1
|
Hernandez-Prera JC. Molecular Pathology of Thyroid Tumors: Old Problems and New Concepts. Clin Lab Med 2024; 44:305-324. [PMID: 38821646 DOI: 10.1016/j.cll.2023.08.006] [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] [Indexed: 06/02/2024]
Abstract
The molecular signatures of many thyroid tumors have been uncovered. These discoveries have translated into clinical practice and are changing diagnostic and tumor classification paradigms. Here, the findings of recent studies are presented with special emphasis on how molecular insights are impacting the understating of RAS mutant thyroid nodules, Hürthel cell neoplasms, and unusual thyroid tumors, such as hyalinizing trabecular tumor, secretory carcinoma of the thyroid, and sclerosing mucoepidermoid carcinoma with eosinophilia. In addition, the utility of detecting actionable molecular alterations by immunohistochemistry in advanced and aggressive thyroid cancer is also discussed.
Collapse
Affiliation(s)
- Juan C Hernandez-Prera
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA.
| |
Collapse
|
2
|
Marotta V, Cennamo M, La Civita E, Vitale M, Terracciano D. Cell-Free DNA Analysis within the Challenges of Thyroid Cancer Management. Cancers (Basel) 2022; 14:cancers14215370. [PMID: 36358788 PMCID: PMC9654679 DOI: 10.3390/cancers14215370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Simple Summary Liquid biopsy is a minimally invasive method that emerged as a new promising tool for improving diagnosis, risk stratification, follow-up, and treatment of cancer patients. To date, the majority of the research in the area of liquid biopsy has focused on plasma-based cell-free DNA as a potential surrogate for tumor DNA obtained from a tissue biopsy. In the last decades, breakthrough advancements have been performed in the knowledge of thyroid cancer genetics, and the role of molecular characterization in clinical decision-making is continuously rising, from diagnosis completion to the personalization of treatment approach. Hence, it is expectable for cell-free DNA to be applicable in thyroid cancer management. This review aims to investigate the cell-free DNA utility for thyroid cancer patients’ care. Abstract Thyroid cancer is the most frequent endocrine malignancy with an increasing incidence trend during the past forty years and a concomitant rise in cancer-related mortality. The circulating cell-free DNA (cfDNA) analysis is a patient’s friendly and repeatable procedure allowing to obtain surrogate information about the genetics and epigenetics of the tumor. The aim of the present review was to address the suitability of cfDNA testing in different forms of thyroid cancer, and the potential clinical applications, as referred to the clinical weaknesses. Despite being limited by the absence of standardization and by reproducibility and validity issues, cfDNA assessment has great potential for the improvement of thyroid cancer management. cfDNA may support the pre-surgical definition of thyroid nodules by complementing invasive thyroid fine needle aspiration cytology. In addition, it may empower risk stratification and could be used as a biomarker for monitoring the post-surgical disease status, both during active surveillance and in the case of anti-tumor treatment.
Collapse
Affiliation(s)
- Vincenzo Marotta
- UOC Clinica Endocrinologica e Diabetologica, AOU San Giovanni di Dio e Ruggi d’Aragona, 84131 Salerno, Italy
- Correspondence: ; Tel.: +39-333-852-1005
| | - Michele Cennamo
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80138 Naples, Italy
| | - Evelina La Civita
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80138 Naples, Italy
| | - Mario Vitale
- Dipartimento di Medicina, Chirurgia e Odontoiatria, Università di Salerno, 84081 Baronissi, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80138 Naples, Italy
| |
Collapse
|
3
|
Shonka DC, Ho A, Chintakuntlawar AV, Geiger JL, Park JC, Seetharamu N, Jasim S, Abdelhamid Ahmed AH, Bible KC, Brose MS, Cabanillas ME, Dabekaussen K, Davies L, Dias-Santagata D, Fagin JA, Faquin WC, Ghossein RA, Gopal RK, Miyauchi A, Nikiforov YE, Ringel MD, Robinson B, Ryder MM, Sherman EJ, Sadow PM, Shin JJ, Stack BC, Tuttle RM, Wirth LJ, Zafereo ME, Randolph GW. American Head and Neck Society Endocrine Surgery Section and International Thyroid Oncology Group consensus statement on mutational testing in thyroid cancer: Defining advanced thyroid cancer and its targeted treatment. Head Neck 2022; 44:1277-1300. [PMID: 35274388 DOI: 10.1002/hed.27025] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The development of systemic treatment options leveraging the molecular landscape of advanced thyroid cancer is a burgeoning field. This is a multidisciplinary evidence-based statement on the definition of advanced thyroid cancer and its targeted systemic treatment. METHODS An expert panel was assembled, a literature review was conducted, and best practice statements were developed. The modified Delphi method was applied to assess the degree of consensus for the statements developed by the author panel. RESULTS A review of the current understanding of thyroid oncogenesis at a molecular level is presented and characteristics of advanced thyroid cancer are defined. Twenty statements in topics including the multidisciplinary management, molecular evaluation, and targeted systemic treatment of advanced thyroid cancer are provided. CONCLUSIONS With the growth in targeted treatment options for thyroid cancer, a consensus definition of advanced disease and statements regarding the utility of molecular testing and available targeted systemic therapy is warranted.
Collapse
Affiliation(s)
- David C Shonka
- Department of Otolaryngology - Head and Neck Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Alan Ho
- Department of Hematology and Medical Oncology, Solid Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Jessica L Geiger
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Jong C Park
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nagashree Seetharamu
- Division of Hematology-Oncology, Donald and Barbara Zucker School of Medicine at Hofstra University, New Hyde Park, New York, USA
| | - Sina Jasim
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, School of Medicine, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Amr H Abdelhamid Ahmed
- Department of Otolaryngology - Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Keith C Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Marcia S Brose
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Kirsten Dabekaussen
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Louise Davies
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James A Fagin
- Endocrinology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - William C Faquin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ronald A Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Raj K Gopal
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Yuri E Nikiforov
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew D Ringel
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Bruce Robinson
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Mabel M Ryder
- Division of Endocrinology, Diabetes, Metabolism, & Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric J Sherman
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer J Shin
- Department of Otolaryngology - Head and Neck Surgery, Center for Surgery and Public Health, Harvard Medical School, Boston, Massachusetts, USA
| | - Brendan C Stack
- Department of Otolaryngology - Head and Neck Surgery, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - R Michael Tuttle
- Endocrinology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lori J Wirth
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark E Zafereo
- Department of Head and Neck Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Gregory W Randolph
- Department of Otolaryngology - Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Abi-Raad R, Prasad ML, Adeniran AJ, Cai G. Copy number variations identified in thyroid FNA specimens are associated with Hürthle cell cytomorphology. Cancer Cytopathol 2022; 130:415-422. [PMID: 35332982 DOI: 10.1002/cncy.22569] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The fine-needle aspiration (FNA) diagnosis of thyroid Hürthle cell neoplasms (HCNs) remains challenging. This study explored a possible association of copy number variations (CNVs) with Hürthle cell lesions of the thyroid. METHODS Thyroid FNA cases that were diagnosed as follicular lesion of undetermined significance (FLUS) or follicular neoplasm (FN)/HCN for which the ThyroSeq version 3 genomic classifier test was performed were retrieved. RESULTS A total of 324 thyroid FNA cases (228 FLUS cases, 46 HCN cases, and 50 FN cases) were included in the study. FLUS cases were further classified as Hürthle cell type (follicular lesion of undetermined significance-Hürthle cell type [FLUS-HCT]; 20 cases) or non-Hürthle cell type (follicular lesion of undetermined significance-non-Hürthle cell type [FLUS-NHCT]; 208 cases). HCN and FLUS-HCT cases showed a higher prevalence of CNVs (23 of 66 [35%]) in comparison with those classified as FN or FLUS-NHCT (14 of 258 [5%]; P < .001). A total of 105 patients had histopathologic follow-up. Cases with CNVs were more likely to be neoplastic (18 of 26 [69%]) and associated with Hürthle cell changes (14 of 26 [54%]) in comparison with cases without any molecular alterations (neoplastic, 8 of 24 [33%]; Hürthle cell changes, 2 of 24 [8%]; P < .05). In HCN/FLUS-HCT cases with CNVs (n = 14), Hürthle cell changes (13 of 14 [93%]) and neoplasms (9 of 14 [64%]) were more likely to be seen on surgical follow-up in comparison with the 17 cases without CNVs (Hürthle cell changes, 6 of 17 [35%]; neoplastic, 3 of 17 [18%]; P < .05). CONCLUSIONS CNVs identified in thyroid FNA cases are associated with Hürthle cell morphology and are suggestive of a neoplasm with Hürthle cell features in thyroid FNAs classified as FLUS-HCT/HCN. This finding may be helpful in triaging patients who would benefit from surgical management.
Collapse
Affiliation(s)
- Rita Abi-Raad
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Manju L Prasad
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Adebowale J Adeniran
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Guoping Cai
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
5
|
Abstract
The molecular signatures of many thyroid tumors have been uncovered. These discoveries have translated into clinical practice and are changing diagnostic and tumor classification paradigms. Here, the findings of recent studies are presented with special emphasis on how molecular insights are impacting the understating of RAS mutant thyroid nodules, Hürthel cell neoplasms, and unusual thyroid tumors, such as hyalinizing trabecular tumor, secretory carcinoma of the thyroid, and sclerosing mucoepidermoid carcinoma with eosinophilia. In addition, the utility of detecting actionable molecular alterations by immunohistochemistry in advanced and aggressive thyroid cancer is also discussed.
Collapse
Affiliation(s)
- Juan C Hernandez-Prera
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA.
| |
Collapse
|
6
|
Theurer S, Rawitzer J, Ting S, Schmid KW. [Diagnostic principles of thyroid tumors in pathology : Relevant changes due to the current WHO classification]. DER PATHOLOGE 2021; 42:125-139. [PMID: 33415346 DOI: 10.1007/s00292-020-00908-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The current edition of the WHO classification of thyroid tumors (2017) contains a number of very relevant changes with considerable consequences for the diagnostic assessment of thyroid specimens. This applies to both the histomorphological examination of surgical specimens and the preoperative fine needle biopsy (FNB). In addition, molecular pathological examinations are becoming increasingly important in the diagnosis of thyroid tumors. Changes affect practically all areas of thyroid tumor diagnostics. Some of these changes have far-reaching consequences that justify a comprehensive commentary and query of the knowledge acquired in the form of this CME article.
Collapse
Affiliation(s)
- Sarah Theurer
- Institut für Pathologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Deutschland.
| | - Josefine Rawitzer
- Institut für Pathologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Saskia Ting
- Institut für Pathologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Kurt Werner Schmid
- Institut für Pathologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| |
Collapse
|
7
|
Aydemirli MD, Corver W, Beuk R, Roepman P, Solleveld-Westerink N, van Wezel T, Kapiteijn E, Morreau H. Targeted Treatment Options of Recurrent Radioactive Iodine Refractory Hürthle Cell Cancer. Cancers (Basel) 2019; 11:E1185. [PMID: 31443247 PMCID: PMC6721552 DOI: 10.3390/cancers11081185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/13/2019] [Indexed: 01/29/2023] Open
Abstract
Objective: To evaluate the efficacy and treatment rationale of Hürthle cell carcinoma (HCC) following a patient with progressive and metastatic HCC. HCC was recently shown to harbor a distinct genetic make-up and the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kiase (PI3K)/AKT signaling pathways are potential targets for anti-cancer agents in the management of recurrent HCC. The presence or absence of gene variants can give a rationale for targeted therapies that could be made available in the context of drug repurposing trials. Methods: Treatment included everolimus, sorafenib, nintedanib, lenvatinib, and panitumumab. Whole genome sequencing (WGS) of metastatic tumor material obtained before administration of the last drug, was performed. We subsequently evaluated the rationale and efficacy of panitumumab in thyroid cancer and control cell lines after epidermal growth factor (EGF) stimulation and treatment with panitumumab using immunofluorescent Western blot analysis. EGF receptor (EGFR) quantification was performed using flow cytometry. Results: WGS revealed a near-homozygous genome (NHG) and a somatic homozygous TSC1 variant, that was absent in the primary tumor. In the absence of RAS variants, panitumumab showed no real-life efficacy. This might be explained by high constitutive AKT signaling in the two thyroid cancer cell lines with NHG, with panitumumab only being a potent inhibitor of pEGFR in all cancer cell lines tested. Conclusions: In progressive HCC, several treatment options outside or inside clinical trials are available. WGS of metastatic tumors might direct the timing of therapy. Unlike other cancers, the absence of RAS variants seems to provide insufficient justification of single-agent panitumumab administration in HCC cases harboring a near-homozygous genome.
Collapse
Affiliation(s)
- Mehtap Derya Aydemirli
- Department of Medical Oncolosgy, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Willem Corver
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Ruben Beuk
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Paul Roepman
- Hartwig Medical Foundation, 1098 XH Amsterdam, The Netherlands
| | | | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncolosgy, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
8
|
Hao Y, Duh QY, Kloos RT, Babiarz J, Harrell RM, Traweek ST, Kim SY, Fedorowicz G, Walsh PS, Sadow PM, Huang J, Kennedy GC. Identification of Hürthle cell cancers: solving a clinical challenge with genomic sequencing and a trio of machine learning algorithms. BMC SYSTEMS BIOLOGY 2019; 13:27. [PMID: 30952205 PMCID: PMC6450053 DOI: 10.1186/s12918-019-0693-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Identification of Hürthle cell cancers by non-operative fine-needle aspiration biopsy (FNAB) of thyroid nodules is challenging. Resultingly, non-cancerous Hürthle lesions were conventionally distinguished from Hürthle cell cancers by histopathological examination of tissue following surgical resection. Reliance on histopathological evaluation requires patients to undergo surgery to obtain a diagnosis despite most being non-cancerous. It is highly desirable to avoid surgery and to provide accurate classification of benignity versus malignancy from FNAB preoperatively. In our first-generation algorithm, Gene Expression Classifier (GEC), we achieved this goal by using machine learning (ML) on gene expression features. The classifier is sensitive, but not specific due in part to the presence of non-neoplastic benign Hürthle cells in many FNAB. RESULTS We sought to overcome this low-specificity limitation by expanding the feature set for ML using next-generation whole transcriptome RNA sequencing and called the improved algorithm the Genomic Sequencing Classifier (GSC). The Hürthle identification leverages mitochondrial expression and we developed novel feature extraction mechanisms to measure chromosomal and genomic level loss-of-heterozygosity (LOH) for the algorithm. Additionally, we developed a multi-layered system of cascading classifiers to sequentially triage Hürthle cell-containing FNAB, including: 1. presence of Hürthle cells, 2. presence of neoplastic Hürthle cells, and 3. presence of benign Hürthle cells. The final Hürthle cell Index utilizes 1048 nuclear and mitochondrial genes; and Hürthle cell Neoplasm Index leverages LOH features as well as 2041 genes. Both indices are Support Vector Machine (SVM) based. The third classifier, the GSC Benign/Suspicious classifier, utilizes 1115 core genes and is an ensemble classifier incorporating 12 individual models. CONCLUSIONS The accurate algorithmic depiction of this complex biological system among Hürthle subtypes results in a dramatic improvement of classification performance; specificity among Hürthle cell neoplasms increases from 11.8% with the GEC to 58.8% with the GSC, while maintaining the same sensitivity of 89%.
Collapse
Affiliation(s)
- Yangyang Hao
- Department of Research & Development, Veracyte, Inc, 6000 Shoreline Court, Suite 300, South San Francisco, CA 94080 USA
| | - Quan-Yang Duh
- Department of Surgery, Section of Endocrine Surgery, University of California San Francisco, San Francisco, CA USA
| | - Richard T. Kloos
- Department of Medical Affairs, Veracyte, Inc, South San Francisco, USA
| | - Joshua Babiarz
- Department of Research & Development, Veracyte, Inc, 6000 Shoreline Court, Suite 300, South San Francisco, CA 94080 USA
| | - R. Mack Harrell
- The Memorial Center for Integrative Endocrine Surgery, Hollywood, FL USA
- The Memorial Center for Integrative Endocrine Surgery, Weston, FL USA
- The Memorial Center for Integrative Endocrine Surgery, Boca Raton, FL USA
| | | | - Su Yeon Kim
- Department of Research & Development, Veracyte, Inc, 6000 Shoreline Court, Suite 300, South San Francisco, CA 94080 USA
| | - Grazyna Fedorowicz
- Department of Research & Development, Veracyte, Inc, 6000 Shoreline Court, Suite 300, South San Francisco, CA 94080 USA
| | - P. Sean Walsh
- Department of Research & Development, Veracyte, Inc, 6000 Shoreline Court, Suite 300, South San Francisco, CA 94080 USA
| | - Peter M. Sadow
- Department of Pathology, Head and Neck Pathology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Jing Huang
- Department of Research & Development, Veracyte, Inc, 6000 Shoreline Court, Suite 300, South San Francisco, CA 94080 USA
| | - Giulia C. Kennedy
- Department of Research & Development, Veracyte, Inc, 6000 Shoreline Court, Suite 300, South San Francisco, CA 94080 USA
- Department of Medical Affairs, Veracyte, Inc, South San Francisco, USA
| |
Collapse
|
9
|
Xu B, Reznik E, Tuttle RM, Knauf J, Fagin JA, Katabi N, Dogan S, Aleynick N, Seshan V, Middha S, Enepekides D, Casadei GP, Solaroli E, Tallini G, Ghossein R, Ganly I. Outcome and molecular characteristics of non-invasive encapsulated follicular variant of papillary thyroid carcinoma with oncocytic features. Endocrine 2019; 64:97-108. [PMID: 30689169 PMCID: PMC6657696 DOI: 10.1007/s12020-019-01848-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/17/2019] [Indexed: 02/01/2023]
Abstract
PURPOSE In 2016, non-invasive encapsulated follicular variant of papillary thyroid carcinoma (NI-EFVPTC) was renamed as noninvasive thyroid follicular neoplasm with papillary-like nuclear features (NIFTP). However, as the study cohort did not mention tumors with oncocytic features, such lesions are still labeled by some as FVPTC. It is therefore crucial to evaluate the outcome and molecular profile of oncocytic NI-EFVPTC. METHODS A multi-institutional clinico-pathologic review was conducted to select 61 patients having oncocytic NI-EFVPTC. A detailed molecular profile was carried out in 15 patients. RESULTS Oncocytic NI-EFVPTCs predominantly affected women in their 50s. There was no distant metastasis, lymph node metastases, or structural recurrence in the entire cohort. Among patients with ≥5 years of FU, all 33 individuals did not recur with a median FU of 10.2 years. Oncocytic NI-EFVPTC commonly had RAS (33%) mutations, a high frequency of mitochondrial DNA mutations (67%) and multiple chromosomal gains/losses (53%). No fusion genes were detected. CONCLUSIONS Oncocytic NI-EFVPTC, when stringently selected for, lacks metastasis at presentation and follows an extremely indolent clinical course, even when treated conservatively with lobectomy alone without RAI therapy. These tumors share a similar mutational profile as NIFTP, FVPTC, and follicular neoplasm and are predominantly RAS-related. Like Hurthle cell neoplasms, they harbor a high frequency of mitochondrial DNA mutations, which contribute to the oncocytic cytomorphology. However, they lack the widespread chromosomal alterations observed in Hurthle cell carcinoma. Consideration should be given to include oncocytic NI-EFVPTCs as NIFTP in order to avoid overtreatment of these highly indolent tumors.
Collapse
Affiliation(s)
- Bin Xu
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - R Michael Tuttle
- Department of Medicine, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - Jeffrey Knauf
- Department of Medicine, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - James A Fagin
- Department of Medicine, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - Nora Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - Nathaniel Aleynick
- Department of Pathology, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - Venkatraman Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - Sumit Middha
- Department of Pathology, Memorial Sloan Kettering Cancer center, New York, NY, USA
| | - Danny Enepekides
- Department of Otolaryngology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | | - Giovanni Tallini
- Department of Experimental, Diagnostic and Specialty Medicine-Anatomic Pathology, University of Bologna School of Medicine, Bologna, Italy
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer center, New York, NY, USA.
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer center, New York, NY, USA.
| |
Collapse
|
10
|
Gopal RK, Kübler K, Calvo SE, Polak P, Livitz D, Rosebrock D, Sadow PM, Campbell B, Donovan SE, Amin S, Gigliotti BJ, Grabarek Z, Hess JM, Stewart C, Braunstein LZ, Arndt PF, Mordecai S, Shih AR, Chaves F, Zhan T, Lubitz CC, Kim J, Iafrate AJ, Wirth L, Parangi S, Leshchiner I, Daniels GH, Mootha VK, Dias-Santagata D, Getz G, McFadden DG. Widespread Chromosomal Losses and Mitochondrial DNA Alterations as Genetic Drivers in Hürthle Cell Carcinoma. Cancer Cell 2018; 34:242-255.e5. [PMID: 30107175 PMCID: PMC6121811 DOI: 10.1016/j.ccell.2018.06.013] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/30/2018] [Accepted: 06/27/2018] [Indexed: 12/24/2022]
Abstract
Hürthle cell carcinoma of the thyroid (HCC) is a form of thyroid cancer recalcitrant to radioiodine therapy that exhibits an accumulation of mitochondria. We performed whole-exome sequencing on a cohort of primary, recurrent, and metastatic tumors, and identified recurrent mutations in DAXX, TP53, NRAS, NF1, CDKN1A, ARHGAP35, and the TERT promoter. Parallel analysis of mtDNA revealed recurrent homoplasmic mutations in subunits of complex I of the electron transport chain. Analysis of DNA copy-number alterations uncovered widespread loss of chromosomes culminating in near-haploid chromosomal content in a large fraction of HCC, which was maintained during metastatic spread. This work uncovers a distinct molecular origin of HCC compared with other thyroid malignancies.
Collapse
Affiliation(s)
- Raj K Gopal
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Kirsten Kübler
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Calvo
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Paz Polak
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Dimitri Livitz
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Peter M Sadow
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Braidie Campbell
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Samuel E Donovan
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Salma Amin
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Zenon Grabarek
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Julian M Hess
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chip Stewart
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Peter F Arndt
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Scott Mordecai
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Angela R Shih
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Frances Chaves
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tiannan Zhan
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Carrie C Lubitz
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA; Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jiwoong Kim
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Lori Wirth
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Sareh Parangi
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | | | - Gilbert H Daniels
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Thyroid Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Vamsi K Mootha
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Gad Getz
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - David G McFadden
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Thyroid Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Internal Medicine, Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
11
|
Genomic and transcriptomic characterization of the mitochondrial-rich oncocytic phenotype on a thyroid carcinoma background. Mitochondrion 2018; 46:123-133. [PMID: 29631022 DOI: 10.1016/j.mito.2018.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/22/2022]
Abstract
We conducted the first systematic omics study of the oncocytic phenotype in 488 papillary thyroid carcinomas (PTC) from The Cancer Genome Atlas. Oncocytic phenotype is secondary to PTC, being unrelated to several pathologic scores. The nuclear genome had low impact on this phenotype (except in specific copy number variation), which was mostly driven by the significant accumulation of mitochondrial DNA non-synonymous and frameshift mutations at high heteroplasmy levels. Energy and mitochondrial-related pathways were significantly enriched in oncocytic tumors that also displayed increased levels of expression for genes involved in autophagy and fusion of mitochondria. Our in vitro tests confirmed that autophagy is increased and functional while mitophagy is decreased in these tumors.
Collapse
|
12
|
Corver WE, Demmers J, Oosting J, Sahraeian S, Boot A, Ruano D, Wezel TV, Morreau H. ROS-induced near-homozygous genomes in thyroid cancer. Endocr Relat Cancer 2018; 25:83-97. [PMID: 29066502 DOI: 10.1530/erc-17-0288] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/24/2017] [Indexed: 12/17/2022]
Abstract
A near-homozygous genome (NHG) is especially seen in a subset of follicular thyroid cancer of the oncocytic type (FTC-OV). An NHG was also observed in the metabolically relatively quiescent cell lines XTC.UC1, a model for FTC-OV, and in FTC-133, -236 and -238, the latter three derived from one single patient with follicular thyroid cancer. FTC-236 subclones showed subtle whole-chromosome differences indicative of sustained reciprocal mitotic missegregations. Reactive oxygen species (ROS) scavenger experiments reduced the number of chromosomal missegregations in XTC.UC1 and FTC-236, while pCHK2 was downregulated in these cells. Treatment with antimycin A increased ROS indicated by enhanced MitoSOX Red and pCHK2 fluorescence in metaphase cells. In a selected set of oncocytic follicular thyroid tumors, increasing numbers of whole-chromosome losses were observed toward an aggressive phenotype, but with retention of chromosome 7. Together, ROS activates CHK2 and links to the stepwise loss of whole chromosomes during tumor progression in these lesions. We postulate that sequential loss of whole chromosomes is a dominant driver of the oncogenesis of a subset of follicular thyroid tumors.
Collapse
Affiliation(s)
- Willem E Corver
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| | - Joris Demmers
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| | - Jan Oosting
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| | - Shima Sahraeian
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| | - Arnoud Boot
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| | - Dina Ruano
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| | - Tom van Wezel
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| | - Hans Morreau
- Department of Pathology Leiden University Medical CenterLeiden, Netherlands
| |
Collapse
|
13
|
Fluorescence in situ hybridization of thyroid fine-needle aspiration biopsy distinguishes between neoplastic and non-neoplastic Hürthle cell lesions. J Am Soc Cytopathol 2018; 7:195-200. [PMID: 31043276 DOI: 10.1016/j.jasc.2018.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/18/2018] [Accepted: 04/23/2018] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Fine-needle aspiration (FNA) biopsy of Hürthle cell proliferations can be difficult to characterize based purely on morphologic features. Studies have shown Hürthle cell neoplasms often demonstrate gains in chromosomes 5, 7, and 12. This study examined fluorescence in situ hybridization (FISH) performance characteristics in non-neoplastic and neoplastic Hürthle cell proliferations sampled by FNA biopsy in order to assess chromosome patterns. MATERIALS AND METHODS FNA biopsies of Hürthle cell proliferations, including nodular hyperplasia (NH), Hürthle cell adenoma (HCA), and Hürthle cell carcinoma (HCC), that had subsequent surgical excision were selected. FISH was performed on an air-dried, modified Wright-Giemsa-stained, aspirate smear slide from each case using a 3-color panel consisting of 1 subtelomeric and 2 centromeric probes for chromosomes 5, 7, and 12. Chromosomal probe patterns were recorded in up to 50 cells. A positive result was considered when >15% of cells showed a polysomy in 2 or more chromosomes. RESULTS A total of 25 cases were included in the study. All cases of NH were negative, and 7 of 9 (78%) HCAs and 8 of 12 (67%) HCCs were positive. Of the positive cases, 2 of the 7 (29%) HCAs showed >50% of cells with polysomy, and 5 of the 8 (63%) HCCs showed >50% of the cells with polysomy. CONCLUSION Thyroid FNA biopsy can identify Hürthle cell proliferations; risk stratification based on morphology is difficult, however. FISH chromosomal evaluation of thyroid FNA biopsies is useful to distinguish neoplastic from non-neoplastic Hürthle cell proliferation.
Collapse
|
14
|
Acquaviva G, Visani M, Repaci A, Rhoden KJ, de Biase D, Pession A, Giovanni T. Molecular pathology of thyroid tumours of follicular cells: a review of genetic alterations and their clinicopathological relevance. Histopathology 2018; 72:6-31. [PMID: 29239040 DOI: 10.1111/his.13380] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 02/05/2023]
Abstract
Thyroid cancer is the most common endocrine malignancy. Knowledge of the molecular pathology of thyroid tumours originating from follicular cells has greatly advanced in the past several years. Common molecular alterations, such as BRAF p.V600E, RAS point mutations, and fusion oncogenes (RET-PTC being the prototypical example), have been, respectively, associated with conventional papillary carcinoma, follicular-patterned tumours (follicular adenoma, follicular carcinoma, and the follicular variant of papillary carcinoma/non-invasive follicular thyroid neoplasm with papillary-like nuclear features), and with papillary carcinomas from young patients and arising after exposure to ionising radiation, respectively. The remarkable correlation between genotype and phenotype shows how specific, mutually exclusive molecular changes can promote tumour development and initiate a multistep tumorigenic process that is characterised by aberrant activation of mitogen-activated protein kinase and phosphoinositide 3-kinase-PTEN-AKT signalling. Molecular alterations are becoming useful biomarkers for diagnosis and risk stratification, and as potential treatment targets for aggressive forms of thyroid carcinoma. What follows is a review of the principal genetic alterations of thyroid tumours originating from follicular cells and of their clinicopathological relevance.
Collapse
Affiliation(s)
- Giorgia Acquaviva
- Anatomical Pathology, Molecular Diagnostic Unit, University of Bologna School of Medicine, Azienda USL di Bologna, Bologna, Italy
| | - Michela Visani
- Anatomical Pathology, Molecular Diagnostic Unit, University of Bologna School of Medicine, Azienda USL di Bologna, Bologna, Italy
| | - Andrea Repaci
- Endocrinology Unit, University of Bologna School of Medicine, Bologna, Italy
| | - Kerry J Rhoden
- Medical Genetics Unit, University of Bologna School of Medicine, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, Molecular Diagnostic Unit, University of Bologna, Azienda USL di Bologna, Bologna, Italy
| | - Annalisa Pession
- Department of Pharmacy and Biotechnology, Molecular Diagnostic Unit, University of Bologna, Azienda USL di Bologna, Bologna, Italy
| | - Tallini Giovanni
- Anatomical Pathology, Molecular Diagnostic Unit, University of Bologna School of Medicine, Azienda USL di Bologna, Bologna, Italy
| |
Collapse
|
15
|
Giordano TJ. Genomic Hallmarks of Thyroid Neoplasia. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:141-162. [PMID: 29083981 DOI: 10.1146/annurev-pathol-121808-102139] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The genomic landscape of thyroid cancers that are derived from follicular cells has been substantially elucidated through the coordinated application of high-throughput genomic technologies. Here, I review the common genetic alterations across the spectrum of thyroid neoplasia and present the resulting model of thyroid cancer initiation and progression. This model illustrates the striking correlation between tumor differentiation and overall somatic mutational burden, which also likely explains the highly variable clinical behavior and outcome of patients with thyroid cancers. These advances are yielding critical insights into thyroid cancer pathogenesis, which are being leveraged for the development of new diagnostic tools, prognostic and predictive biomarkers, and novel therapeutic approaches.
Collapse
Affiliation(s)
- Thomas J Giordano
- Departments of Pathology and Internal Medicine, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA;
| |
Collapse
|
16
|
Mussazhanova Z, Akazawa Y, Matsuda K, Shichijo K, Miura S, Otsubo R, Oikawa M, Yoshiura KI, Mitsutake N, Rogounovitch T, Saenko V, Kozykenova Z, Zhetpisbaev B, Shabdarbaeva D, Sayakenov N, Amantayev B, Kondo H, Ito M, Nakashima M. Association between p53-binding protein 1 expression and genomic instability in oncocytic follicular adenoma of the thyroid. Endocr J 2016; 63:457-67. [PMID: 26935218 DOI: 10.1507/endocrj.ej15-0629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Oncocytic follicular adenomas (FAs) of the thyroid are neoplasms of follicular cell origin that are predominantly composed of large polygonal cells with eosinophilic and granular cytoplasm. However, the pathological characteristics of these tumors are largely unexplored. Both the initiation and progression of cancer can be caused by an accumulation of genetic mutations that can induce genomic instability. Thus, the aim of this study was to evaluate the extent of genomic instability in oncocytic FA. As the presence of p53-binding protein 1 (53BP1) in nuclear foci has been found to reflect DNA double-strand breaks that are triggered by various stresses, the immunofluorescence expression pattern of 53BP-1 was assessed in oncocytic and conventional FA. The association with the degree of DNA copy number aberration (CNA) was also evaluated using array-based comparative genomic hybridization. Data from this study demonstrated increased 53BP1 expression (i.e., "unstable" expression) in nuclear foci of oncocytic FA and a higher incidence of CNAs compared with conventional FA. There was also a particular focus on the amplification of chromosome 1p36 in oncocytic FA, which includes the locus for Tumor protein 73, a member of the p53 family implicated as a factor in the development of malignancies. Further evaluations revealed that unstable 53BP1 expression had a significant positive correlation with the levels of expression of Tumor protein 73. These data suggest a higher level of genomic instability in oncocytic FA compared with conventional FA, and a possible relationship between oncocytic FA and abnormal amplification of Tumor protein 73.
Collapse
Affiliation(s)
- Zhanna Mussazhanova
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Shawky M, Sakr M. Hurthle Cell Lesion: Controversies, Challenges, and Debates. Indian J Surg 2016; 78:41-8. [PMID: 27186039 PMCID: PMC4848220 DOI: 10.1007/s12262-015-1381-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/20/2015] [Indexed: 02/05/2023] Open
Abstract
Hurthle cell lesion is one of the most questionable clinico-pathological entities in most of its aspects. Literature has used the terms oncocytic, oxyphilic, Hurthle, and Ashkanazy interchangeably; what does each term denote? Who first described these cells? What is the cell of origin? How much Hurthle cells should be present to define the lesion as Hurthle cell one? Is it possible to differentiate hyperplastic from neoplastic and benign from malignant Hurthle cell lesion on a non-histopathologic ground? Does it belong to follicular or to papillary neoplasms or should it be classified separately? Can we anticipate its clinical behavior or predict its outcome? How can we manage? We will try to answer these questions in light of the ongoing relevant arguments with the aim of resolving some uncertainties and suggesting how to solve others.
Collapse
Affiliation(s)
- Michael Shawky
- Department of Head and Neck and Endocrine Surgery, Faculty of Medicine, University of Alexandria, Third Floor-Surgery Building, El Sultan Hussein St, Azarita, Alexandria, Egypt
| | - Mahmoud Sakr
- Department of Head and Neck and Endocrine Surgery, Faculty of Medicine, University of Alexandria, Third Floor-Surgery Building, El Sultan Hussein St, Azarita, Alexandria, Egypt
| |
Collapse
|
18
|
Brauner E, Holmes BJ, Krane JF, Nishino M, Zurakowski D, Hennessey JV, Faquin WC, Parangi S. Performance of the Afirma Gene Expression Classifier in Hürthle Cell Thyroid Nodules Differs from Other Indeterminate Thyroid Nodules. Thyroid 2015; 25:789-96. [PMID: 25962906 DOI: 10.1089/thy.2015.0049] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The recently introduced Afirma gene expression classifier (AGEC) provides binary results (benign or suspicious) to guide management of cytologically indeterminate thyroid nodules. The AGEC is intended to reduce unnecessary surgeries for benign nodules, and management algorithms favor surgery for suspicious results. Limited data are available on the performance of this test for Hürthle cell nodules (HCNs). This study hypothesized that a predominance of Hürthle cells leads to an increased rate of suspicious AGEC results with a potential for overtreatment, despite a relatively low risk of malignancy. METHODS The pathology databases from three tertiary care facilities were queried from 2010 to 2014 for fine-needle aspirates (FNAs) diagnosed as suspicious for Hürthle cell neoplasm (SHCN) or atypia of undetermined significance/follicular lesion of undetermined significance concerning for Hürthle cell neoplasm (AFHCN). Cytology diagnoses were rendered internally prior to AGEC testing. The patient demographics, FNA diagnosis, AGEC result, surgical procedure, and pathologic outcomes were recorded. RESULTS The cohort consisted of 134 patients with HCNs. Prior to AGEC availability, 62 patients underwent surgery: 81% (50/62) of patients had surgery, and 34% (17/50) of the resected index nodules were malignant. After introduction of the AGEC, 72 patients underwent AGEC testing: 65% (47/72) of patients had surgery, and 13% (6/46) of the resected nodules were malignant. Thirty-two percent (23/72) of patients had a benign AGEC result and did not undergo surgery, and 4% (3/72) had surgery despite a benign AGEC result with benign final pathology, whereas 63% (45/72) of patients had suspicious AGEC results, with 96% of these patients (43/45) undergoing surgery, and 14% (6/43) of these index nodules were malignant. CONCLUSIONS While 32% of tested patients declined surgery based on a benign AGEC, 86% of patients with suspicious AGEC findings had unnecessary surgery, reflecting a substantially lower rate of malignancy from what was previously reported for all indeterminate nodules. Given the approximate pretest malignancy risk of 25-35% for an FNA diagnosis of SHCN or AFHCN, a suspicious AGEC diagnosis does not increase the probability of malignancy in an HCN, and patients should be counseled accordingly.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/diagnosis
- Adenocarcinoma, Follicular/genetics
- Adenoma, Oxyphilic
- Adult
- Aged
- Biopsy, Fine-Needle
- Carcinoma/diagnosis
- Carcinoma/genetics
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Papillary
- Cohort Studies
- Diagnosis, Differential
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Middle Aged
- Oxyphil Cells/metabolism
- Oxyphil Cells/pathology
- Retrospective Studies
- Thyroid Cancer, Papillary
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/genetics
- Thyroid Nodule/diagnosis
- Thyroid Nodule/genetics
Collapse
Affiliation(s)
- Eran Brauner
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts
| | - Brittany J Holmes
- 2 Department of Pathology, Massachusetts General Hospital , Boston, Massachusetts
| | - Jeffrey F Krane
- 3 Department of Pathology, Brigham and Women's Hospital , Boston, Massachusetts
| | - Michiya Nishino
- 4 Department of Pathology, Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - David Zurakowski
- 5 Departments of Surgery and Anesthesia, Boston Children's Hospital , Boston, Massachusetts
| | - James V Hennessey
- 6 Department of Medicine, Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - William C Faquin
- 2 Department of Pathology, Massachusetts General Hospital , Boston, Massachusetts
| | - Sareh Parangi
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts
| |
Collapse
|
19
|
Le Pennec S, Konopka T, Gacquer D, Fimereli D, Tarabichi M, Tomás G, Savagner F, Decaussin-Petrucci M, Trésallet C, Andry G, Larsimont D, Detours V, Maenhaut C. Intratumor heterogeneity and clonal evolution in an aggressive papillary thyroid cancer and matched metastases. Endocr Relat Cancer 2015; 22:205-16. [PMID: 25691441 DOI: 10.1530/erc-14-0351] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The contribution of intratumor heterogeneity to thyroid metastatic cancers is still unknown. The clonal relationships between the primary thyroid tumors and lymph nodes (LN) or distant metastases are also poorly understood. The objective of this study was to determine the phylogenetic relationships between matched primary thyroid tumors and metastases. We searched for non-synonymous single-nucleotide variants (nsSNVs), gene fusions, alternative transcripts, and loss of heterozygosity (LOH) by paired-end massively parallel sequencing of cDNA (RNA-Seq) in a patient diagnosed with an aggressive papillary thyroid cancer (PTC). Seven tumor samples from a stage IVc PTC patient were analyzed by RNA-Seq: two areas from the primary tumor, four areas from two LN metastases, and one area from a pleural metastasis (PLM). A large panel of other thyroid tumors was used for Sanger sequencing screening. We identified seven new nsSNVs. Some of these were early events clonally present in both the primary PTC and the three matched metastases. Other nsSNVs were private to the primary tumor, the LN metastases and/or the PLM. Three new gene fusions were identified. A novel cancer-specific KAZN alternative transcript was detected in this aggressive PTC and in dozens of additional thyroid tumors. The PLM harbored an exclusive whole-chromosome 19 LOH. We have presented the first, to our knowledge, deep sequencing study comparing the mutational spectra in a PTC and both LN and distant metastases. This study has yielded novel findings concerning intra-tumor heterogeneity, clonal evolution and metastases dissemination in thyroid cancer.
Collapse
Affiliation(s)
- Soazig Le Pennec
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Tomasz Konopka
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - David Gacquer
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Danai Fimereli
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Maxime Tarabichi
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Gil Tomás
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Frédérique Savagner
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Myriam Decaussin-Petrucci
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Christophe Trésallet
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Guy Andry
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Denis Larsimont
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Vincent Detours
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Carine Maenhaut
- IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium IRIBHMWELBIOUniversité libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, BelgiumCHU d'AngersBâtiment IRIS, 4 rue Larrey, Angers F-49033, FranceEA 3143Université d'Angers, F-49033 Angers, FranceService d'Anatomie et Cytologie PathologiquesCentre de Biologie Sud - Bâtiment 3D, Centre Hospitalier Lyon Sud, 69495 Pierre Bénite Cedex, FranceHôpital Pitié-SalpêtrièreUniversité Pierre et Marie Curie, 47 Boulevard de l'Hôpital, 75013 Paris, FranceInstitut Jules Bordet121 Boulevard de Waterloo, 1000 Brussels, Belgium
| |
Collapse
|
20
|
Corver WE, van Wezel T, Molenaar K, Schrumpf M, van den Akker B, van Eijk R, Ruano Neto D, Oosting J, Morreau H. Near-haploidization significantly associates with oncocytic adrenocortical, thyroid, and parathyroid tumors but not with mitochondrial DNA mutations. Genes Chromosomes Cancer 2014; 53:833-44. [DOI: 10.1002/gcc.22194] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 05/28/2014] [Indexed: 01/06/2023] Open
Affiliation(s)
- Willem E. Corver
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Tom van Wezel
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Kees Molenaar
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Melanie Schrumpf
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Brendy van den Akker
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Ronald van Eijk
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Dina Ruano Neto
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Jan Oosting
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| | - Hans Morreau
- Department of Pathology; Leiden University Medical Center; RC Leiden Netherlands
| |
Collapse
|
21
|
Kurelac I, MacKay A, Lambros MBK, Di Cesare E, Cenacchi G, Ceccarelli C, Morra I, Melcarne A, Morandi L, Calabrese FM, Attimonelli M, Tallini G, Reis-Filho JS, Gasparre G. Somatic complex I disruptive mitochondrial DNA mutations are modifiers of tumorigenesis that correlate with low genomic instability in pituitary adenomas. Hum Mol Genet 2012; 22:226-38. [PMID: 23049073 DOI: 10.1093/hmg/dds422] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mitochondrial DNA (mtDNA) mutations leading to the disruption of respiratory complex I (CI) have been shown to exhibit anti-tumorigenic effects, at variance with those impairing only the function but not the assembly of the complex, which appear to contribute positively to cancer development. Owing to the challenges in the analysis of the multi-copy mitochondrial genome, it is yet to be determined whether tumour-associated mtDNA lesions occur as somatic modifying factors or as germ-line predisposing elements. Here we investigated the whole mitochondrial genome sequence of 20 pituitary adenomas with oncocytic phenotype and identified pathogenic and/or novel mtDNA mutations in 60% of the cases. Using highly sensitive techniques, namely fluorescent PCR and allele-specific locked nucleic acid quantitative PCR, we identified the most likely somatic nature of these mutations in our sample set, since none of the mutations was detected in the corresponding blood tissue of the patients analysed. Furthermore, we have subjected a series of 48 pituitary adenomas to a high-resolution array comparative genomic hybridization analysis, which revealed that CI disruptive mutations, and the oncocytic phenotype, significantly correlate with low number of chromosomal aberrations in the nuclear genome. We conclude that CI disruptive mutations in pituitary adenomas are somatic modifiers of tumorigenesis most likely contributing not only to the development of oncocytic change, but also to a less aggressive tumour phenotype, as indicated by a stable karyotype.
Collapse
Affiliation(s)
- Ivana Kurelac
- Dip. di Scienze Mediche e Chirurgiche, U.O. Genetica Medica, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Corver WE, Ruano D, Weijers K, den Hartog WCE, van Nieuwenhuizen MP, de Miranda N, van Eijk R, Middeldorp A, Jordanova ES, Oosting J, Kapiteijn E, Hovens G, Smit J, van Wezel T, Morreau H. Genome haploidisation with chromosome 7 retention in oncocytic follicular thyroid carcinoma. PLoS One 2012; 7:e38287. [PMID: 22675538 PMCID: PMC3365880 DOI: 10.1371/journal.pone.0038287] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 05/03/2012] [Indexed: 12/20/2022] Open
Abstract
Background Recurrent non-medullary thyroid carcinoma (NMTC) is a rare disease. We initially characterized 27 recurrent NMTC: 13 papillary thyroid cancers (PTC), 10 oncocytic follicular carcinomas (FTC-OV), and 4 non-oncocytic follicular carcinomas (FTC). A validation cohort composed of benign and malignant (both recurrent and non-recurrent) thyroid tumours was subsequently analysed (n = 20). Methods Data from genome-wide SNP arrays and flow cytometry were combined to determine the chromosomal dosage (allelic state) in these tumours, including mutation analysis of components of PIK3CA/AKT and MAPK pathways. Results All FTC-OVs showed a very distinct pattern of genomic alterations. Ten out of 10 FTC-OV cases showed near-haploidisation with or without subsequent genome endoreduplication. Near-haploidisation was seen in 5/10 as extensive chromosome-wide monosomy (allelic state [A]) with near-haploid DNA indices and retention of especially chromosome 7 (seen as a heterozygous allelic state [AB]). In the remaining 5/10 chromosomal allelic states AA with near diploid DNA indices were seen with allelic state AABB of chromosome 7, suggesting endoreduplication after preceding haploidisation. The latter was supported by the presence of both near-haploid and endoreduplicated tumour fractions in some of the cases. Results were confirmed using FISH analysis. Relatively to FTC-OV limited numbers of genomic alterations were identified in other types of recurrent NMTC studied, except for chromosome 22q which showed alterations in 6 of 13 PTCs. Only two HRAS, but no mutations of EGFR or BRAF were found in FTC-OV. The validation cohort showed two additional tumours with the distinct pattern of genomic alterations (both with oncocytic features and recurrent). Conclusions We demonstrate that recurrent FTC-OV is frequently characterised by genome-wide DNA haploidisation, heterozygous retention of chromosome 7, and endoreduplication of a near-haploid genome. Whether normal gene dosage on especially chromosome 7 (containing EGFR, BRAF, cMET) is crucial for FTC-OV tumour survival is an important topic for future research. Microarrays Data are made available at GEO (GSE31828).
Collapse
MESH Headings
- Adenocarcinoma, Follicular
- Aged
- Aged, 80 and over
- Alleles
- Carcinoma, Neuroendocrine
- Chromosomes, Human, Pair 7/genetics
- Cohort Studies
- DNA Mutational Analysis
- DNA, Neoplasm/genetics
- Disease Progression
- Female
- Flow Cytometry
- Gene Dosage/genetics
- Genes, Neoplasm/genetics
- Genome, Human/genetics
- Haploidy
- Homozygote
- Humans
- Male
- Middle Aged
- Models, Biological
- Oxyphil Cells/pathology
- Phenotype
- Polymorphism, Single Nucleotide/genetics
- Recurrence
- Reproducibility of Results
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/pathology
Collapse
Affiliation(s)
- Willem E. Corver
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail: (WEC); (HM)
| | - Dina Ruano
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karin Weijers
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Noel de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ronald van Eijk
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anneke Middeldorp
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jan Oosting
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen Kapiteijn
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Guido Hovens
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Smit
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail: (WEC); (HM)
| |
Collapse
|
23
|
|
24
|
Geyer FC, de Biase D, Lambros MBK, Ragazzi M, Lopez-Garcia MA, Natrajan R, Mackay A, Kurelac I, Gasparre G, Ashworth A, Eusebi V, Reis-Filho JS, Tallini G. Genomic profiling of mitochondrion-rich breast carcinoma: chromosomal changes may be relevant for mitochondria accumulation and tumour biology. Breast Cancer Res Treat 2012; 132:15-28. [PMID: 21509527 DOI: 10.1007/s10549-011-1504-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 04/04/2011] [Indexed: 02/08/2023]
Abstract
Oncocytic carcinomas are composed of mitochondrion-rich cells. Though recognised by the WHO classification as a histological special type of breast cancer, their status as a discrete pathological entity remains a matter of contention. Given that oncocytic tumours of other anatomical sites display distinct clinico-pathological and molecular features, we sought to define the molecular genetic features of mitochondrion-rich breast tumours and to compare them with a series of histological grade- and oestrogen receptor status-matched invasive ductal carcinomas of no special type. Seventeen mitochondrion-rich breast carcinomas, including nine bona fide oncocytic carcinomas, were profiled with antibodies against oestrogen, progesterone and androgen receptors, HER2, Ki67, GCDFP-15, chromogranin, epithelial membrane antigen, cytokeratin 7, cytokeratin 14, CD68 and mitochondria antigen. These tumours were microdissected and DNA extracted from samples with >70% of tumour cells. Fourteen cases yielded DNA of sufficient quality/quantity and were subjected to high-resolution microarray comparative genomic hybridisation analysis. The genomic profiles were compared to those of 28 grade- and oestrogen receptor status-matched invasive ductal carcinomas of no special type. Oncocytic and other mitochondrion-rich tumours did not differ significantly between themselves. As a group, mitochondrion-rich carcinomas were immunophenotypically heterogenous. Recurrent copy number changes were similar to those described in unselected breast cancers. However, unsupervised and supervised analysis identified a subset of mitochondrion-rich cancers, which often displayed gains of 11q13.1-q13.2 and 19p13. Changes in the latter two chromosomal regions have been shown to be associated with oncocytic tumours of the kidney and thyroid, respectively, and host several nuclear genes with specific mitochondrial function. Our results indicate that in a way akin to oncocytic tumours of other anatomical sites, at least a subset of mitochondrion-rich breast carcinomas may be underpinned by a distinct pattern of chromosomal changes potentially relevant for mitochondria accumulation and constitute a discrete molecular entity.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma/genetics
- Carcinoma/metabolism
- Carcinoma/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Chromosome Aberrations
- Chromosomes, Human/genetics
- Chromosomes, Human/metabolism
- Cluster Analysis
- Comparative Genomic Hybridization
- Female
- Humans
- Middle Aged
- Mitochondria/pathology
- Mitochondrial Proteins/genetics
- Neoplasm Grading
- Phenotype
- Receptors, Estrogen/metabolism
Collapse
Affiliation(s)
- Felipe C Geyer
- The Breakthrough Breast Cancer Research Centre, ICR, 237 Fulham Road, London SW3 6JB, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Da Silva L, James D, Simpson PT, Walker D, Vargas AC, Jayanthan J, Lakhani SR, McNicol AM. Tumor heterogeneity in a follicular carcinoma of thyroid: a study by comparative genomic hybridization. Endocr Pathol 2011; 22:103-7. [PMID: 21499728 DOI: 10.1007/s12022-011-9154-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We report a follicular carcinoma of thyroid that showed a range of histologic appearances, with microfollicular, macrofollicular/pseudopapillary, oncocytic, and poorly differentiated areas. We used comparative genomic hybridization to detect the major DNA copy number changes in each component, in order to study the inter-relationships among them. All showed gains in 11q and 17q, suggesting that these were early events in the development of the tumor, and these were the only changes in the follicular component. The other components each showed additional gains and losses, some unique to one component. The oncocytic component showed most changes, including loss on 16q in the region of the E-cadherin gene. This was associated with reduced intensity of immunostaining for E-cadherin specifically in that component. No mutations in the E-cadherin gene were detected in this component. The demonstration that some DNA copy number changes are consistent across each component suggests that they are all clonally related. The additional chromosomal and immunohistochemical heterogeneity across the macrofollicular/pseudopapillary, oncocytic, and poorly differentiated components would be consistent with the emergence of subclones, possibly as part of tumor progression.
Collapse
Affiliation(s)
- Leonard Da Silva
- The University of Queensland, UQ Centre for Clinical Research, Herston, Building 918/B71, Royal Brisbane and Women's Hospital, Herston, QLD, 4029, Australia
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Gasparre G, Bonora E, Tallini G, Romeo G. Molecular features of thyroid oncocytic tumors. Mol Cell Endocrinol 2010; 321:67-76. [PMID: 20184940 DOI: 10.1016/j.mce.2010.02.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 02/15/2010] [Accepted: 02/17/2010] [Indexed: 01/17/2023]
Abstract
Thyroid oncocytic neoplasms are tumors composed of cells characterized by an aberrant increase of mitochondrial mass. They represent a subset of thyroid tumors whose classification and clinical features has been a matter of controversy for clinicians and pathologists alike. The prevalence of oncocytic tumors in the thyroid gland, the relevance of the issues debated, and the obvious cellular derangement of oncocytic cells, namely a complete deregulation of the mitochondrial mass and metabolism, have spurred many investigators to study the molecular mechanism underlying the genesis of this peculiar cancer phenotype. Their findings, which are unraveling the tumor pathobiology, are the subject of the present review.
Collapse
Affiliation(s)
- Giuseppe Gasparre
- Dip. Scienze Ginecologiche, Ostetriche e Pediatriche, U.O. Genetica Medica, Pol. S.Orsola-Malpighi, Università di Bologna, Italy.
| | | | | | | |
Collapse
|
27
|
Parameswaran R, Brooks S, Sadler GP. Molecular pathogenesis of follicular cell derived thyroid cancers. Int J Surg 2010; 8:186-93. [PMID: 20097316 DOI: 10.1016/j.ijsu.2010.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Revised: 01/02/2010] [Accepted: 01/10/2010] [Indexed: 01/24/2023]
Abstract
Thyroid cancers are the most common endocrine malignancy. Radiation exposure, family history of thyroid cancer and some inherited conditions are the most important predisposing factors for the development of thyroid cancer. Three mitogenic signalling pathways have been described in the thyroid cell, which are influenced by various stimulatory and inhibitory hormones, growth factors and neurotransmitters. Various proto-oncogenes and oncogenes like ras, braf, trk, met and RET also play a role in the signal transduction systems. Two theories have been described in thyroid cancer pathogenesis, the foetal cell carcinogenesis theory and the more common, multistep carcinogenesis theory. The multistep carcinogenesis theory is now the accepted model in many human cancers, including thyroid cancer. The early events of tumour formation are the consequence of activation of either various growth factors or the proto-oncogenes like ras, met or ret. This results in the formation of differentiated thyroid cancers like the papillary, follicular or Hurthle cell cancers. The later stages of tumour formation involve further activation of proto-oncogenes and loss or inactivation of tumour suppressor genes like p53. Based on this theory, follicular carcinomas are generated from follicular adenomas and papillary carcinomas from precursor cells generated from thyrocytes. Anaplastic carcinoma may develop from papillary or follicular carcinoma by dedifferentiation. In this review article, we highlight the molecular pathogenesis of thyroid tumours.
Collapse
Affiliation(s)
- Rajeev Parameswaran
- Department of Endocrine Surgery, John Radcliffe Hospital, Headington, Oxford OX3 9DY, United Kingdom
| | | | | |
Collapse
|
28
|
Pita JM, Banito A, Cavaco BM, Leite V. Gene expression profiling associated with the progression to poorly differentiated thyroid carcinomas. Br J Cancer 2009; 101:1782-91. [PMID: 19809427 PMCID: PMC2778548 DOI: 10.1038/sj.bjc.6605340] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Poorly differentiated thyroid carcinomas (PDTC) represent a heterogeneous, aggressive entity, presenting features that suggest a progression from well-differentiated carcinomas. To elucidate the mechanisms underlying such progression and identify novel therapeutic targets, we assessed the genome-wide expression in normal and tumour thyroid tissues. METHODS Microarray analyses of 24 thyroid carcinomas - 7 classic papillary, 8 follicular variants of papillary (fvPTC), 4 follicular (FTC) and 5 PDTC - were performed and correlated with RAS, BRAF, RET/PTC and PAX8-PPARG alterations. Selected genes were validated by quantitative RT-PCR in an independent set of 28 thyroid tumours. RESULTS Unsupervised analyses showed that gene expression similarity was higher between PDTC and fvPTC, particularly for tumours harbouring RAS mutations. Poorly differentiated thyroid carcinomas presented molecular signatures related to cell proliferation, poor prognosis, spindle assembly checkpoint and cell adhesion. Compared with normal tissues, PTC had 307 out of 494 (60%) genes over-expressed, FTC had 137 out of 171 (80%) genes under-expressed, whereas PDTC had 92 out of 107 (86%) genes under-expressed, suggesting that gene downregulation is involved in tumour dedifferentiation. Significant UHRF1 and ITIH5 deregulated gene expression in PDTC, relatively to normal tissues, was confirmed by quantitative RT-PCR. CONCLUSION Our findings suggest that fvPTC are possible precursors of PDTC. Furthermore, UHRF1 and ITIH5 have a potential therapeutic/prognostic value for aggressive thyroid tumours.
Collapse
Affiliation(s)
- J M Pita
- Centro de Investigação de Patobiologia Molecular (CIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa 1099-023, Portugal
| | | | | | | |
Collapse
|
29
|
Baloch ZW, LiVolsi VA, Asa SL, Rosai J, Merino MJ, Randolph G, Vielh P, DeMay RM, Sidawy MK, Frable WJ. Diagnostic terminology and morphologic criteria for cytologic diagnosis of thyroid lesions: a synopsis of the National Cancer Institute Thyroid Fine-Needle Aspiration State of the Science Conference. Diagn Cytopathol 2008; 36:425-37. [PMID: 18478609 DOI: 10.1002/dc.20830] [Citation(s) in RCA: 537] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The National Cancer Institute (NCI) sponsored the NCI Thyroid Fine-needle Aspiration (FNA) State of the Science Conference on October 22-23, 2007 in Bethesda, MD. The two-day meeting was accompanied by a permanent informational website and several on-line discussion periods between May 1 and December 15, 2007 (http://thyroidfna.cancer.gov). This document summarizes matters regarding diagnostic terminology/classification scheme for thyroid FNA interpretation and cytomorphologic criteria for the diagnosis of various benign and malignant thyroid lesions. (http://thyroidfna.cancer.gov/pages/info/agenda/).
Collapse
Affiliation(s)
- Zubair W Baloch
- Department of Pathology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Affiliation(s)
- Stan Sidhu
- Royal North Shore Hospital, University of Sydney Endocrine Surgical Unit, Sydney, New South Wales, Australia
| |
Collapse
|
31
|
Sippel RS, Elaraj DM, Khanafshar E, Zarnegar R, Kebebew E, Duh QY, Clark OH. Tumor Size Predicts Malignant Potential in Hürthle Cell Neoplasms of the Thyroid. World J Surg 2008; 32:702-7. [DOI: 10.1007/s00268-007-9416-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Baloch ZW, LiVolsi VA. Our approach to follicular-patterned lesions of the thyroid. J Clin Pathol 2007; 60:244-50. [PMID: 16798933 PMCID: PMC1860564 DOI: 10.1136/jcp.2006.038604] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2006] [Indexed: 01/08/2023]
Abstract
Follicular-patterned lesions of the thyroid are common; these include hyperplastic/adenomatoid nodules, follicular adenoma, follicular carcinoma and follicular variants of papillary carcinoma. Most of these lesions can be diagnosed with ease; however, there is a controversial subgroup. In this review, we present our diagnostic approach based on our experience with the histological diagnosis of these tumours, which can help in appropriate clinical management.
Collapse
Affiliation(s)
- Zubair W Baloch
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
33
|
Cocco L, Manzoli L, Palka G, Martelli AM. Nuclear phospholipase C beta1, regulation of the cell cycle and progression of acute myeloid leukemia. ACTA ACUST UNITED AC 2005; 45:126-35. [PMID: 16024064 DOI: 10.1016/j.advenzreg.2005.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A large number of observations have hinted at the fact that location impinges on function of some of the main players of nuclear inositol lipid cycle. PLC beta1 is a well-known example, given that it has been shown that only the enzyme located in the nucleus targets the cyclin D3/cdk4 complex, playing, in turn, a key role in the control of normal progression through the G1 phase of the cell cycle. The PLC beta1 gene, which is constituted of 36 small exons and large introns, maps on the short arm of human chromosome 20 (20pl2, nearby markers D20S917 and D20S177) with the specific probe (PAC clone HS881E24) spanning from exon 19 to 32 of the gene itself. The chromosome band 20pl2 has been shown to be rearranged in human diseases such as solid tumors without a more accurate definition of the alteration, maybe because of the absence of candidate genes or specific probes. Moreover, non-specific alterations in chromosome 20 have been found in patients affected by MDS and acute myeloid leukemia AML. MDS is an adult hematological disease that evolves into AML in about 30% of the cases. The availability of a highly specific probe gave an opportunity to perform in patients affected with MDS/AML, associated with normal karyotype, painting and FISH analysis aimed to check the PLC beta1 gene, given that this signaling molecule is a key player in the control of some checkpoints of the normal progression through the cell cycle. FISH analysis disclosed in a small group of MDS/AML patients with normal karyotype the monoallelic deletion of the PLC beta1 gene. In contrast, PLC beta4, another gene coding for a signaling molecule, located on 20pl2.3 at a distance as far as less than 1 Mb from PLC beta1, is unaffected in MDS patients with the deletion of PLC beta1 gene, hinting at an interstitial deletion. The MDS patients, bearing the deletion, rapidly evolved to AML, whilst the normal karyotype MDS patients, showing non-deletion of PLC beta1 gene, are still alive at least 24 months after the diagnosis. The immunocytochemical analysis using an anti PLC beta1 monoclonal antibody showed that all the AML/MDS patients who were normal at FISH analysis also had normal staining of the nucleus, which is a preferential site for PLC beta1. In contrast, the monoallelic deletion gave rise to a dramatic decrease of the nuclear staining suggesting a decreased expression of the nuclear PLC beta1. The reported data strengthen the contention of a key role played by PLC beta1 in the nucleus, suggest a possible involvement of PLC beta1 in the progression of MDS to AML and pave the way for a larger investigation aimed at identifying a possible high risk group among MDS patients with a normal karyotype.
Collapse
Affiliation(s)
- Lucio Cocco
- Cellular Signaling Laboratory, Department of Anatomical Sciences, University of Bologna, Via Irnerio 48, Bologna 40126, Italy.
| | | | | | | |
Collapse
|
34
|
Freier K, Flechtenmacher C, Walch A, Ohl S, Devens F, Burke B, Hassfeld S, Lichter P, Joos S, Hofele C. Copy number gains on 22q13 in adenoid cystic carcinoma of the salivary gland revealed by comparative genomic hybridization and tissue microarray analysis. ACTA ACUST UNITED AC 2005; 159:89-95. [PMID: 15860365 DOI: 10.1016/j.cancergencyto.2004.09.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2004] [Revised: 09/13/2004] [Accepted: 09/14/2004] [Indexed: 11/18/2022]
Abstract
Adenoid cystic carcinoma (ACC) of the salivary gland is a neoplasm characterized by slow but inevitable local progression and terminal hematogenous metastasis. To detect novel imbalanced chromosomal regions associated with tumorigenesis, we used chromosomal comparative genomic hybridization to screen 27 ACC. The most common aberration was copy number gain of 22q13 (nine cases) followed by gains of 16p (seven cases) and 17q (four cases) and copy number losses on 6q (six cases). To further delineate the prevalence of 22q13 copy number gains in ACC, fluorescence in situ hybridization was performed for five bacterial/phage artificial chromosome (BAC/PAC) probes from the 22q13 consensus region with 57 ACC on a tissue microarray. The overall prevalence of copy number gains on 22q13 was 30% of the tumors in the fluorescence in situ hybridization analysis, irrespective of histologic differentiation (cribriform/tubular vs. solid) or tumor event (primary vs. recurrent). We therefore assume that copy number gain of 22q13 is a novel frequent finding in ACC that may be involved in the initial pathogenesis of this neoplasm by proto-oncogene activation.
Collapse
Affiliation(s)
- Kolja Freier
- Abteilung Molekulare Genetik, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Hürthle cell carcinoma (HCC) has been reported to have variable clinical behavior. The objective of this study was to determine the important prognostic factors in patients with HCC. It was a retrospective investigation of 33 patients with HCC treated in our institution from 1976 to 2002. The average age of our 33 patients with HCC was 55.2 years (range 20-82 years; 23 women, 10 men). Fifteen patients (45.5%) presented with a T2 tumor, 7 (21.2%) with a T3 tumor, and 8 (24.2%) with a T4 tumor; the tumor stage was unknown in three patients. Coexisting papillary carcinoma occurred in six patients (18%), 9% of whom presented with metastatic disease. Twelve patients (36.4%) had metastases, four of whom (12.2%) had persistent HCC, and 8 (24.2%) developed recurrent HCC (range 1-9 years). Eight of these twelve patients (66.7%) died from the HCC. The average follow-up time was 5.5 years (range 1-16 years). Altogether, 3 of 23 women and 5 of 10 men died from their HCC ( p < 0.05). Of the 22 patients with T2-T3 tumors, 5 (22.7%) developed metastases and 1 patient died; 5 of 8 patients (62.5%) with T4 tumors developed metastases and died ( p < 0.01). Patients who were treated by less than total thyroidectomy had a worse prognosis by univariate analysis ( p < 0.01) but not by multivariate analysis. Survival time for patients with persistent disease was shorter than for those with recurrent disease ( p < 0.05). Multivariate analysis, however, revealed no difference for extent of operation or those with persistent and recurrent disease. The cause-specific survivals were 74% and 49% at 5 and 10 years, respectively. Disease-free survivals were 65.0% and 40.5% at 5 and 10 years, respectively. Our findings show that gender and stage of disease influence the prognosis of patients with HCC.
Collapse
Affiliation(s)
- Yevgeniya Kushchayeva
- Endocrine Surgical Oncology, Department of Surgery, University of California-San Francisco/Mount Zion Medical Center, 1600 Divisadero Street, San Francisco, CA 94143-1674, USA
| | | | | | | |
Collapse
|
36
|
Stankov K, Pastore A, Toschi L, McKay J, Lesueur F, Kraimps JL, Bonneau D, Gibelin H, Levillain P, Volante M, Papotti M, Romeo G. Allelic loss on chromosomes 2q21 and 19p 13.2 in oxyphilic thyroid tumors. Int J Cancer 2004; 111:463-7. [PMID: 15221978 DOI: 10.1002/ijc.20259] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hürthle thyroid tumors are characterized by frequent numerical chromosomal aberrations, including aneuploidy or polyploidy, losses and gains of some chromosomal regions and DNA fragmentation. In recent years, great attention has been paid to the combined analysis of morphologic and genetic features of oxyphilic tumors and to the elucidation of their pathogenesis. We analyzed for loss of heterozygosity (LOH) of the candidate regions for TCO (thyroid tumor with cell oxyphilia) and NMTC1 (nonmedullary thyroid carcinoma 1), 2 loci already mapped on chromosomes 19p13.2 and 2q21, respectively. Matched normal and tumor DNA samples from 70 patients with sporadic oxyphilic thyroid tumors and 20 with sporadic follicular tumors were subjected to microsatellite analysis using 10 markers on 19p13.2 and 6 markers on 2q21. This approach led us to the observation of a more significant LOH in oxyphilic than in follicular tumors. Allelic loss in tumor samples was evenly distributed in both 19p13.2 and 2q21 regions, in accordance with the established linkage of TCO and NMTC1 for inherited tumors. In order to investigate the possible contribution of both susceptibility loci in oxyphilic tumors, the family that led to the original mapping of TCO locus was reanalyzed for the markers in the 2q21 region. This led to the exclusion of linkage with the NMTC1 locus and to the refutation of the digenic inheritance hypothesis at least in this family.
Collapse
Affiliation(s)
- Karmen Stankov
- Dipartimento di Medicina Interna, Cardioangiologia ed Epatologia, Unità Operativa di Genetica Medica, Policlinico S. Orsola-Malpighi, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Thyroid tumorigenesis and carcinogenesis accompany progressive loss of thyroid-specific differentiated functions. Some thyroid cancers are or become dedifferentiated, and they become refractory to efficacy-proven conventional therapies such as radioiodine ablation therapy and thyrotropin (TSH)-suppressive therapy. Redifferentiation therapy by either redifferentiating agents or gene transfer of differentiation-related genes may retard tumor growth and make tumors respond to conventional therapies.
Collapse
Affiliation(s)
- Jin-Woo Park
- Department of Surgery, College of Medicine, Chungbuk National University, San 62 Kaeshin-dong, Heungdok-gu, Cheongju, 361-763 South Korea
| | | |
Collapse
|
38
|
Dettori T, Frau DV, Garcia JL, Pierantoni G, Lee C, Hernandez JM, Fusco A, Morton CC, Vanni R. Comprehensive conventional and molecular cytogenetic characterization of B-CPAP, a human papillary thyroid carcinoma-derived cell line. ACTA ACUST UNITED AC 2004; 151:171-7. [PMID: 15172756 DOI: 10.1016/j.cancergencyto.2003.09.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2003] [Revised: 09/05/2003] [Accepted: 09/24/2003] [Indexed: 11/19/2022]
Abstract
Cell lines derived from different thyroid tumor histotypes are useful for the in vitro study of both the phenotypic and genetic features of these cancers. Although karyotypic changes are known to be associated with thyroid lesions, the chromosome patterns of only a few cell lines have been published. Herein, we report an extensive conventional and molecular cytogenetic investigation of the human papillary thyroid carcinoma derived cell line B-CPAP. Morphological studies and expression of tumor markers in this cell line have been reported previously, but no detailed characterization on the origin of the chromosome markers is available. B-CPAP cells have a rather stable hypertriploid karyotype, with chromosome polysomies and structural chromosome abnormalities featuring whole chromosome arm imbalances. Chromosome banding revealed a main clone with nine chromosome markers, and fluorescence in situ hybridization (FISH) with whole chromosome paint (wcp), partial chromosome paint (pcp), and centromeric probes clarified their origin. The use of centromeric probes provided accurate refinement of the rearrangements classified as whole-arm translocations by banding and FISH with wcp probes. Both chromosomal and array-based comparative genomic hybridization experiments confirmed the cytogenetic characterization of this cell line. Moreover, the use of fluorescence immunophenotyping and interphase cytogenetics as a tool for the investigation of neoplasms (FICTION) technique, which simultaneously shows nuclear ploidy and cytoplasmic immunofluorescence, detailed the oncocytic feature of the cells. Intriguingly, despite their origin, they lack most of the features expressed in papillary thyroid tumor cells and have a chromosomal pattern reminiscent of that of a subgroup of oncocytic malignant thyroid tumors.
Collapse
Affiliation(s)
- T Dettori
- Dipartimento di Scienze e Tecnologie Biomediche, Università di Cagliari, Cittadella Universitaria, Monserrato, Cagliari 09042, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Lo Vasco VR, Calabrese G, Manzoli L, Palka G, Spadano A, Morizio E, Guanciali-Franchi P, Fantasia D, Cocco L. Inositide-specific phospholipase c β1 gene deletion in the progression of myelodysplastic syndrome to acute myeloid leukemia. Leukemia 2004; 18:1122-6. [PMID: 15085153 DOI: 10.1038/sj.leu.2403368] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Myelodysplastic syndrome (MDS) is an adult hematological disease that evolves into acute myeloid leukemia (AML) in about 30% of the cases. The availability of a highly specific probe moved us to perform in patients affected with MDS/AML, associated with normal karyotype, painting and fluorescence in situ hybridization (FISH) analysis aimed to check the inositide-specific phospholipase C (PI-PLC) beta1 gene, a player in the control of some checkpoints of the cell cycle. Here we present a preliminary observation in which FISH analysis disclosed in a small group of MDS/AML patients with normal karyotype the monoallelic deletion of the PI-PLCbeta1 gene. On the contrary, PI-PLC beta4, another gene coding for a signaling molecule, located on 20p12.3 at a distance as far as less than 1Mb from PI-PLCbeta1, is unaffected in MDS patients with the deletion of PI-PLC beta1 gene, hinting at an interstitial deletion. The MDS patients, bearing the deletion, rapidly evolved to AML. The data suggest the possible involvement of PI-PLCbeta1 in the progression of the disease and pave the way for a larger investigation aimed at identifying a possible high-risk group among MDS patients with a normal karyotype.
Collapse
Affiliation(s)
- V R Lo Vasco
- Cellular Signalling Laboratory, Department of Anatomical Sciences, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Jin-Woo Park
- Department of Surgery, Chungbuk National University, College of Medicine & Hospital, Korea.
| |
Collapse
|
41
|
Dettori T, Frau DV, Lai ML, Mariotti S, Uccheddu A, Daniele GM, Tallini G, Faa G, Vanni R. Aneuploidy in oncocytic lesions of the thyroid gland: diffuse accumulation of mitochondria within the cell is associated with trisomy 7 and progressive numerical chromosomal alterations. Genes Chromosomes Cancer 2003; 38:22-31. [PMID: 12874783 DOI: 10.1002/gcc.10238] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oncocytic cells are characterized by a greatly increased number of mitochondria that distend the cell cytoplasm and result in a distinctive granular appearance of the cell on conventional histology sections. Oncocytes are frequently found in metabolically active human tissues including the thyroid gland, and, as a general rule, when their proportion in a thyroid tumor is greater than 75% the tumor is referred to as oncocytic (Hürthle cell) adenoma or carcinoma. Such tumors represent a subset of thyroid lesions, and recently, both interphase fluorescence in situ hybridization (FISH) and comparative genomic hybridization (CGH) studies reported that they may show aneuploidy, with widespread numerical chromosomal alterations. In contrast, very few cases have been studied by conventional cytogenetic analysis. Whether the cells with chromosomal changes are the same as those with mitochondrial accumulation or whether lesions only partially composed of oncocytic cells also have cytogenetic alterations is unclear. To investigate the relationship between acquisition of the oncocytic phenotype and numerical chromosomal changes, we analyzed a random selection of thyroid lesions with (18 cases) and without (11 cases) morphological evidence of oncocytic differentiation. Lesions with oncocytes included hyperplastic nodules, adenomas, Hürthle cell tumors, and papillary carcinomas with lymphocytic stroma (Whartin-like tumors of the thyroid). Karyotypic changes were analyzed by cytogenetic analysis, FISH, or CGH, and the results were compared with in situ analysis of mitochondrial accumulation by immunofluorescence. A striking correlation between the presence of oncocytes and the presence of aneuploid katyotypes was seen in the oncocytic follicular thyroid nodules, but not in the oncocytic papillary tumors. Structural chromosome changes or normal karyotypes were observed in the lesions lacking oncocytic features. Extending the FICTION technique to the evaluation of a cytoplasmic antigen (mitochondrial membrane antigen), we pursued the simultaneous visualization of both mitochondrial increase and numerical chromosomal alterations, and showed that oncocytes of follicular lesions are prone to become aneuploid. Our data support the contention that follicular tumors composed of oncocytes should be regarded as a distinct subset.
Collapse
Affiliation(s)
- Tinuccia Dettori
- Department of Biomedical Sciences and Technology, University of Cagliari, Cagliari, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Machens A, Holzhausen HJ, Lautenschläger C, Thanh PN, Dralle H. Enhancement of lymph node metastasis and distant metastasis of thyroid carcinoma. Cancer 2003; 98:712-9. [PMID: 12910514 DOI: 10.1002/cncr.11581] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The mechanisms of local and distant metastases are imperfectly understood. The goal of the current study was to add to the body of knowledge regarding local and distant metastases of thyroid malignancies. METHODS The authors performed multivariate analysis of 573 patients who underwent surgery between November 1994 and May 2002 for follicular (FTC; n = 100), papillary (PTC; n = 236), or medullary thyroid carcinoma (MTC; n = 237) at a university hospital. RESULTS In multivariate analysis, extrathyroidal extension consistently evolved as the key risk factor for both lymph node metastasis and distant metastasis. This correlation was most pronounced in MTC and least pronounced in FTC. The risk of lymph node metastasis also increased with reoperative status in patients with MTC and with primary tumor diameter in patients with MTC (tumor diameter > 10 mm) and patients with PTC (tumor diameter > 20 mm). In the PTC group, lymph node metastasis was more common among patients younger than age 45. In the MTC group, extrathyroidal growth and distant metastasis were associated exclusively with lymph node metastasis. Lymph node metastasis was the only secondary risk factor for distant metastasis. In the analysis of risk factors for distant metastasis in the FTC and PTC groups, no interaction was found between extrathyroidal growth and lymph node metastasis. This finding suggests that extrathyroidal growth and lymph node metastasis of FTC and PTC, and presumably also MTC, represent separate mechanisms and routes of distant metastasis. CONCLUSIONS Screening for both local residual disease and distant metastases should be intensified in the high-risk population of patients whose primary tumors exhibit large diameters, extrathyroidal growth, or lymph node metastasis.
Collapse
Affiliation(s)
- Andreas Machens
- Department of General, Visceral and Vascular Surgery, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany.
| | | | | | | | | |
Collapse
|
43
|
Brunaud L, Zarnegar R, Wada N, Magrane G, Wong M, Duh QY, Davis O, Clark OH. Chromosomal aberrations by comparative genomic hybridization in thyroid tumors in patients with familial nonmedullary thyroid cancer. Thyroid 2003; 13:621-9. [PMID: 12964966 DOI: 10.1089/105072503322239952] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE Nonmedullary thyroid cancer is the most common form of thyroid cancer and its familial form (FNMTC) is increasingly recognized as a distinct clinical entity. However, the genetic background of FNMTC is still poorly understood and the causative gene(s) have not yet been identified. METHODS Because comparative genomic hybridization allows for screening of the entire tumor genome simultaneously for chromosomal gains and/or losses without prior knowledge of potential aberrations, we used this technique in thyroid normal and neoplastic samples from FNMTC patients (1) to analyze whether chromosomal aberrations would correlate with inheritance pattern, and/or clinicopathologic features and (2) to compare comparative genomic hybridization (CGH) findings in familial tumors with those already known in sporadic differentiated thyroid cancers. RESULTS No common germline or somatic chromosomal aberrations were observed in patients with FNMTC because the frequencies and most locations of chromosomal aberrations in familial tumors were also common in sporadic tumors. However, some somatic aberrations were only found in familial tumors (gains in 2q, 3q, 18p, and 19p). Common aberrations in familial tumors corresponded to several locations of candidate genes already reported for sporadic thyroid tumorigenesis. CONCLUSIONS Our findings suggest that chromosomal aberrations in thyroid tumors in patients with FNMTC are not related to inheritance pattern but rather to tumorigenesis.
Collapse
Affiliation(s)
- Laurent Brunaud
- Endocrine Surgical Oncology Fellow, University of California San Francisco UCSF/Mount Zion Medical Center, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Impact of molecular cytogenetics on localization and identification of cancer-related genes in endocrine tumor development. ACTA ACUST UNITED AC 2003. [DOI: 10.1097/00060793-200306000-00004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
45
|
Hu J, Khanna V, Jones MW, Surti U. Comparative study of primary and recurrent ovarian serous carcinomas: comparative genomic hybridization analysis with a potential application for prognosis. Gynecol Oncol 2003; 89:369-75. [PMID: 12798697 DOI: 10.1016/s0090-8258(03)00056-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVES The purpose of this study is to comparatively characterize genomic imbalances in primary and recurrent ovarian serous carcinomas and to identify genomic alterations that may be used as a marker for prognosis. METHODS Twenty ovarian serous carcinomas were studied by comparative genomic hybridization (CGH). RESULTS Genomic alterations were found in all of the tumors. The most common regions involving gain of DNA copy numbers are 1q41q44, 8q22q24, 19p12q13.1, 20q12q13, 3q26q29, 12p12p13, 2p22p25, 7p14p21, 5p15.2p15.3, and 17q22q25. The most common regions with loss of DNA copy numbers are Xp11.2q13, 4q31q35, Xp21p22.3, 18q22q23, 13q22q31, 9p22p24, and 16q22q24. High-level gains were detected at chromosomal regions of 1q41q44, 2p22p25, 3q26q29, and 19p12q13.1. Comparative analysis of primary and recurrent tumors showed that gains of 2p22p25, 19p12q13.1, and 20q12q13 and loss of 5q14q22 were more common in the recurrent high-grade tumors. About 85% of the tumors showed increases in DNA copy numbers in the regions (2p and 8q) harboring the myc family gene. Patients with tumor containing fewer than seven chromosomal aberrations showed longer survival time. CONCLUSION The myc oncogene family may play a role in the pathogenesis of ovarian serous carcinomas. Our study suggests that tumors with gains of 2p22p25, 19p12q13.1, and 20q12q13 and loss of 5q14q22 may be at high risk for recurrence. Furthermore, the patients' survival time inversely correlates with the numbers of chromosomal alterations found in their tumors. CGH analysis may have a clinical application in predicting prognosis and risk of recurrence in patients with ovarian serous carcinomas.
Collapse
Affiliation(s)
- J Hu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | | | | | | |
Collapse
|