1
|
Yip AT, Kim TH, Peluso EM, Jacobsen SE, Yeh MW, Lechner MG. Medullary Thyroid Carcinoma and Clinical Outcomes in Heterozygous Carriers of the RET K666N Germline Pathogenic Variant. JCEM CASE REPORTS 2025; 3:luaf002. [PMID: 39963300 PMCID: PMC11831074 DOI: 10.1210/jcemcr/luaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Indexed: 02/20/2025]
Abstract
Medullary thyroid carcinoma (MTC) is a rare neuroendocrine tumor of the thyroid parafollicular C-cells associated with activating mutations in the rearranged during transfection (RET) kinase proto-oncogene. We report the clinical outcomes of a family with a rare germline RET K666N pathogenic variant discovered incidentally by genetic testing performed for breast cancer risk stratification in an asymptomatic 24-year-old woman. Subsequent genetic testing identified the same pathogenic variant in her 21-year-old sister, 60-year-old father, and 84-year-old paternal grandmother. The proband and her sister had no biochemical or imaging evidence of MTC. The 60-year-old father had mildly elevated serum calcitonin and multiple thyroid nodules on ultrasound. Fine-needle aspirate thyroid biopsy cytology suggested MTC so he underwent total thyroidectomy. Surgical pathology demonstrated bilateral subcentimeter foci of MTC and C-cell hyperplasia. The 84-year-old grandmother was also found to have multiple thyroid nodules and elevated calcitonin but declined further evaluation. There was no biochemical evidence of other multiple endocrine neoplastic type 2 (MEN2)-associated tumors (ie, parathyroid adenoma, pheochromocytoma) in the family. These data, along with prior rare reports in the literature, suggest that monoallelic germline RET K666N pathogenic variants carry a risk of familial MTC that demonstrate age-dependent expressivity but low penetrance of other MEN2 tumors in affected individuals.
Collapse
Affiliation(s)
- Allison T Yip
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Teresa H Kim
- Department of Pathology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Esther M Peluso
- UCLA/Caltech Medical Scientist Training Program, UCLA Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Steven E Jacobsen
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
- Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael W Yeh
- Department of Endocrine Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Melissa G Lechner
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
2
|
Tomuleasa C, Tigu AB, Munteanu R, Moldovan CS, Kegyes D, Onaciu A, Gulei D, Ghiaur G, Einsele H, Croce CM. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct Target Ther 2024; 9:201. [PMID: 39138146 PMCID: PMC11323831 DOI: 10.1038/s41392-024-01899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Receptor tyrosine kinases (RTKs), a category of transmembrane receptors, have gained significant clinical attention in oncology due to their central role in cancer pathogenesis. Genetic alterations, including mutations, amplifications, and overexpression of certain RTKs, are critical in creating environments conducive to tumor development. Following their discovery, extensive research has revealed how RTK dysregulation contributes to oncogenesis, with many cancer subtypes showing dependency on aberrant RTK signaling for their proliferation, survival and progression. These findings paved the way for targeted therapies that aim to inhibit crucial biological pathways in cancer. As a result, RTKs have emerged as primary targets in anticancer therapeutic development. Over the past two decades, this has led to the synthesis and clinical validation of numerous small molecule tyrosine kinase inhibitors (TKIs), now effectively utilized in treating various cancer types. In this manuscript we aim to provide a comprehensive understanding of the RTKs in the context of cancer. We explored the various alterations and overexpression of specific receptors across different malignancies, with special attention dedicated to the examination of current RTK inhibitors, highlighting their role as potential targeted therapies. By integrating the latest research findings and clinical evidence, we seek to elucidate the pivotal role of RTKs in cancer biology and the therapeutic efficacy of RTK inhibition with promising treatment outcomes.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania.
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Cristian-Silviu Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
3
|
Zhou L, Li J, Zhang X, Xu Z, Yan Y, Hu K. An integrative pan cancer analysis of RET aberrations and their potential clinical implications. Sci Rep 2022; 12:13913. [PMID: 35978072 PMCID: PMC9386015 DOI: 10.1038/s41598-022-17791-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/31/2022] [Indexed: 11/18/2022] Open
Abstract
RET (rearranged during transfection), encoding a tyrosine kinase receptor, is a novel therapeutic target for cancers. The aberrations of RET are commonly found in cancers. Here, we profiled a comprehensive genomic landscape of RET mutations, copy number variants (CNVs), co-occurrence of RET and its mRNA expression and methylation levels in pan cancer, paving the way to the development of new RET-targeted therapies in clinic. Analysis of RET somatic mutations, CNVs, co-occurrence, mRNA expression and methylation were performed among 32 cancer types from The Cancer Genome Atlas (TCGA) dataset covering a total of 10,953 patients with 10,967 samples. RET aberrations were found in 3.0% of diverse cancers. The top two RET-altered tumors were skin cutaneous melanoma (SKCM) and uterine corpus endometrial carcinoma (UCEC) with dominant mutations in the other and PKinase_Tyr domains. RET-G823E and RET-S891L were most commonly found in SKCM and UCEC. Thyroid carcinoma (THCA) demonstrated the highest rate of coiled-coil domain containing 6 (CCDC6)-RET fusions, which constitutively activate RET kinase. Two FDA-approved RET inhibitors-pralsetinib and selpercatinib have been implied for the treatment of patients with RET S891L mutant UCEC and the treatment of patients with metastatic RET-fusion positive THCA and non-small cell lung cancer (NSCLC) at therapeutic level 1. We also identified four RET M918T-altered cases in patients with pheochromocytoma and paraganglioma (PCPG), which may induce drug resistance against multikinase inhibitors. Next, 273 co-occurring aberrations, most frequently in Notch signaling, TGF-β pathway, cell cycle, and Ras-Raf-MEK-Erk/JNK signaling, were uncovered among 311 RET altered cases. TP53 mutations (162 patients) leads to the most significant co-occurrence associated with RET aberrations. Furthermore, the RET expression was found most significantly increased in breast invasive carcinoma (BRCA) and neck squamous cell carcinoma (HNSC), as compared to their corresponding normal tissues. At last, patients with higher expression and sequence variant frequency have a worse prognosis, such as sarcoma patients. This work provided a profound and comprehensive analysis of RET and co-occurred alterations, RET mRNA expression and the clinical significance in pan cancer, offering new insights into targeted therapy for patients with RET anomalies.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Juanni Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaofang Zhang
- Departments of Burn and Plastic, Ningxiang People's Hospital, Hunan University of Chinese Medicine, Changsha, 410600, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuanliang Yan
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Kuan Hu
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
4
|
Román-Gil MS, Pozas J, Rosero-Rodríguez D, Chamorro-Pérez J, Ruiz-Granados Á, Caracuel IR, Grande E, Molina-Cerrillo J, Alonso-Gordoa T. Resistance to RET targeted therapy in Thyroid Cancer: Molecular basis and overcoming strategies. Cancer Treat Rev 2022; 105:102372. [DOI: 10.1016/j.ctrv.2022.102372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/07/2022]
|
5
|
Kohno T, Tabata J, Nakaoku T. REToma: a cancer subtype with a shared driver oncogene. Carcinogenesis 2020; 41:123-129. [PMID: 31711124 DOI: 10.1093/carcin/bgz184] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/17/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
RET (REarranged during Transfection), which encodes a receptor tyrosine kinase for members of the glial cell line-derived neurotrophic factor, plays a role as driver oncogene in a variety of human cancers. Fusion of RET with several partner genes has been detected in papillary thyroid, lung, colorectal, pancreatic and breast cancers, and tyrosine kinase inhibitors (TKIs) for RET (particularly RET-specific inhibitors) show promising therapeutic effects against such cancers. Oncogenic mutations within the extracellular cysteine-rich and intracellular kinase domains of RET drive medullary thyroid carcinogenesis; the same mutations are also observed in a small subset of diverse cancers such as lung, colorectal and breast cancers. Considering the oncogenic nature of RET mutants, lung, colorectal and breast cancers are predicted to respond to RET TKIs in a manner similar to medullary thyroid cancer. In summary, cancers carrying oncogenic RET alterations as a driver mutation could be collectively termed 'REToma' and treated with RET TKIs in a tissue-agnostic manner.
Collapse
Affiliation(s)
- Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Junya Tabata
- Division of Genome Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Takashi Nakaoku
- Division of Genome Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| |
Collapse
|
6
|
Qi XP, Jin BY, Li PF, Wang S, Zhao YH, Cao ZL, Yu XH, Cheng J, Fang XD, Zhao JQ. RET S409Y Germline Mutation and Associated Medullary Thyroid Carcinoma. Thyroid 2019; 29:1447-1456. [PMID: 31364476 DOI: 10.1089/thy.2018.0385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background: Inherited medullary thyroid carcinoma (MTC) is primarily caused by RET mutations that are commonly localized in exons 5, 8, 10, 11, and 13-16. In this study, we report pedigrees for individuals with MTC that harbor a germline S409Y variant within exon 6 of the RET proto-oncogene. Methods: Targeted sequencing was used to diagnose four apparently sporadic MTC index cases carrying the germline RET S409Y (c.1226 C>A) variant. Subsequently, 27 relatives of these individuals underwent clinical and genetic assessments and/or thyroid surgery. Furthermore, in silico analyses and in vitro assays were performed to predict or verify the potential oncogenic activity of the S409Y variant. Results: Overall, 15 of 31 participants were found to carry the RET S409Y variant. Of these, 6 presented with isolated MTC (mean age 50.2 years; range 41-75 years), of which 3 presented with neck lymph node metastases and 2 presented with distant liver or lung metastases. Among the remaining 9 carriers, 3 (mean age 56 years; range 41-76 years) had elevated serum calcium-stimulated calcitonin (sCtn) or concurrent marginally elevated serum calcitonin (Ctn) levels, whereas the other 6 (mean age 37.5 years; range 14-52 years) exhibited typical Ctn/sCtn levels (p < 0.05). None of the 15 carriers in these 4 families presented clinical evidence of pheochromocytoma, hyperparathyroidism, or Hirschsprung's disease. In silico analyses revealed that S409Y was a "possibly damaging" mutation that could affect the RET protein inter-domain interface. An in vitro assay revealed that the phosphorylation level of RET tyrosine 905 was relatively higher in the RET S409Y mutant than in wild-type (WT) RET. Moreover, transfection of HEK 293 cells with S409Y enhanced the phosphorylation activity of AKT, ERK pathways, and it increased cell proliferation compared with WT RET, but to a lesser degree than that for the RET C618Y and C634Y mutations. Conclusions: This study demonstrates that the novel germline RET S409Y variant is likely pathogenic and is associated with lower penetrance of MTC than that for the C618Y and C634Y mutations. Individuals with S409Y should be managed using a personalized approach, and additionally, "at-risk" family members should be evaluated. Additional studies are needed to elucidate the correlation between the S409Y mutation and multiple endocrine neoplasia type 2-specific tumors.
Collapse
Affiliation(s)
- Xiao-Ping Qi
- Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Wenzhou Medical University, Hangzhou, China
| | - Bai-Ye Jin
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng-Fei Li
- Department of Research and Development, XY Biotechnology Co. Ltd., Hangzhou, China
| | - Sheng Wang
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Jeddah, Saudi Arabia
| | - Yi-Hua Zhao
- Department of Urologic Surgery, Yueqing People's Hospital, Wenzhou Medical University, Yueqing, China
| | - Zhi-Lie Cao
- Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Wenzhou Medical University, Hangzhou, China
| | - Xiu-Hua Yu
- Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Wenzhou Medical University, Hangzhou, China
| | - Jun Cheng
- Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Wenzhou Medical University, Hangzhou, China
| | - Xu-Dong Fang
- Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Wenzhou Medical University, Hangzhou, China
| | - Jian-Qiang Zhao
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
7
|
Ahmed AA, Vundamati D, Farooqi M, Repnikova E, Zinkus T, Hetherington M, Paulson L. Next-Generation Sequencing in the Diagnosis of Rare Pediatric Sinonasal Tumors. EAR, NOSE & THROAT JOURNAL 2019; 100:NP263-NP268. [PMID: 31550935 DOI: 10.1177/0145561319863371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The diagnosis of desmoid fibromatosis or other spindle cell tumors in the sinonasal region is very rare in children and needs to be thoroughly confirmed with immunohistochemical and/or molecular tests. We report 2 patients with such rare tumors and describe the use of next-generation sequencing in their evaluation. A 3-year-old female had a 4.4-cm midline nasal cavity mass involving the bony septum and extending into the base of the skull bilaterally. The moderate cellular fibroblastic proliferation revealed areas of thick keloid-like collagen bands and other areas with myxoid edematous stroma. Deep targeted sequencing identified a novel G34V mutation in the CTNNB1 gene consistent with desmoid fibromatosis. An 11-month-old male infant presented with a right nasal mass that extended through the cribriform plate into the anterior cranial fossa and involved the right ethmoid sinus and adjacent right orbit. Histology revealed an infiltrative atypical fibrous proliferation with focal calcifications that was negative for CTNNB1 and GNAS mutations. A novel RET E511K variant was identified in the tumor and later was also found in the germline and hence rendered of unknown significance. Both cases highlight the utility of next-generation sequencing in the evaluation of pediatric sinonasal spindle cell tumors that may have overlapping pathologic features. Reporting of rare or novel variants in tumor-only sequencing should be cautiously evaluated in children and pairing with germline sequencing may be needed to avoid the pitfall of assigning uncommon variants.
Collapse
Affiliation(s)
- Atif A Ahmed
- Department of Pathology, 4204Children's Mercy Hospital/University of Missouri, Kansas City, MO, USA
| | - Divya Vundamati
- Department of Pathology, 4204Children's Mercy Hospital/University of Missouri, Kansas City, MO, USA
| | - Midhat Farooqi
- Department of Pathology, 4204Children's Mercy Hospital/University of Missouri, Kansas City, MO, USA
| | - Elena Repnikova
- Department of Pathology, 4204Children's Mercy Hospital/University of Missouri, Kansas City, MO, USA
| | - Timothy Zinkus
- Department of Radiology, 4204Children's Mercy Hospital/University of Missouri, Kansas City, MO, USA
| | - Maxine Hetherington
- Department of Pediatrics Hematology-Oncology, 4204Children's Mercy Hospital/University of Missouri, Kansas City, MO, USA
| | - Lorien Paulson
- Department of Surgery, 4204Children's Mercy Hospital/University of Missouri, Kansas City, MO, USA
| |
Collapse
|
8
|
Khan SA, Ci B, Xie Y, Gerber DE, Beg MS, Sherman SI, Cabanillas ME, Busaidy NL, Burtness BA, Heilmann AM, Bailey M, Ross JS, Sher DJ, Ali SM. Unique mutation patterns in anaplastic thyroid cancer identified by comprehensive genomic profiling. Head Neck 2019; 41:1928-1934. [PMID: 30758123 DOI: 10.1002/hed.25634] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 08/27/2018] [Accepted: 12/19/2018] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Anaplastic thyroid cancer (ATC) is a highly aggressive thyroid cancer. Those ATC with genomic alterations (GAs) in TSC2, ALK, and BRAF may respond to targeted therapies. METHODS Comprehensive genomic profiling on 90 ATC specimens identified base substitutions, short insertions and deletions, amplifications, copy number alterations, and genomic rearrangements in up to 315 cancer-related genes and 28 genes commonly rearranged in cancer. RESULTS Median patient age was 65 (range, 33-86) years, 50 patients were male. There was a mean of 4.2 GA per case, range 1-11. The most common GA were TP53 (66%), BRAF (34%), TERT (32%), CDKN2A (32%), and NRAS (26%). BRAF V600E and NRAS/HRAS/KRAS alteration were mutually exclusive. BRAF, CDKN2A, PIK3CA, and JAK2 were more frequent in patients >70 years of age; while myc, PTEN, and NRAS were more common in those ≤50 years. CONCLUSION ATC shows many GA with potential therapeutic significance and suggesting different molecular pathways can lead to ATC.
Collapse
Affiliation(s)
- Saad A Khan
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bo Ci
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yang Xie
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - David E Gerber
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Muhammad S Beg
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Steven I Sherman
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara A Burtness
- Department of Internal Medicine, Yale School of Medicine; Developmental Therapeutics Program, Yale Cancer Center, New Haven, Connecticut
| | | | - Mark Bailey
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | | | - David J Sher
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Siraj M Ali
- Foundation Medicine, Inc., Cambridge, Massachusetts
| |
Collapse
|
9
|
Paratala BS, Chung JH, Williams CB, Yilmazel B, Petrosky W, Williams K, Schrock AB, Gay LM, Lee E, Dolfi SC, Pham K, Lin S, Yao M, Kulkarni A, DiClemente F, Liu C, Rodriguez-Rodriguez L, Ganesan S, Ross JS, Ali SM, Leyland-Jones B, Hirshfield KM. RET rearrangements are actionable alterations in breast cancer. Nat Commun 2018; 9:4821. [PMID: 30446652 PMCID: PMC6240119 DOI: 10.1038/s41467-018-07341-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 10/25/2018] [Indexed: 11/23/2022] Open
Abstract
Fusions involving the oncogenic gene RET have been observed in thyroid and lung cancers. Here we report RET gene alterations, including amplification, missense mutations, known fusions, novel fusions, and rearrangements in breast cancer. Their frequency, oncogenic potential, and actionability in breast cancer are described. Two out of eight RET fusions (NCOA4-RET and a novel RASGEF1A-RET fusion) and RET amplification were functionally characterized and shown to activate RET kinase and drive signaling through MAPK and PI3K pathways. These fusions and RET amplification can induce transformation of non-tumorigenic cells, support xenograft tumor formation, and render sensitivity to RET inhibition. An index case of metastatic breast cancer progressing on HER2-targeted therapy was found to have the NCOA4-RET fusion. Subsequent treatment with the RET inhibitor cabozantinib led to a rapid clinical and radiographic response. RET alterations, identified by genomic profiling, are promising therapeutic targets and are present in a subset of breast cancers. Fusions of the gene RET have been described in thyroid and lung cancers. Here, the AUs identify RET gene alterations, including known fusions, novel fusions, and rearrangements in breast cancer (BC) that are involved in the tumorigenic process and show the benefit of RET therapy in a recurrent BC patient carrying the NCOA4-RET fusion.
Collapse
Affiliation(s)
- Bhavna S Paratala
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | - Jon H Chung
- Foundation Medicine, Cambridge, MA, 02139, USA
| | - Casey B Williams
- Avera Cancer Institute Center for Precision Oncology, Sioux Falls, SD, 57105, USA
| | | | - Whitney Petrosky
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | - Kirstin Williams
- Avera Cancer Institute Center for Precision Oncology, Sioux Falls, SD, 57105, USA
| | | | | | - Ellen Lee
- University Radiology Group, New Brunswick, NJ, 08901, USA
| | - Sonia C Dolfi
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | - Kien Pham
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School and Rutgers Robert Wood Johnson Medical School, Newark, NJ, 07103, USA
| | - Stephanie Lin
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | - Ming Yao
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | - Atul Kulkarni
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | - Frances DiClemente
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School and Rutgers Robert Wood Johnson Medical School, Newark, NJ, 07103, USA
| | - Lorna Rodriguez-Rodriguez
- Rutgers University, Piscataway, NJ, 08854, USA.,Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Shridar Ganesan
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA.,Rutgers University, Piscataway, NJ, 08854, USA
| | | | - Siraj M Ali
- Foundation Medicine, Cambridge, MA, 02139, USA
| | - Brian Leyland-Jones
- Avera Cancer Institute Center for Precision Oncology, Sioux Falls, SD, 57105, USA.
| | - Kim M Hirshfield
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA. .,Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
10
|
Jaber T, Hyde SM, Cote GJ, Grubbs EG, Giles WH, Stevens CA, Dadu R. A Homozygous RET K666N Genotype With an MEN2A Phenotype. J Clin Endocrinol Metab 2018; 103:1269-1272. [PMID: 29408964 DOI: 10.1210/jc.2017-02402] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/29/2018] [Indexed: 02/13/2023]
Abstract
CONTEXT Germline RET K666N mutation has been described as a pathogenic mutation with low disease penetrance for medullary thyroid cancer (MTC) without other features of multiple endocrine neoplasia type 2A. We describe a patient with homozygous RET K666N mutation with MTC and bilateral pheochromocytoma (PHEO). CASE DESCRIPTION A 59-year-old woman received a diagnosis of MTC after biopsy of two thyroid nodules. Coincident biochemical and radiologic testing was suspicious for bilateral PHEO, confirmed after bilateral adrenalectomy. There was no evidence of primary hyperparathyroidism (PHPT). She had a total thyroidectomy with neck dissection revealing bilateral MTC with lymph node metastases. Germline RET testing identified homozygous K666N mutations. Genetic testing of family members showed that both adult children harbor a heterozygous K666N mutation. Her 32-year-old son had an elevated calcitonin level and underwent thyroidectomy, which identified MTC. Her 30-year-old daughter had a normal calcitonin level. Prophylactic thyroidectomy showed C-cell hyperplasia only. Three of seven other family members were tested and found to carry the mutation. All had normal calcitonin levels, and none had biochemical evidence of PHEO or PHPT. Given the absence of PHEO in reported RET K666N families, our proband underwent genetic testing for causes of hereditary paragangliomas or PHEO. No additional mutations were identified. CONCLUSIONS Here we report a case of a homozygous RET K666N mutation leading to coincident MTC and PHEO. Heterozygous presentations of RET K666N mutations have low penetrance for isolated MTC. We believe that the gene dosage associated with the homozygosity of this variant contributed to the occurrence of bilateral PHEO.
Collapse
Affiliation(s)
- Tania Jaber
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Samuel M Hyde
- Department of Clinical Cancer Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Gilbert J Cote
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth G Grubbs
- Department of Surgical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Wesley H Giles
- Department of Surgery, The University of Tennessee College of Medicine, Chattanooga, Tennessee
| | - Cathy A Stevens
- Department of Pediatrics, Division of Medical Genetics, The University of Tennessee College of Medicine, Chattanooga, Tennessee
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
11
|
Lebeault M, Pinson S, Guillaud-Bataille M, Gimenez-Roqueplo AP, Carrie A, Barbu V, Pigny P, Bezieau S, Rey JM, Delvincourt C, Giraud S, Veyrat-Durebex C, Saulnier P, Bouzamondo N, Chabbert M, Blin J, Mohamed A, Romanet P, Borson-Chazot F, Rohmer V, Barlier A, Mirebeau-Prunier D. Nationwide French Study of RET Variants Detected from 2003 to 2013 Suggests a Possible Influence of Polymorphisms as Modifiers. Thyroid 2017; 27:1511-1522. [PMID: 28946813 DOI: 10.1089/thy.2016.0399] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The presence of single nucleotide polymorphisms (SNPs) in the REarranged during Transfection (RET) gene has been investigated with regard to their potential role in the development or progression of medullary thyroid cancer or pheochromocytomas (PHEO) in patients with the multiple endocrine neoplasia type 2 (MEN2) syndrome. The aim of this study was to evaluate the spectrum of RET variants in France between 2003 and 2013, and to evaluate the impact of SNPs on the MEN2 A phenotype. METHODS In this retrospective cohort study, RET variants were screened in 5109 index cases, and RET pathogenic variants were screened in 2214 relatives. Exons 5, 8, 10, 11, 13, 14, 15, and 16 were characterized by Sanger sequencing. RET pathogenic variants, RET variants with unknown functional significance (VUS), and four RET SNP variants-G691S (rs1799939), L769L (rs1800861), S836S (rs1800862), and S904S (rs1800863)-were characterized and are reported in index cases. In silico analysis and classification following the recommendation of the American College of Medical Genetics and Genomics was performed for RET VUS. Each patient's age at the time of diagnosis, sex, and the endocrine neoplasias present at molecular diagnosis were recorded. RESULTS Twenty-six single VUS in RET without any well-defined risk profiles were found in 33 patients. Nine of these were considered probably pathogenic, 11 of uncertain significance, and six as probably benign. Three double pathogenic variants found in three patients were classified as pathogenic. A study of the entire cohort showed that patients carrying pathogenic variants or VUS in RET together with PHEO were diagnosed earlier than the others. The presence of the G691S SNP, or a combination of SNPs, increased the risk of developing PHEO but did not modify the date of the diagnosis. No association was found between SNPs and medullary thyroid cancer or hyperparathyroidism. CONCLUSIONS The findings propose a classification of 15 of the 26 VUS in RET without any well-defined risk profiles and suggest that the G691S SNP, or a combination of SNPs, may be associated with the development of PHEO.
Collapse
Affiliation(s)
| | - Stéphane Pinson
- 2 Laboratoire de Génétique Moléculaire , CHU Lyon, Lyon France
- 3 Réseau TenGen , France
| | - Marine Guillaud-Bataille
- 3 Réseau TenGen , France
- 4 Département de Biologie et Pathologie Médicale, Gustave Roussy, Université de Paris-Saclay, Villejuif, France
| | - Anne-Paule Gimenez-Roqueplo
- 3 Réseau TenGen , France
- 5 Service de Génétique, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Alain Carrie
- 3 Réseau TenGen , France
- 6 Centre de Génétique Moléculaire Chromosomique, Assistance Publique Hôpitaux de Paris , Paris, France
| | - Véronique Barbu
- 3 Réseau TenGen , France
- 7 Laboratoire Commun de Biologie et Génétique Moléculaires , HUEP, SAT, AP-HP Paris, France
| | - Pascal Pigny
- 3 Réseau TenGen , France
- 8 Laboratoire de Biochimie et Oncologie Moléculaire , CHU Lille, Lille, France
| | - Stéphane Bezieau
- 3 Réseau TenGen , France
- 9 Laboratoire de Génétique Moléculaire , CHU Nantes, Nantes, France
| | - Jean-Marc Rey
- 3 Réseau TenGen , France
- 10 Laboratoire de Biopathologie Cellulaire et Tissulaire des Tumeurs , CHU Montpellier, Montpellier, France
| | - Chantal Delvincourt
- 3 Réseau TenGen , France
- 11 Laboratoire de Biologie Oncologique , CHU Reims, Reims, France
| | - Sophie Giraud
- 2 Laboratoire de Génétique Moléculaire , CHU Lyon, Lyon France
- 3 Réseau TenGen , France
| | - Charlotte Veyrat-Durebex
- 3 Réseau TenGen , France
- 12 UMR CNRS 6015-INSERMU1083, Laboratoire MITOVASC, Université d'Angers, Angers, France
- 13 Département de Biochimie et Génétique, CHU Angers , Angers, France
| | - Patrick Saulnier
- 14 Cellule de Méthodologie et Biostatistiques, Délégation à la Recherche Clinique et l'Innovation-DRCI, CHU Angers , Angers, France
| | - Nathalie Bouzamondo
- 3 Réseau TenGen , France
- 13 Département de Biochimie et Génétique, CHU Angers , Angers, France
| | - Marie Chabbert
- 12 UMR CNRS 6015-INSERMU1083, Laboratoire MITOVASC, Université d'Angers, Angers, France
| | - Julien Blin
- 15 Institut National du Cancer-INCa , Paris, France
| | - Amira Mohamed
- 3 Réseau TenGen , France
- 16 Aix Marseille Univ, CNRS, CRN2M, UMR 7286, and APHM La Conception Hospital, Molecular Biology Laboratory, Marseille, France
| | - Pauline Romanet
- 3 Réseau TenGen , France
- 16 Aix Marseille Univ, CNRS, CRN2M, UMR 7286, and APHM La Conception Hospital, Molecular Biology Laboratory, Marseille, France
| | - Francoise Borson-Chazot
- 3 Réseau TenGen , France
- 17 Hospices Civils de Lyon, Pôle IMER; Université Claude Bernard Lyon 1, HESPER EA 7425 Lyon, France
| | - Vincent Rohmer
- 1 Service d'Endocrinologie, CHU Angers , Angers, France
- 3 Réseau TenGen , France
| | - Anne Barlier
- 3 Réseau TenGen , France
- 16 Aix Marseille Univ, CNRS, CRN2M, UMR 7286, and APHM La Conception Hospital, Molecular Biology Laboratory, Marseille, France
| | - Delphine Mirebeau-Prunier
- 3 Réseau TenGen , France
- 12 UMR CNRS 6015-INSERMU1083, Laboratoire MITOVASC, Université d'Angers, Angers, France
- 13 Département de Biochimie et Génétique, CHU Angers , Angers, France
| |
Collapse
|
12
|
Hyde SM, Cote GJ, Grubbs EG. Genetics of Multiple Endocrine Neoplasia Type 1/Multiple Endocrine Neoplasia Type 2 Syndromes. Endocrinol Metab Clin North Am 2017; 46:491-502. [PMID: 28476233 DOI: 10.1016/j.ecl.2017.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Multiple endocrine neoplasia syndromes types 1 and 2 represent well-characterized yet clinically heterogeneous hereditary conditions for which diagnostic and management recommendations exist; genetic testing for these inherited endocrinopathies is included in these guidelines and is an important part of identifying affected patients and their family members. Understanding of these mature syndromes is challenged as more individuals undergo genetic testing and genetic data are amassed, with the potential to create clinical conundrums that may have an impact on individualized approaches to management and counseling. Clinicians who diagnose and treat patients with MEN syndromes should be aware of these possibilities.
Collapse
Affiliation(s)
- Samuel M Hyde
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, USA; Department of Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, USA
| | - Gilbert J Cote
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, USA
| | - Elizabeth G Grubbs
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Xu JY, Grubbs EG, Waguespack SG, Jimenez C, Gagel RF, Sosa JA, Sellin RV, Dadu R, Hu MI, Trotter CS, Jackson M, Rich TA, Hyde SM, Sherman SI, Cote GJ. Medullary Thyroid Carcinoma Associated with Germline RET K666N Mutation. Thyroid 2016; 26:1744-1751. [PMID: 27673361 PMCID: PMC5175438 DOI: 10.1089/thy.2016.0374] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Multiple endocrine neoplasia type 2 is an autosomal dominant inherited syndrome caused by activating mutations in the RET proto-oncogene. The RETK666N DNA variant was previously reported in two isolated medullary thyroid carcinoma (MTC) cases, but no family studies are available, and its oncogenic significance remains unknown. METHODS The clinical features, genetic data, and family information of eight index MTC patients with a germline RETK666N variant were assessed. RESULTS Four probands presented with MTC and extensive nodal metastasis, one with biopsy-confirmed distant metastasis. Two additional probands presented with localized disease. However, nodal status was not available. Of the final two probands, one had an incidental 1.5 mm MTC and C-cell hyperplasia uncovered after surgery for papillary thyroid carcinoma, and one had two foci of MTC (largest dimension 2.3 cm) detected after surgery for dysphagia. Genetic screening identified 16 additional family members carrying the K666N variant (aged 5-90 years), 11 of whom have documented evaluation for MTC. Of these, only two were found to have elevated basal serum calcitonin upon screening, and the remaining patients had calcitonin levels within the reference range. One patient who elected to have a thyroidectomy at 70 years of age was confirmed to have MTC. The other subject, 57 years old, elected surveillance. Four prophylactic thyroidectomies were performed, with one case of C-cell hyperplasia at 20 years and three cases that revealed normal pathology at ages 21, 30, and 30 years. None of the K666N DNA variant carriers had evidence of primary hyperparathyroidism or pheochromocytoma. CONCLUSIONS From this case series, the largest such experience to date, it is concluded that the RETK666N variant is likely pathogenic and associated with low penetrance of MTC. However, the findings are insufficient to define its pathogenicity clearly and make firm recommendations for screening and treatment. Given the potential benefit associated with early detection of aberrant C-cell growth, and the noninvasive nature of genetic testing, "at risk" individuals should be screened, and if the K666N variant is identified, they should be managed using a personalized screening approach for detection of MTC.
Collapse
Affiliation(s)
- Jian Yu Xu
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
- Division of Endocrinology, Baylor College of Medicine, Houston, Texas
| | - Elizabeth G. Grubbs
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven G. Waguespack
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert F. Gagel
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julie A. Sosa
- Departments of Surgery and Medicine, Duke Cancer Institute, and Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina
| | - Rena V. Sellin
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mimi I. Hu
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chardria S. Trotter
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michelle Jackson
- Department of Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thereasa A. Rich
- Department of Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samuel M. Hyde
- Department of Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven I. Sherman
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gilbert J. Cote
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
14
|
Kato S, Subbiah V, Marchlik E, Elkin SK, Carter JL, Kurzrock R. RET Aberrations in Diverse Cancers: Next-Generation Sequencing of 4,871 Patients. Clin Cancer Res 2016; 23:1988-1997. [PMID: 27683183 DOI: 10.1158/1078-0432.ccr-16-1679] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/22/2016] [Accepted: 09/04/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Aberrations in genetic sequences encoding the tyrosine kinase receptor RET lead to oncogenic signaling that is targetable with anti-RET multikinase inhibitors. Understanding the comprehensive genomic landscape of RET aberrations across multiple cancers may facilitate clinical trial development targeting RETExperimental Design: We interrogated the molecular portfolio of 4,871 patients with diverse malignancies for the presence of RET aberrations using Clinical Laboratory Improvement Amendments-certified targeted next-generation sequencing of 182 or 236 gene panels.Results: Among diverse cancers, RET aberrations were identified in 88 cases [1.8% (88/4, 871)], with mutations being the most common alteration [38.6% (34/88)], followed by fusions [30.7% (27/88), including a novel SQSTM1-RET] and amplifications [25% (22/88)]. Most patients had coexisting aberrations in addition to RET anomalies [81.8% (72/88)], with the most common being in TP53-associated genes [59.1% (52/88)], cell cycle-associated genes [39.8% (35/88)], the PI3K signaling pathway [30.7% (27/88)], MAPK effectors [22.7% (20/88)], or other tyrosine kinase families [21.6% (19/88)]. RET fusions were mutually exclusive with MAPK signaling pathway alterations. All 72 patients harboring coaberrations had distinct genomic portfolios, and most [98.6% (71/72)] had potentially targetable coaberrations with either an FDA-approved or an investigational agent. Two cases with lung (KIF5B-RET) and medullary thyroid carcinoma (RET M918T) that responded to a vandetanib (multikinase RET inhibitor)-containing regimen are shown.Conclusions:RET aberrations were seen in 1.8% of diverse cancers, with most cases harboring actionable, albeit distinct, coexisting alterations. The current report suggests that optimal targeting of patients with RET anomalies will require customized combination strategies. Clin Cancer Res; 23(8); 1988-97. ©2016 AACR.
Collapse
Affiliation(s)
- Shumei Kato
- Department of Medicine, Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California, San Diego, Moores Cancer Center, San Diego, California.
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Razelle Kurzrock
- Department of Medicine, Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California, San Diego, Moores Cancer Center, San Diego, California
| |
Collapse
|
15
|
Silva AL, Carmo F, Moura MM, Domingues R, Espadinha C, Leite V, Cavaco B, Bugalho MJ. Identification and characterization of two novel germline RET variants associated with medullary thyroid carcinoma. Endocrine 2015; 49:366-72. [PMID: 25725622 DOI: 10.1007/s12020-015-0559-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 02/19/2015] [Indexed: 12/18/2022]
Abstract
Activating germline mutations in the RET proto-oncogene are responsible for about 98 % of the familial forms of medullary thyroid carcinoma (MTC), which represent 25 % of all MTC cases. The search for germline mutations in this gene is important for the recognition of hereditary forms of MTC and further identification of at-risk relatives who may benefit from early clinical intervention. Genotype-phenotype correlations are well established for most disease-causing RET mutations, allowing risk stratification. The association of a new RET variant with the MTC phenotype and familial predisposition requires the assessment of its functional and clinical significance. The aim of this study was to evaluate the oncogenic potential of two newly identified RET germline variants associated with late-onset MTC. In vitro functional assays were designed to address the transforming potential of novel RET variants, through their expression in non-transformed cells, and comparing their effect with wild-type RET. The new variants were identified in codons 515 (p.C515W) and 636 (p.T636M) located, respectively, in exons 8 and 11, thus resulting in amino acid substitutions in the extracellular region of the tyrosine kinase receptor RET. Through functional assays, we observed increased cell growth and proliferation, loss of contact inhibition, and a stimulation of cell migration, suggesting that these new RET variants hold some relevant transforming potential. The transforming potential of these novel RET variants was of low-grade, when compared to that of RET MEN2A-causing mutation p.C634R, probably explaining the mild phenotype characterized by late onset and low clinical aggressiveness.
Collapse
Affiliation(s)
- A L Silva
- Unidade de Investigação de Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Rua Professor Lima Basto, 1099-023, Lisbon, Portugal,
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Yamazaki M, Hanamura T, Ito KI, Uchino S, Sakurai A, Komatsu M. A newly identified missense mutation in RET codon 666 is associated with the development of medullary thyroid carcinoma. Endocr J 2014; 61:1141-4. [PMID: 25319874 DOI: 10.1507/endocrj.ej14-0334] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A 38-year-old woman with a thyroid nodule measuring approximately 2 cm was suspected to have medullary thyroid carcinoma (MTC) because of markedly elevated serum calcitonin and carcinoembryonic antigen levels. There were no signs of pheochromocytoma, whereas primary hyperparathyroidism was suspected based on the findings of inappropriate hypersecretion of parathyroid hormone although no parathyroid tumor was detected with imaging studies. RET mutation analysis revealed a novel germline missense mutation in codon 666, c.1997A>G (p.K666R). She underwent total thyroidectomy with lymphadenectomy and simultaneous total parathyroidectomy with autotransplantation of parathyroid tissue. She was given calcium lactate and alfacalcidol to prevent postoperative hypocalcemia. Pathological findings of the thyroid tumor were compatible with MTC, but the resected parathyroid glands were intact. To our knowledge, c.1997A>G (p.K666R) is a new RET mutation. This is a minor variant, but it is significant because of the possible pathogenicity in tumor formation. It is often difficult to determine whether MTC is generated as part of MEN2-related disease or familial MTC when it is a unique manifestation. In addition, it is still unclear whether all missense mutations in this codon reported previously will lead to the same clinical course and prognosis. Further careful observations of clinical presentation are required to determine the clinical features associated with this variant.
Collapse
Affiliation(s)
- Masanori Yamazaki
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
Medullary thyroid cancer (MTC) accounts for ~10% of thyroid carcinomas and occurs in sporadic and hereditary forms. Early diagnosis significantly impacts the clinical course, management and outcome of the disease. The identification of germline-activating mutations of the rearranged during transfection oncogene in patients with hereditary MTC led to significant progress in the diagnostic and therapeutic approach, thus improving the quality of care provided, and consequently, disease prognosis. In the present review, various aspects of genetic screening (GS) in MTC will be covered, which elucidate the value of GS in guiding clinical decision making, therapy selection and appropriate genetic counseling of the affected families. GS should be offered to every MTC patient, based on the personal and family medical history, to allow optimal clinical management and follow-up.
Collapse
Affiliation(s)
- Theodora Pappa
- a Department of Medical Therapeutics, Endocrine Unit, Athens University School of Medicine, Alexandra Hospital, Athens, Greece
| | - Maria Alevizaki
- a Department of Medical Therapeutics, Endocrine Unit, Athens University School of Medicine, Alexandra Hospital, Athens, Greece
- b Department of Endocrinology, Metabolism and Diabetes, Athens University School of Medicine, Evgenideion Hospital, Athens, Greece
| |
Collapse
|
18
|
Valente FOF, Dias da Silva MR, Camacho CP, Kunii IS, Bastos AU, da Fonseca CCN, Simião HPC, Tamanaha R, Maciel RMB, Cerutti JM. Comprehensive analysis of RET gene should be performed in patients with multiple endocrine neoplasia type 2 (MEN 2) syndrome and no apparent genotype-phenotype correlation: an appraisal of p.Y791F and p.C634Y RET mutations in five unrelated Brazilian families. J Endocrinol Invest 2013; 36:975-81. [PMID: 23723040 DOI: 10.3275/8997] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND We previously identified a four-generation family with medullary thyroid cancer (MTC) and a germline p.Y791F RET mutation whose cancer lacked a strong genotype-phenotype correlation. The entire gene coding region of the RET gene should be sequenced when genotype-phenotype discrepancies are observed in patients with multiple endocrine neoplasia type 2 (MEN 2), even if a RET hotspot mutation has been identified. METHODS A new genetic test was performed in the index case of this family with the p.Y791F RET germline mutation. The entire coding region of the RET gene was investigated by direct sequencing of PCR products. Once a mutation was identified, the target exon was sequenced in all at-risk relatives. RESULTS An additional p.C634Y germline mutation in the RET gene was identified in the reported family. The double mutation occurred in cis and segregated with the phenotype. Through the Brazilian Genetic Screening Program developed at our institution, we additionally report the combination of these two mutations (p.C634Y/p.Y791F) in the RET gene in four other unrelated families. The overall penetrance of MTC and pheochromocytoma in patients with the p.C634Y/p.Y791F mutations was 79% and 13%, respectively. CONCLUSION Our data emphasises that a comprehensive analysis of the RET gene may reveal multiple germline mutations in MEN 2 patients who exhibit an atypical clinical course of the disease.
Collapse
Affiliation(s)
- F O F Valente
- Laboratory of Molecular and Translational Endocrinology, Division of Endocrinology, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Toledo SPA, Lourenço DM, Toledo RA. A differential diagnosis of inherited endocrine tumors and their tumor counterparts. Clinics (Sao Paulo) 2013; 68:1039-56. [PMID: 23917672 PMCID: PMC3715026 DOI: 10.6061/clinics/2013(07)24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 03/26/2013] [Indexed: 12/15/2022] Open
Abstract
Inherited endocrine tumors have been increasingly recognized in clinical practice, although some difficulties still exist in differentiating these conditions from their sporadic endocrine tumor counterparts. Here, we list the 12 main topics that could add helpful information and clues for performing an early differential diagnosis to distinguish between these conditions. The early diagnosis of patients with inherited endocrine tumors may be performed either clinically or by mutation analysis in at-risk individuals. Early detection usually has a large impact in tumor management, allowing preventive clinical or surgical therapy in most cases. Advice for the clinical and surgical management of inherited endocrine tumors is also discussed. In addition, recent clinical and genetic advances for 17 different forms of inherited endocrine tumors are briefly reviewed.
Collapse
Affiliation(s)
- Sergio P A Toledo
- Division of Endocrinology, Endocrine Genetics Unit (LIM-25), Faculdade de Medicina da Universidade de São Paulo, São Paulo/SP, Brazil.
| | | | | |
Collapse
|
20
|
The optimal range of RET mutations to be tested: European comments to the guidelines of the American Thyroid Association. Thyroid Res 2013; 6 Suppl 1:S8. [PMID: 23514012 PMCID: PMC3599734 DOI: 10.1186/1756-6614-6-s1-s8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In the 9th ETA-CRN Meeting (September 2009, Lisbon) some recommendations from the American Thyroid Association (ATA) guidelines for the management of medullary thyroid cancer (MTC) were discussed by an European Panel of Experts (EPE). Among the 12 ATA recommendations related to hereditary MTC and to the optimal range of RET mutations to be tested (recommendations 1-5 and 9-15), 7 were shared and 5 were not shared by the EPE. In the present paper, the related comments and suggestions will be reported and discussed.
Collapse
|
21
|
Sarika HL, Papathoma A, Garofalaki M, Vasileiou V, Vlassopoulou B, Anastasiou E, Alevizaki M. High prevalence of exon 8 G533C mutation in apparently sporadic medullary thyroid carcinoma in Greece. Clin Endocrinol (Oxf) 2012; 77:857-62. [PMID: 22676047 DOI: 10.1111/j.1365-2265.2012.04462.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Genetic screening for ret mutation has become routine practice in the evaluation of medullary thyroid carcinoma (MTC). Approximately 25% of these tumours are familial, and they occur as components of the multiple endocrine neoplasia type 2 syndromes (MEN 2A and 2B) or familial MTC. In familial cases, the majority of mutations are found in exons 10, 11, 13, 14 or 15 of the ret gene. A rare mutation involving exon 8 (G533C) has recently been reported in familial cases of MTC in Brazil and Greece; some of these cases were originally thought to be sporadic. The aim of this study was to re-evaluate a series of sporadic cases of MTC, with negative family history, and screen them for germline mutations in exon 8. DESIGN AND PATIENTS Genomic DNA was extracted from peripheral lymphocytes in 129 unrelated individuals who had previously been characterized as 'sporadic' based on the negative family history and negative screening for ret gene mutations. Samples were analysed in Applied Biosystems 7500 real-time PCR and confirmed by sequencing. MEASUREMENTS AND RESULTS The G533C exon 8 mutation was identified in 10 of 129 patients with sporadic MTC. Asymptomatic gene carriers were subsequently identified in other family members. CONCLUSION In our study, we found that 7·75% patients with apparently sporadic MTC do carry G533C mutation involving exon 8 of ret. We feel that there is now a need to include exon 8 mutation screening in all patients diagnosed as sporadic MTC, in Greece.
Collapse
Affiliation(s)
- H L Sarika
- 1st Endocrine Section and Diabetes Center, Alexandra Hospital, Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
22
|
Mastroianno S, Torlontano M, Scillitani A, D'Aloiso L, Verrienti A, Bonfitto N, De Bonis A, D'Agruma L, Muscarella LA, Guarnieri V, Dicembrino F, Maranghi M, Durante C, Filetti S. Coexistence of multiple endocrine neoplasia type 1 and type 2 in a large Italian family. Endocrine 2011; 40:481-5. [PMID: 21678021 DOI: 10.1007/s12020-011-9501-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 06/03/2011] [Indexed: 10/18/2022]
Abstract
To describe the coexistence of mutations of both the multiple endocrine neoplasia type 1 (MEN1) and type 2 (MEN2) genes in a large Italian family and evaluate if it could be associated with more aggressive clinical manifestations of the two syndromes. Blood samples were obtained for genetic and biochemical analyses. The RET gene exons (8, 10, 11, 13, 14, 15, 16, 18) and the MEN1 coding regions, including the exon-intron boundaries, were amplified by PCR and directly sequenced. We identified two germline mutations in the proband: the first one, K666M, located at the exon 11 of RET proto-oncogene and the second one, IVS4+1G>T, located in the MEN1 gene. The functional characterization of IVS4+1G>T variation, located in the splicing donor site of exon 4 of MEN1 gene, caused the in-frame junction of exon 3 to exon 5, thus obtaining a shorter protein. The same proband's germline mutations were found in 16 relatives out of 21 screened subjects: 8 carried IVS4+1G>T, 4 RET K666M, and 4 both the mutations. This is the second report in literature of coexistence in the same family of germline mutations of both RET proto-oncogene and MEN1 gene. The simultaneous presence of the two mutations was not apparently associated with more aggressive diseases, since at last follow-up all patients appeared to be disease-free or well compensated by medical therapy; finally, no one exhibited metastatic diseases.
Collapse
Affiliation(s)
- Sandra Mastroianno
- Unit of Cardiology, Scientific Institute Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Oliveira MNL, Hemerly JP, Bastos AU, Tamanaha R, Latini FRM, Camacho CP, Impellizzeri A, Maciel RMB, Cerutti JM. The RET p.G533C mutation confers predisposition to multiple endocrine neoplasia type 2A in a Brazilian kindred and is able to induce a malignant phenotype in vitro and in vivo. Thyroid 2011; 21:975-85. [PMID: 21834681 DOI: 10.1089/thy.2010.0190] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND We have previously described a p.G533C substitution in the rearranged during transfection (RET) oncogene in a large family with medullary thyroid carcinoma. Here, we explore the functional transforming potential of RET p.G533C mutation. METHODS Plasmids expressing RET mutants (p.G533C and p.C634Y) and RET wild type were stable transfected into a rat thyroid cell line (PCCL3). Biological and biochemical effects of RET p.G533C were investigated both in vitro and in vivo. Moreover, we report the first case of pheochromocytoma among the RET p.G533C-carriers in this Brazilian family and explore the RET mutational status in DNA isolated from pheochromocytoma. RESULTS Ectopic expression of RET p.G533C and p.C634Y activates RET/MAPK/ERK pathway at similar levels and significantly increased cell proliferation, compared with RET wild type. We additionally show that p.G533C increased cell viability, anchorage-independent growth, and micronuclei formation while reducing apoptosis, hallmarks of the malignant phenotype. RET p.G533C down-regulates the expression of thyroid specific genes in PCCL3. Moreover, RET p.G533C-expressing cells were able to induce liver metastasis in nude mice. Finally, we described two novel RET variants (G548V and S556T) in the DNA isolated from pheochromocytoma while they were absent in the DNA isolated from blood. CONCLUSIONS Our in vitro and in vivo analysis indicates that this mutation confers a malignant phenotype to PCCL3 cells. These findings, in association with the report of first case of pheochromocytoma in the Brazilian kindred, suggest that this noncysteine mutation may be more aggressive than was initially considered.
Collapse
MESH Headings
- Adolescent
- Adrenal Gland Neoplasms/enzymology
- Adrenal Gland Neoplasms/genetics
- Adrenal Gland Neoplasms/pathology
- Analysis of Variance
- Animals
- Apoptosis
- Brazil
- Carcinoma, Medullary/congenital
- Cell Adhesion
- Cell Line
- Cell Proliferation
- Cell Survival
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Enzyme Activation
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Humans
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/secondary
- Male
- Mice
- Mice, Nude
- Micronuclei, Chromosome-Defective
- Middle Aged
- Multiple Endocrine Neoplasia Type 2a/enzymology
- Multiple Endocrine Neoplasia Type 2a/genetics
- Multiple Endocrine Neoplasia Type 2a/secondary
- Mutation
- Neoplastic Syndromes, Hereditary/enzymology
- Neoplastic Syndromes, Hereditary/genetics
- Pedigree
- Phenotype
- Pheochromocytoma/enzymology
- Pheochromocytoma/genetics
- Pheochromocytoma/pathology
- Phosphorylation
- Proto-Oncogene Proteins c-ret/genetics
- Proto-Oncogene Proteins c-ret/metabolism
- Rats
- Rats, Inbred F344
- Thyroid Gland/enzymology
- Thyroid Gland/pathology
- Thyroid Neoplasms/enzymology
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/secondary
- Transfection
Collapse
Affiliation(s)
- Mariana N L Oliveira
- Division of Genetics, Genetic Bases of Thyroid Tumors Laboratory, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Borrello MG, Aiello A, Peissel B, Rizzetti MG, Mondellini P, Degl'Innocenti D, Catalano V, Gobbo M, Collini P, Bongarzone I, Pierotti MA, Greco A, Seregni E. Functional characterization of the MTC-associated germline RET-K666E mutation: evidence of oncogenic potential enhanced by the G691S polymorphism. Endocr Relat Cancer 2011; 18:519-27. [PMID: 21690267 DOI: 10.1530/erc-10-0306] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Activating mutations of RET, a gene encoding two isoforms of a tyrosine kinase receptor physiologically expressed in several neural crest-derived cell lineages, are associated with the inherited forms of medullary thyroid carcinoma (MTC). The identification and characterization of novel RET mutations involved in MTC is valuable, as RET gene testing plays a crucial role in the management of these patients. In an MTC patient, we have identified a germline c.1996A>G transition in heterozygosis leading to K666E substitution. In addition, the conservative S904S (c.2712C>G) and the non-conservative functional G691S (c.2071G>A) polymorphisms have been identified. Through functional studies, we demonstrate for the first time that K666E is a gain-of-function mutation with oncogenic potential, based on its ability to transform NIH3T3 cells. It was not possible to define whether K666E is a de novo or inherited RET variant in the patient, as the family history was negative for MTC, and the carrier status of family members could not be tested. Our results, together with a recent report of co-segregation of the mutation in three MTC families, suggest that K666E is a causative MTC mutation. As we have shown that the same patient allele carries both K666E and G691S variants, the latter known to increase downstream RET signaling, a possible role for the G691S polymorphism has also been investigated. We have demonstrated that, although RET-G691S is not oncogenic per se, it enhances the transforming activity of the RET-K666E mutant, thus suggesting a modifier role for this functional polymorphism.
Collapse
Affiliation(s)
- Maria Grazia Borrello
- Operative Unit Molecular Mechanisms, Department of Experimental Oncology, IRCCS Istituto Nazionale dei Tumori Foundation, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Prazeres H, Couto JP, Rodrigues F, Vinagre J, Torres J, Trovisco V, Martins TC, Sobrinho-Simões M, Soares P. In vitro transforming potential, intracellular signaling properties, and sensitivity to a kinase inhibitor (sorafenib) of RET proto-oncogene variants Glu511Lys, Ser649Leu, and Arg886Trp. Endocr Relat Cancer 2011; 18:401-12. [PMID: 21551259 DOI: 10.1530/erc-10-0258] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Multiple endocrine neoplasia type 2 and a subset of apparently sporadic medullary thyroid carcinoma (AS-MTC) are caused by germ line activating point mutations of the rearranged during transfection (RET) proto-oncogene. RET encodes a receptor with tyrosine kinase activity that targets several intracellular signaling cascades, such as RAS-RAF-ERK1/2, PIK3-AKT, and STAT transcription factors. The objective of this study was to assess the function of three germ line RET variants Arg886Trp, Ser649Leu, and Glu511Lys of undetermined pathogenic significance, which were found in three kindreds of isolated AS-MTC. For this purpose, we employed vectors expressing each of the RET variants and measured the number of NIH3T3 transformation foci and soft agar colonies, the degree of activation of known RET intracellular signaling targets (ERK1/2, STAT1, STAT3, and TCF4), and the extent of ERK1/2 inhibition on sorafenib treatment. We found that RET variants Arg886Trp and Glu511Lys have shown increased in vitro transforming potential in a glial-derived neurotrophic factor-dependent manner. In contrast, the Ser649Leu variant did not significantly increased the number of foci and agar colonies relative to wild-type RET (RET-WT). The variants Glu511Lys and Arg886Trp showed 10- and 12.5-fold ERK1/2 activation respectively, that was significantly higher than that observed for RET-WT (fivefold). Increased levels of STAT1 and TCF4 activation were only observed for RET Arg886Trp (2.5- and 3-fold versus 1.2- and 2-fold in RET-WT respectively). The three RET variants analyzed here were sensitive to treatment with sorafenib. In conclusion, our results allow to classify previously uncharacterized RET genotypes, which may be of use to define follow-up and therapeutic regimens.
Collapse
Affiliation(s)
- Hugo Prazeres
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) - Cancer Biology, Rua Dr Roberto Frias, Porto, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rohmer V, Vidal-Trecan G, Bourdelot A, Niccoli P, Murat A, Wemeau JL, Borson-Chazot F, Schvartz C, Tabarin A, Chabre O, Chabrier G, Caron P, Rodien P, Schlumberger M, Baudin E. Prognostic factors of disease-free survival after thyroidectomy in 170 young patients with a RET germline mutation: a multicenter study of the Groupe Francais d'Etude des Tumeurs Endocrines. J Clin Endocrinol Metab 2011; 96:E509-18. [PMID: 21190982 DOI: 10.1210/jc.2010-1234] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND In hereditary medullary thyroid carcinoma (HMTC), prophylactic surgery is the only curative option, which should be properly defined both in time and extent. OBJECTIVES To identify and characterize prognostic factors associated with disease-free survival (DFS) in children from HMTC families. DESIGN We conducted a retrospective analysis of a multi-center cohort of 170 patients below age 21 at surgery. Demographic, clinical, genetic, biological data [basal and pentagastrine-stimulated calcitonin (CT and CT/Pg, respectively)], and tumor node metastasis (TNM) status were collected. DFS was assessed based on basal CT levels. Kaplan-Meier curves, Cox regression, and logistic regression models were used to determine factors associated with DFS and TNM staging. RESULTS No patients with a preoperative basal CT <31 ng/ml had persistent or recurrent disease. Medullary thyroid carcinoma defined by a diameter ≥10 mm [hazard ratio (HR): 6.0; 95% confidence interval (95% CI): 1.8-19.8] and N1 status (HR: 20.8; 95% CI: 3.9-109.8) were independently associated with DFS. Class D genotype [odds ratio (OR): 48.5, 95% CI: 10.6-225.1], preoperative basal CT >30 ng/liter (OR: 43.4, 95% CI: 5.2-359.8), and age >10 (OR: 5.5, 95% CI: 1.4-21.8) were associated with medullary thyroid carcinoma ≥10 mm. No patient with a preoperative basal CT <31 ng/ml had a N1 status. Class D genotype (OR: 48.6, 95% CI: 8.6-274.1), and age >10 (OR: 4.6, 95% CI: 1.1-19.0) were associated with N1 status. CONCLUSION In HMTC patients, DFS is best predicted by TNM staging and preoperative basal CT level below 30 pg/ml. Basal CT, class D genotype, and age constitute key determinants to decide preoperatively timely surgery.
Collapse
Affiliation(s)
- V Rohmer
- Centre hospitalier universitaire Angers, Endocrinologie, Faculté de médecine, Université Angers, Angers 49933, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|