregulation of immune regulatory pathways was noted among fusion-positive pPTC metastases, suggesting that these tumors might facilitate spread through immune evasive mechanisms.
Collapse
Affiliation(s)
- Adam Stenman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - Samuel Backman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Klara Johansson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Johan O Paulsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan Zedenius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
- Correspondence should be addressed to C C Juhlin:
| |
Collapse
|
20
|
Intratumoral Heterogeneity in Differentiated Thyroid Tumors: An Intriguing Reappraisal in the Era of Personalized Medicine. J Pers Med 2021; 11:jpm11050333. [PMID: 33922518 PMCID: PMC8146970 DOI: 10.3390/jpm11050333] [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: 03/30/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
Differentiated thyroid tumors (DTTs) are characterized by significant molecular variability in both spatial and temporal intra-tumoral heterogeneity (ITH), that could influence the therapeutic management. ITH phenomenon appears to have a relevant role in tumor growth, aggressive behavior and drug resistance. Accordingly, characteristics and consequences of ITH in DTTs should be better analyzed and understood in order to guide clinical practice, improving survival. Consequently, in the present review, we investigated morphological and molecular ITH of DTTs in benign, borderline neoplasms and in malignant entities, summarizing the most significant data. Molecular testing in DTTs documents a high risk for recurrence of cancer associated with BRAFV600E, RET/PTC 1/3, ALK and NTRK fusions, while the intermediate risk may be related to BRAFK601E, H/K/N RAS and PAX8/PPARγ. In addition, it may be suggested that tumor genotype is associated with peculiar phenotype.
Collapse
|
21
|
Eszlinger M, Khalil M, Gillmor AH, Huang H, Stewardson P, McIntyre JB, Morrissy S, Paschke R. Histology-based molecular profiling improves mutation detection for advanced thyroid cancer. Genes Chromosomes Cancer 2021; 60:531-545. [PMID: 33749950 DOI: 10.1002/gcc.22949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/21/2023] Open
Abstract
Advanced cancers frequently show histologic and molecular intratumoral heterogeneity. Therefore, we comprehensively characterized advanced, metastatic, radioiodine-resistant (RAIR) thyroid carcinomas at the molecular level in the context of histologic heterogeneity with the aim to identify potentially actionable mutations that may guide the use of specific tyrosine kinase inhibitor (TKI) treatment. Whole exome sequencing (WES) was applied to 29 macrodissected tissue samples of histologically heterogeneous and homogeneous areas, lymph node and lung metastases from six clinically and histologically well-characterized metastatic RAIR thyroid cancer patients with structural incomplete response to treatment. WES data were analyzed to identify potential driver mutations in oncogenic pathways, copy number alterations, microsatellite instability, mutant-allele tumor heterogeneity, and the relevance of histologic heterogeneity to molecular profiling. In addition to known driver mutations in BRAF, NRAS, EIF1AX, NCOA4-RET, and TERT, further potentially actionable drivers were identified in AKT1, ATM, E2F1, HTR2A, and MLH3. The analysis of the evolutionary history of the mutations and the reconstruction of the molecular phylogeny of the cancers show a remarkable association between histologic and molecular heterogeneity. A comprehensive molecular analysis of the primary tumor guided by histologic analysis may help to better stratify patients for precision medicine approaches. Given the association between the molecular and the histologic heterogeneity, the selection of tumor samples for molecular analysis should be based on meticulous histologic evaluation of the entire tumor.
Collapse
Affiliation(s)
- Markus Eszlinger
- Departments of Oncology, Pathology and Laboratory Medicine, Biochemistry and Molecular Biology, and Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Institute of Pathology, University Hospital Halle, Halle, Germany
| | - Moosa Khalil
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Aaron Hill Gillmor
- Department of Graduate Sciences and Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Helen Huang
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul Stewardson
- Department of Medical Science and Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - John B McIntyre
- Precision Oncology Hub Laboratory, Alberta Health Services, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Sorana Morrissy
- Department of Biochemistry and Molecular Biology, and Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ralf Paschke
- Departments of Medicine, Oncology, Pathology and Laboratory Medicine, Biochemistry and Molecular Biology, and Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
22
|
Zhou Y, Liu J, Chu L, Dong M, Feng L. Whole-Exome Sequencing Reveals Novel Variations in Patients with Familial Von Hippel-Lindau Syndrome. World Neurosurg 2021; 150:e696-e704. [PMID: 33774214 DOI: 10.1016/j.wneu.2021.03.089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Von Hippel-Lindau (VHL) syndrome is a rare disease that occurs in an autosomal-dominant genetic pattern. Due to the high genetic variability of VHL diseases, current studies have limited clinical value. Moreover, casual genetic variations in patients with VHL syndrome are still unclear. METHODS Here, we performed whole-exome sequencing of 25 individuals to identify reliable disease-related variations. Systemic computational analysis was performed for variant detection, and Sanger sequencing was used to validate detected mutations. RESULTS Most of the known mutations in the VHL gene were observed in the studied population. In addition, a large fragment deletion in VHL exon 2 in the immediate family members of the last family was detected. This had not been reported earlier. Moreover, we identified 3 novel mutation sites in the MAP2K3 gene that may be involved in the occurrence and development of the VHL disease. CONCLUSIONS These results demonstrated that the heterogeneous nature of VHL syndrome and novel mutational signatures may help to improve the diagnostic ability of VHL syndrome.
Collapse
Affiliation(s)
- Yuhao Zhou
- Guizhou Medical University, Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Jian Liu
- Guizhou Medical University, Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, P. R. China.
| | - Liangzhao Chu
- Department of Neurosurgery, Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Minghao Dong
- Department of Neurosurgery, Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Luqian Feng
- Department of Neurosurgery, Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| |
Collapse
|
23
|
Gomes-Lima CJ, Shobab L, Wu D, Ylli D, Bikas A, McCoy M, Feldman R, Lee W, Rao SN, Jensen K, Vasko V, Castro LC, Jonklaas J, Wartofsky L, Burman KD. Do Molecular Profiles of Primary Versus Metastatic Radioiodine Refractory Differentiated Thyroid Cancer Differ? Front Endocrinol (Lausanne) 2021; 12:623182. [PMID: 33716974 PMCID: PMC7949910 DOI: 10.3389/fendo.2021.623182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Management of metastatic radioiodine refractory differentiated thyroid cancer (DTC) can be a therapeutic challenge. Generally, little is known about the paired molecular profile of the primary tumor and the metastases and whether they harbor the same genetic abnormalities. The present study compared the molecular profile of paired tumor specimens (primary tumor/metastatic sites) from patients with radioiodine refractory DTC in order to gain insight into a possible basis for resistance to radioiodine. Twelve patients with radioiodine refractory metastases were studied; median age at diagnosis of 61 years (range, 25-82). Nine patients had papillary TC (PTC), one had follicular TC (FTC), and two had Hürthle cell TC (HTC). Distant metastases were present in the lungs (n = 10), bones (n = 4), and liver (n = 1). The molecular profiling of paired tumors was performed with a panel of 592 genes for Next Generation Sequencing, RNA-sequencing, and immunohistochemistry. Digital microfluidic PCR was used to investigate TERT promoter mutations. The genetic landscape of all paired sites comprised BRAF, NRAS, HRAS, TP53, ATM, MUTYH, POLE, and NTRK genes, including BRAF and NTRK fusions. BRAF V600E was the most common point mutation in the paired specimens (5/12). TERT promoter mutation C228T was detected in one case. PD-L1 expression at metastatic sites was highly positive (95%) for one patient with HTC. All specimens were stable for microsatellite instability testing, and the tumor mutation burden was low to intermediate. Therefore, the molecular profile of DTC primary and metastatic lesions can show heterogeneity, which may help explain some altered responses to therapeutic intervention.
Collapse
Affiliation(s)
- Cristiane J. Gomes-Lima
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
- University of Brasilia School of Health Sciences, Postgraduate Program, Brasilia, Brazil
| | - Leila Shobab
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Di Wu
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Dorina Ylli
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Athanasios Bikas
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Matthew McCoy
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, United States
| | - Rebecca Feldman
- Caris Life Sciences, Medical Affairs, Phoenix, AZ, United States
| | - Wen Lee
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Sarika N. Rao
- Division of Endocrinology, Mayo Clinic, Jacksonville, FL, United States
| | - Kirk Jensen
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Vasily Vasko
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Luiz Claudio Castro
- Department of Pediatrics, University of Brasilia School of Medicine, Brasilia, Brazil
| | - Jacqueline Jonklaas
- Department of Medicine, Georgetown University, Washington, DC, United States
| | - Leonard Wartofsky
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Kenneth D. Burman
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| |
Collapse
|
24
|
Muzza M. The clonal origin of multifocal papillary thyroid cancer: intrathyroidal spread or independent tumors? Minerva Endocrinol (Torino) 2020; 46:35-44. [PMID: 33045819 DOI: 10.23736/s2724-6507.20.03302-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multifocality is a common finding in papillary thyroid cancer but the molecular pathogenesis, prognosis and management of multifocal papillary thyroid cancer are debated. The clonal origin of multifocal papillary thyroid cancer represents a controversial aspect, as two opposite viewpoints have been proposed: independent origin or intraglandular spread. Different approaches have been used for inferring the clonality of multifocal papillary thyroid cancer, including X-chromosome inactivation, mutational analysis, determination of loss of heterozygosity and, more recently, next-generation sequencing. Next-generation sequencing, able to provide information on genetic heterogeneity and phylogenetic evolution in multifocal tumors, represents the most reliable approach. While most evidences indicated an independent origin of multifocal papillary thyroid cancer, a minority of studies suggested that multifocal papillary thyroid tumors might be monoclonally derived. This discrepancy may reflect technical limitations; nevertheless, studies based on next-generation sequencing indicated that both independent and clonal origins are possible. The co-existence of multiple tumors implies a high degree of genetic heterogeneity, which may influence the best and targeted therapeutic strategy. On the other hand, intrathyroidal dissemination may indicate metastatic potential of the dominant tumor, thereby prompting more aggressive treatments. In conclusion, data available in the literature indicated that multifocal papillary thyroid cancer may derived from both intraglandular spread and independent tumor foci. The understanding of the clonal origin of multifocal papillary thyroid tumors might represent an important issue in patient treatment.
Collapse
Affiliation(s)
- Marina Muzza
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy -
| |
Collapse
|
25
|
Rajan N, Khanal T, Ringel MD. Progression and dormancy in metastatic thyroid cancer: concepts and clinical implications. Endocrine 2020; 70:24-35. [PMID: 32779092 PMCID: PMC7530083 DOI: 10.1007/s12020-020-02453-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023]
Abstract
Distant metastasis classically has been defined as a late-stage event in cancer progression. However, it has become clear that metastases also may occur early in the "lifetime" of a cancer and that they may remain stable at distant sites. This stability of metastatic cancer deposits has been termed "metastatic dormancy" or, as we term it, "metastatic progression dormancy" as the progression either may reflect growth of already existing metastases or new cancer spread. Biologically, dormancy is the presence of nongrowing, static metastatic cells that survive over time. Clinically, dormancy is defined by stability in tumor markers, imaging, and clinical course. Metastatic well-differentiated thyroid cancer offers an excellent tumor type to understand these processes for several reasons: (1) primary therapy often includes removal of the entire gland with ablation of residual normal tissue thereby removing one source for new metastases; (2) the presence of a sensitive biochemical and radiographic monitoring tests enabling monitoring of metastasis throughout the progression process; and (3) its tendency toward prolonged clinical dormancy that can last for years or decades be followed by progression. This latter factor provides opportunities to define therapeutic targets and/or markers of progression. In this review, we will discuss concepts of metastatic progression dormancy and the factors that drive both long-term stability and loss of dormancy with a focus on thyroid cancer.
Collapse
Affiliation(s)
- Neel Rajan
- Division of Endocrinology, Diabetes, and Metabolism, Arthur G. James Comprehensive Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Tilak Khanal
- Division of Endocrinology, Diabetes, and Metabolism, Arthur G. James Comprehensive Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Matthew D Ringel
- Division of Endocrinology, Diabetes, and Metabolism, Arthur G. James Comprehensive Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
26
|
Szpak-Ulczok S, Pfeifer A, Rusinek D, Oczko-Wojciechowska M, Kowalska M, Tyszkiewicz T, Cieslicka M, Handkiewicz-Junak D, Fujarewicz K, Lange D, Chmielik E, Zembala-Nozynska E, Student S, Kotecka-Blicharz A, Kluczewska-Galka A, Jarzab B, Czarniecka A, Jarzab M, Krajewska J. Differences in Gene Expression Profile of Primary Tumors in Metastatic and Non-Metastatic Papillary Thyroid Carcinoma-Do They Exist? Int J Mol Sci 2020; 21:E4629. [PMID: 32610693 PMCID: PMC7369779 DOI: 10.3390/ijms21134629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/22/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Molecular mechanisms of distant metastases (M1) in papillary thyroid cancer (PTC) are poorly understood. We attempted to analyze the gene expression profile in PTC primary tumors to seek the genes associated with M1 status and characterize their molecular function. One hundred and twenty-three patients, including 36 M1 cases, were subjected to transcriptome oligonucleotide microarray analyses: (set A-U133, set B-HG 1.0 ST) at transcript and gene group level (limma, gene set enrichment analysis (GSEA)). An additional independent set of 63 PTCs, including 9 M1 cases, was used to validate results by qPCR. The analysis on dataset A detected eleven transcripts showing significant differences in expression between metastatic and non-metastatic PTC. These genes were validated on microarray dataset B. The differential expression was positively confirmed for only two genes: IGFBP3, (most significant) and ECM1. However, when analyzed on an independent dataset by qPCR, the IGFBP3 gene showed no differences in expression. Gene group analysis showed differences mainly among immune-related transcripts, indicating the potential influence of tumor immune infiltration or signal within the primary tumor. The differences in gene expression profile between metastatic and non-metastatic PTC, if they exist, are subtle and potentially detectable only in large datasets.
Collapse
Affiliation(s)
- Sylwia Szpak-Ulczok
- Nuclear Medicine and Endocrine Oncology Department; Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (S.S.-U.); (D.H.-J.); (A.K.-B.); (A.K.-G.); (B.J.)
| | - Aleksandra Pfeifer
- Department of Genetic and Molecular Diagnostics of Cancer, Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (A.P.); (D.R.); (M.O.-W.); (M.K.); (T.T.); (M.C.)
| | - Dagmara Rusinek
- Department of Genetic and Molecular Diagnostics of Cancer, Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (A.P.); (D.R.); (M.O.-W.); (M.K.); (T.T.); (M.C.)
| | - Malgorzata Oczko-Wojciechowska
- Department of Genetic and Molecular Diagnostics of Cancer, Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (A.P.); (D.R.); (M.O.-W.); (M.K.); (T.T.); (M.C.)
| | - Malgorzata Kowalska
- Department of Genetic and Molecular Diagnostics of Cancer, Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (A.P.); (D.R.); (M.O.-W.); (M.K.); (T.T.); (M.C.)
| | - Tomasz Tyszkiewicz
- Department of Genetic and Molecular Diagnostics of Cancer, Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (A.P.); (D.R.); (M.O.-W.); (M.K.); (T.T.); (M.C.)
| | - Marta Cieslicka
- Department of Genetic and Molecular Diagnostics of Cancer, Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (A.P.); (D.R.); (M.O.-W.); (M.K.); (T.T.); (M.C.)
| | - Daria Handkiewicz-Junak
- Nuclear Medicine and Endocrine Oncology Department; Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (S.S.-U.); (D.H.-J.); (A.K.-B.); (A.K.-G.); (B.J.)
| | - Krzysztof Fujarewicz
- Institute of Automatic Control, Silesian University of Technology, 44-100 Gliwice, Poland; (K.F.); (S.S.)
| | - Dariusz Lange
- Tumor Pathology Department; Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (D.L.); (E.C.); (E.Z.-N.)
| | - Ewa Chmielik
- Tumor Pathology Department; Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (D.L.); (E.C.); (E.Z.-N.)
| | - Ewa Zembala-Nozynska
- Tumor Pathology Department; Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (D.L.); (E.C.); (E.Z.-N.)
| | - Sebastian Student
- Institute of Automatic Control, Silesian University of Technology, 44-100 Gliwice, Poland; (K.F.); (S.S.)
| | - Agnieszka Kotecka-Blicharz
- Nuclear Medicine and Endocrine Oncology Department; Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (S.S.-U.); (D.H.-J.); (A.K.-B.); (A.K.-G.); (B.J.)
| | - Aneta Kluczewska-Galka
- Nuclear Medicine and Endocrine Oncology Department; Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (S.S.-U.); (D.H.-J.); (A.K.-B.); (A.K.-G.); (B.J.)
| | - Barbara Jarzab
- Nuclear Medicine and Endocrine Oncology Department; Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (S.S.-U.); (D.H.-J.); (A.K.-B.); (A.K.-G.); (B.J.)
| | - Agnieszka Czarniecka
- The Oncologic and Reconstructive Surgery Clinic; Maria Sklodowska, Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland;
| | - Michal Jarzab
- Breast Unit; Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland;
| | - Jolanta Krajewska
- Nuclear Medicine and Endocrine Oncology Department; Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-101 Gliwice, Poland; (S.S.-U.); (D.H.-J.); (A.K.-B.); (A.K.-G.); (B.J.)
| |
Collapse
|
27
|
Intratumoral Genetic Heterogeneity in Papillary Thyroid Cancer: Occurrence and Clinical Significance. Cancers (Basel) 2020; 12:cancers12020383. [PMID: 32046148 PMCID: PMC7072350 DOI: 10.3390/cancers12020383] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
Intratumoral heterogeneity (ITH) refers to a subclonal genetic diversity observed within a tumor. ITH is the consequence of genetic instability and accumulation of genetic alterations, two mechanisms involved in the progression from an early tumor stage to a more aggressive cancer. While this process is widely accepted, the ITH of early stage papillary thyroid carcinoma (PTC) is debated. By different genetic analysis, several authors reported the frequent occurrence of PTCs composed of both tumor cells with and without RET/PTC or BRAFV600E genetic alterations. While these data, and the report of discrepancies in the genetic pattern between metastases and the primary tumor, demonstrate the existence of ITH in PTC, its extension and biological significance is debated. The ITH takes on a great significance when involves oncogenes, such as RET rearrangements and BRAFV600E as it calls into question their role of driver genes. ITH is also predicted to play a major clinical role as it could have a significant impact on prognosis and on the response to targeted therapy. In this review, we analyzed several data indicating that ITH is not a marginal event, occurring in PTC at any step of development, and suggesting the existence of unknown genetic or epigenetic alterations that still need to be identified.
Collapse
|