Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
J Nucl Med ; 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38991747

RESUMO

High-activity radioactive iodine (RAI) therapy for metastatic thyroid cancer (TC) requires isolation to minimize radiation exposure to third parties, thus posing challenges for patients needing hands-on care. There are limited data on the approach to high-activity RAI treatment in paraplegic patients. We report a state-of-the-art multidisciplinary approach to the management of bedbound patients, covering necessary radiation safety measures that lead to radiation exposure levels as low as reasonably achievable. Given the limited literature resources on standardized approaches, we provide a practical example of the safe and successful treatment of a woman with BRAFV600E-mutant tall-cell-variant papillary TC and pulmonary metastases, who underwent dabrafenib redifferentiation before RAI therapy. The patient was 69 y old and had become paraplegic because of a motor-vehicle accident. Since caring for a paraplegic patient with neurogenic bowel and bladder dysfunction poses radiation safety challenges, a multidisciplinary team comprising endocrinologists, nuclear medicine physicians, radiation safety specialists, and the nursing department developed a radiation mitigation strategy to ensure patient and staff safety during RAI therapy. The proposed standardized approach includes thorough monitoring of radiation levels in the workplace, providing additional protective equipment for workers who handle radioactive materials or are in direct patient contact, and implementing strict guidelines for safely disposing of radioactive waste such as urine collected in lead-lined containers. This approach requires enhanced training, role preparation, and practice; use of physical therapy equipment to increase the exposure distance; and estimation of the safe exposure time for caregivers based on dosimetry. The effective and safe treatment of metastatic TC in paraplegic patients can be successfully implemented with a comprehensive radiation mitigation strategy and thorough surveying of personnel for contamination.

2.
Thyroid ; 34(6): 674-686, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38717947

RESUMO

Background: Long-term management of intermediate- and high-risk differentiated thyroid cancer (DTC) involves thyrotropin (TSH) suppression with thyroid hormone to prevent potential stimulation of TSH receptors on DTC cells, leading to tumor growth. However, the current guidelines recommending TSH suppression are based on low- to moderate-quality evidence. Methods: We performed a systematic review and meta-analysis of studies evaluating the role of TSH suppression in intermediate- and high-risk DTC patients (≥18 years) treated as per regional guideline-based therapy with a follow-up duration of 5 years (PROSPERO #252396). TSH suppression was defined as "below normal reference range" or, when known, <0.5 mIU/L. Primary outcome measures included (i) composite of progression-free survival (PFS), disease-free survival (DFS), and relapse-free survival (RLFS), and (ii) composite of disease-specific survival (DSS), and overall survival (OS). Secondary outcome included a composite of cardiac or skeletal adverse events. All outcomes and comparisons were represented as TSH suppression versus TSH nonsuppression. Randomized controlled trials, cohort studies, and case-control studies were included for analysis. Pooled hazard ratio (HR) and 95% confidence interval (CI) were calculated using random-effects model. Results: Abstract screening was performed on 6,369 studies. After the exclusion of irrelevant studies and full-text screening, nine studies were selected for the final meta-analysis. Based on seven studies (3,591 patients), the composite outcome of PFS, DFS, and RLFS was not significantly different between TSH suppression and nonsuppression groups (HR: 0.75; 95% CI: 0.48-1.17; I2 = 76%). Similarly, a DSS and OS composite outcome assessment based on four studies (3,616 patients) did not favor TSH suppression (HR: 0.69; 95% CI: 0.31-1.52; I2 = 88%). Even after excluding studies of lower quality, the primary outcomes were not significantly different between the TSH suppression and nonsuppression cohorts. The secondary outcome, obtained from two studies (1,294 patients), was significantly higher in the TSH-suppressed groups (HR: 1.82; 95% CI: 1.30-2.55; I2 = 0%). Significant study heterogeneity was noted for primary outcomes. Conclusion: TSH suppression in intermediate- and high-risk DTC may not improve survival outcomes but may increase the risk of secondary complications. However, the limited evidence and study heterogeneity warrant cautious interpretation of our findings. Registration: PROSPERO #252396.


Assuntos
Neoplasias da Glândula Tireoide , Tireotropina , Humanos , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/mortalidade , Neoplasias da Glândula Tireoide/patologia , Tireotropina/sangue , Intervalo Livre de Doença , Intervalo Livre de Progressão , Resultado do Tratamento
3.
Artigo em Inglês | MEDLINE | ID: mdl-38421044

RESUMO

CONTEXT: Thyroid-stimulating hormone (or thyrotropin) receptor (TSHR) could be a selective target for small molecule ligands to treat thyroid cancer (TC). OBJECTIVE: We report a novel, orally efficacious ligand for TSHR that exhibits proliferation inhibitory activity against human TC in vitro and in vivo, and inhibition of metastasis in vivo. DESIGN: A35 (NCATS-SM4420; NCGC00241808) was selected from a sub-library of >200 TSHR ligands. Cell proliferation assays including BrdU incorporation and WST-1, along with molecular docking studies were done. In vivo activity of A35 was assessed in TC cell-derived xenograft (CDX) models with immunocompromised (NSG) mice. FFPE sections of tumor and lung tissues were observed for the extent of cell death and metastasis. RESULTS: A35 was shown to stimulate cAMP production in some cell types by activating TSHR but not in TC cells, MDA-T32 and MDA-T85. A35 inhibited proliferation of MDA-T32 & MDA-T85 in vitro and in vivo, and pulmonary metastasis of MDA-T85F1 in mice. In vitro, A35 inhibition of proliferation was reduced by a selective TSHR antagonist. Inhibition of CDX tumor growth without decreases in mouse weights and liver function showed A35 to be efficacious without apparent toxicity. Lastly, A35 reduced levels of Ki67 in the tumors and metastatic markers in lung tissues. CONCLUSION: We conclude that A35 is a TSHR-selective inhibitor of TC cell proliferation and metastasis, and suggest that A35 may be a promising lead drug candidate for the treatment of differentiated thyroid cancer in humans.

4.
Front Endocrinol (Lausanne) ; 14: 1275813, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37886645

RESUMO

Purpose: While there are reports of treatment-related endocrine disruptions and catecholamine surges in pheochromocytoma/paraganglioma (PPGL) patients treated with [177Lu]Lu-DOTA-TATE therapy, the spectrum of these abnormalities in the immediate post-treatment period (within 48 hours) has not been previously evaluated and is likely underestimated. Methods: The study population included patients (≥18 years) enrolled in a phase 2 trial for treatment of somatostatin receptor (SSTR)-2+ inoperable/metastatic pheochromocytoma/paraganglioma with [177Lu]Lu-DOTA-TATE (7.4 GBq per cycle for 1 - 4 cycles). Hormonal measurements [adrenocorticotropic hormone (ACTH), cortisol, thyroid stimulating hormone (TSH), free thyroxine (FT4), follicle stimulating hormone (FSH), luteinizing hormone (LH), testosterone, estradiol, growth hormone, prolactin], catecholamines, and metanephrines were obtained on days-1, 2, 3, 30, and 60 per cycle as per trial protocol, and were retrospectively analyzed. Results: Among the 27 patients (age: 54 ± 12.7 years, 48.1% females) who underwent hormonal evaluation, hypoprolactinemia (14.1%), elevated FSH (13.1%), and elevated LH (12.5%) were the most frequent hormonal abnormalities across all 4 cycles combined. On longitudinal follow-up, significant reductions were noted in i. ACTH without corresponding changes in cortisol, ii. TSH, and FT4, and iii. prolactin at or before day-30 of [177Lu]Lu-DOTA-TATE. No significant changes were observed in the gonadotropic axis and GH levels. Levels of all hormones on day-60 were not significantly different from day-1 values, suggesting the transient nature of these changes. However, two patients developed clinical, persistent endocrinopathies (primary hypothyroidism: n=1 male; early menopause: n=1 female). Compared to day-1, a significant % increase in norepinephrine, dopamine, and normetanephrine levels were noted at 24 hours following [177Lu]Lu-DOTA-TATE dose and peaked within 48 hours. Conclusions: [177Lu]Lu-DOTA-TATE therapy is associated with alterations in endocrine function likely from radiation exposure to SSTR2+ endocrine tissues. However, these changes may sometimes manifest as clinically significant endocrinopathies. It is therefore important to periodically assess endocrine function during [177Lu]Lu-DOTA-TATE therapy, especially among symptomatic patients. Clinical trial registration: https://clinicaltrials.gov/ct2/show/NCT03206060?term=NCT03206060&draw=2&rank=1, identifier NCT03206060.


Assuntos
Neoplasias das Glândulas Suprarrenais , Feocromocitoma , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Idoso , Feocromocitoma/radioterapia , Estudos Retrospectivos , Prolactina , Hidrocortisona , Hormônio Adrenocorticotrópico , Hormônio Foliculoestimulante , Catecolaminas , Tireotropina
6.
Front Pharmacol ; 13: 791710, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36249761

RESUMO

Objective: A low-iodine diet (LID) of <50µ iodine/day is recommended as preparation for radioactive iodine (RAI) therapy in patients with differentiated thyroid cancer (DTC). The 24-h urinary iodine excretion (UIE) is utilized to evaluate the iodine-depleted status. The aim of this study was to test the association between UIE and progression-free survival (PFS). Patients and methods: In total, 70 patients with intermediate- or high-risk DTC, post-total thyroidectomy, adhered to 2 weeks of LID and had UIE measured before RAI therapy. A Cox regression model was performed to study the contribution of UIE to PFS. Results: The study group consisted of 68% (48/70) of women, aged 41.5 [IQR 31.0, 54.0] years, with tumor size 2.8 [IQR 1.8-4.5] cm, and presence of distant metastases in 22.9% (16/70) of patients. Patients were treated with 1-5 RAI dosages with the median cumulative activity of 150 [IQR 102-314] mCi (5.5 [IQR 3.8-11.6] GBq). During the follow-up of 3.7 [IQR 1.5-6.5] years, 21.4% (15/70) of patients had disease progression. The risk of progression was significantly higher in patients with UIE ≥200 µg/day at the time of RAI administration than in those with UIE <200 µg/day (HR 3.35, 95% CI 1.09-10.34, and p = 0.02). However, the multivariate Cox proportional hazards regression analysis adjusted for age, tumor size, and presence of distant metastases suggested that only distant metastases were independently significantly associated with the risk of progression (HR 5.80 (1.17-28.67), p = 0.03). Conclusions: Although UIE ≥200 µg/day might be associated with worse PFS in RAI-treated DTC patients, the presence of distant metastases is a strong independent predictor of progression. Less stringent LID might be sufficient to achieve a UIE of <200 µg/day.

7.
Horm Metab Res ; 54(12): 795-812, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35878617

RESUMO

Neuroendocrine tumors (NETs) occur in various regions of the body and present with complex clinical and biochemical phenotypes. The molecular underpinnings that give rise to such varied manifestations have not been completely deciphered. The management of neuroendocrine tumors (NETs) involves surgery, locoregional therapy, and/or systemic therapy. Several forms of systemic therapy, including platinum-based chemotherapy, temozolomide/capecitabine, tyrosine kinase inhibitors, mTOR inhibitors, and peptide receptor radionuclide therapy have been extensively studied and implemented in the treatment of NETs. However, the potential of immune checkpoint inhibitor (ICI) therapy as an option in the management of NETs has only recently garnered attention. Till date, it is not clear whether ICI therapy holds any distinctive advantage in terms of efficacy or safety when compared to other available systemic therapies for NETs. Identifying the characteristics of NETs that would make them (better) respond to ICIs has been challenging. This review provides a summary of the current evidence on the value of ICI therapy in the management of ICIs and discusses the potential areas for future research.


Assuntos
Tumores Neuroendócrinos , Humanos , Tumores Neuroendócrinos/tratamento farmacológico , Inibidores de Checkpoint Imunológico/uso terapêutico
8.
Front Endocrinol (Lausanne) ; 13: 896287, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35712243

RESUMO

The treatment options that are currently available for management of metastatic, progressive radioactive iodine (RAI)-refractory differentiated thyroid cancers (DTCs), and medullary thyroid cancers (MTCs) are limited. While there are several systemic targeted therapies, such as tyrosine kinase inhibitors, that are being evaluated and implemented in the treatment of these cancers, such therapies are associated with serious, sometimes life-threatening, adverse events. Peptide receptor radionuclide therapy (PRRT) has the potential to be an effective and safe modality for treating patients with somatostatin receptor (SSTR)+ RAI-refractory DTCs and MTCs. MTCs and certain sub-types of RAI-refractory DTCs, such as Hürthle cell cancers which are less responsive to conventional modalities of treatment, have demonstrated a favorable response to treatment with PRRT. While the current literature offers hope for utilization of PRRT in thyroid cancer, several areas of this field remain to be investigated further, especially head-to-head comparisons with other systemic targeted therapies. In this review, we provide a comprehensive outlook on the current translational and clinical data on the use of various PRRTs, including diagnostic utility of somatostatin analogs, theranostic properties of PRRT, and the potential areas for future research.


Assuntos
Carcinoma Neuroendócrino , Neoplasias da Glândula Tireoide , Carcinoma Neuroendócrino/patologia , Humanos , Radioisótopos do Iodo/uso terapêutico , Receptores de Somatostatina/uso terapêutico , Neoplasias da Glândula Tireoide/patologia
9.
Pol Arch Intern Med ; 132(3)2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35243857

RESUMO

Hashimoto thyroiditis (HT) is a common autoimmune disorder, affecting women 7-10 times more often than men, that develops because of genetic susceptibility, Xchromosome inactivation patterns modulated by environmental factors as well as microbiome composition, and leads to an imbalance in self­tolerance mechanisms. The consequential thyroid infiltration by lymphocytes, potentiated by antibody­mediated autoimmune response through the antibodies against thyroid peroxidase (TPOAbs), leads to a destruction of thyrocytes. The presence of TPOAbs is associated with a 2 to 4­fold increase in the risk of recurrent miscarriages and preterm birth in pregnant women. The clinical presentation of HT includes: (A) thyrotoxicosis, when stored thyroid hormones are released to circulation from destroyed thyroid follicles; (B) euthyroidism, when preserved thyroid tissue compensates for destroyed thyrocytes; and (C) hypothyroidism, when thyroid hormone production by the affected thyroid gland is insufficient. The management of Hashitoxicosis is based on symptoms control usually with ß­blockers, euthyroidism requires periodical thyroid stimulating hormone measurements to assess for progression to hypothyroidism, and hypothyroidism is treated with thyroid hormone replacement therapy. The dose of levothyroxine (LT4) used for treatment is based on the degree of preserved thyroid functionality and lean body mass, and usually ranges from 1.4 to 1.8 mcg/kg/day. There is insufficient evidence to recommend for or against therapy with triiodothyronine (T3), apart from in pregnancy when only levothyroxine is indicated, as T3 does not sufficiently cross fetal blood­brain barrier. HT is associated with 1.6 times higher risk of papillary thyroid cancer and 60 times higher risk of thyroid lymphoma than in general the population.


Assuntos
Doença de Hashimoto , Hipotireoidismo , Nascimento Prematuro , Feminino , Doença de Hashimoto/diagnóstico , Doença de Hashimoto/terapia , Humanos , Recém-Nascido , Masculino , Gravidez , Nascimento Prematuro/tratamento farmacológico , Hormônios Tireóideos , Tiroxina/uso terapêutico , Tri-Iodotironina/uso terapêutico
10.
J Endocr Soc ; 6(5): bvac032, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35356009

RESUMO

Context: Recombinant human thyrotropin (rhTSH) is currently not Food and Drug Administration approved for the treatment of high-risk patients with differentiated thyroid cancer (DTC). Objective: The goal of our study was to compare the outcomes in higher-risk patients with metastatic DTC prepared for radioiodine (RAI) therapy with rhTSH vs thyroid hormone withdrawal (THW). Methods: A retrospective chart review was performed of patients with metastatic DTC in follow-up at MedStar Washington Hospital Center and MedStar Georgetown University Hospital from 2009 to 2017. Patients were divided according to their preparation for RAI therapy, with assessment of progression-free survival (PFS) and overall survival (OS). Results: Fifty-five patients with distant metastases (16 men, 39 women) were prepared for RAI therapy exclusively either with rhTSH (n = 27) or with THW (n = 28). There were no statistically significant differences between the groups regarding clinicopathological features and history of RAI therapies. The median follow-up time for patients with rhTSH-aided therapies was 4.2 years (range, 3.3-5.5 years) and for patients with THW-aided therapies was 6.8 years (range, 4.2-11.6 years) (P = .002). Multivariate analysis showed that the method of thyrotropin stimulation was not associated with a difference in PFS or OS. Conclusion: As has been shown previously for low-risk DTC, this study indicates that the mode of preparation for RAI therapy does not appear to influence the outcomes of patients with metastatic DTC. PFS and OS were similar for patients with THW-aided or rhTSH-aided RAI therapies.

11.
Cytopathology ; 33(3): 344-349, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34957617

RESUMO

INTRODUCTION: Molecular testing for genetic alterations in thyroid neoplasms, including BRAF V600E (BRAF) mutation, are often applied to thyroid aspirates falling into the Bethesda System for Reporting Thyroid Cytopathology indeterminate categories. Current methods typically use dedicated aspirated material, without morphological determination of containing the cells of interest and may be of elevated cost. We describe our experience with BRAF mutation analysis on material obtained from Papanicolaou (PAP)-stained ThinPrep® (TP) slides. METHODS: Eighty-three cases collected between 2012 and 2019 with more than 100 cells were selected. An electronic record of a whole slide scan was made for each case before testing. The coverslips were removed, and DNA was extracted from material scraped from each slide using the Qiagen QIAamp DNA FFPE Tissue Kit. BRAF testing was performed using a highly sensitive mutation detection assay, either COLD-PCR, castPCR, or droplet digital PCR. RESULTS: Fourteen out of 83 cases had a BRAF mutation. Of these, 8 were classified as atypia of undetermined significance or suspicious for malignancy in which follow-up showed conventional papillary thyroid carcinoma in 5 out of 6 cases. The specificity and positive predictive value were 97% and 91%, respectively. CONCLUSIONS: BRAF mutation analysis can be performed on material obtained from routine clinical PAP-stained TP slides. As a first step, this unconventional effective approach may reduce costs related to the molecular evaluation of thyroid nodule aspirates and provides the opportunity for cytomorphological confirmation that the cells of interest are present in material submitted for BRAF mutation analysis.


Assuntos
Neoplasias da Glândula Tireoide , Nódulo da Glândula Tireoide , Análise Mutacional de DNA , Humanos , Mutação/genética , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias da Glândula Tireoide/diagnóstico , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Nódulo da Glândula Tireoide/patologia
12.
Clin Thyroidol ; 34(7): 286-289, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36937987

RESUMO

Background: Thyroid disorders have been associated with breast cancer. In fact, breast cancer is the most common secondary malignancy in female patients with thyroid cancer (1). Moreover, hyperthyroidism is associated with an 11% increased risk of breast cancer in women (2). Importantly, up to 30% of patients with breast cancer are treated with thyroid hormone replacement therapy (THRT) for overt or subclinical hypothyroidism (3). These observations, coupled with the preclinical data showing growth stimulatory effects of thyroid hormones (THs) in various cancer models (4), formed the rationale for the study by Wahdan-Alaswad et al. aimed at investigating the role of THRT on the outcome of patients with nonmetastatic breast cancer (3). Methods: The authors conducted an observational study analyzing the association between THRT, disease-free survival (DFS), and disease-specific survival (DSS) in two cohorts of patients with nonmetastatic breast cancer. The first cohort consistent of 820 patients followed for a median of 10 years and treated for breast cancer between 1962 and1993, with THRT implemented in 69 patients. The second cohort included 160 patients treated more recently (between 2006 and 2009) and followed for a median of 8.8 years, with 50 patients exposed to THRT. The data on the age, tumor size, presence or absence of steroid (estrogen and/or progesterone) receptors (SR+/SR-), and treatment regimen were incorporated in the multivariate model analyzing the association between DFS/DSS and THRT at 5 and 10 years. To better understand the results of the observational cohort study, the authors performed functional in vitro and in vivo experiments to investigate the molecular mechanisms underlying TH effects on breast cancer cells and to test the interactions between estrogen receptors (ERs) and TH receptors (THRs). Results: In patients with SR+ breast cancer, THRT was associated with a significantly increased risk of recurrence (DFS RR, 2.9; P<0.001) and death (DSS RR, 3.4; P<0.001), independent of age, tumor size and grade, while THRT in patients with SR- breast cancer was not associated with worse outcomes. Moreover, patients with SR+ breast cancer undergoing therapy with aromatase inhibitor combined with THRT were characterized by a shorter DFS (P<0.042) and a higher 10-year recurrence rate of 14%, as compared with 2% in patient treated with the aromatase inhibitor alone.The functional in vitro and in vivo studies revealed growth stimulatory effects of monotherapy with TH or estrogens that were further potentiated with combination therapy in ER+ breast cancer cell lines and mice xenografts. The RNA-Seq analysis revealed that combination therapy was associated with a significant activation of the cell cycle, mismatch repair, homologous recombination, and DNA replication signaling, as well as induced thyroid-specific genes and estrogen-mediated signatures. These effects were abrogated by the knockdown or inhibition of ER and/or THRa, suggesting that cross-talk and nuclear colocalization of ERs and THRs are major drivers of pro-oncogenic signaling in the ER+ breast cancer model. Conclusions: The study reveals clinically significant associations between THRT and worse outcomes in patients with nonmetastatic SR+ breast cancer that are likely driven by interactions between the nuclear ERs and THRs, leading to upregulation of pro-oncogenic signaling. These results suggest that overuse of THRT in patients with hypothyroidism and concurrent breast cancer should be avoided.

13.
Clin Thyroidol ; 34(4): 161-164, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36969797

RESUMO

Background: Exposure to excess ionizing radiation has been identified as a risk factor for the development of thyroid cancer (1). However, there are no well-established biomarkers indicating exposure to radiation as the etiology of thyroid tumors that can be applied in clinical care or in legal claims. Morton et al. analyzed a large cohort of individuals who were exposed to a power plant accident in Chernobyl in 1986 in their childhood and who subsequently developed papillary thyroid cancer (PTC) (2). The goal of the study was to enhance our understanding of radiation-induced carcinogenesis based on assessment of the molecular drivers, transcriptomics, and epigenetic profile of PTC associated with excessive environmental radiation exposure. Methods: The authors performed whole-genome sequencing, single-nucleotide polymorphism (SNP) microarray genotyping, mRNA and microRNA sequencing, methylation profiling, transcriptome analysis, and telomere-length quantification in 440 pathologically confirmed fresh-frozen PTC tissue samples for which matched normal tissue from either nontumor thyroid and/or blood was available. In the study group, 359 subjects were exposed to excess ionizing radiation in childhood or in utero; 81 individuals who were born more than 9 months after the Chernobyl accident served as a reference group. The data were analyzed adjusting for covariates that potentially affect PTC incidence, including sex, age at diagnosis, age at radiation exposure, and latency (defined as the time from exposure to PTC diagnosis). Results: The median age at exposure to ionizing radiation was 7.3 years and the median latency before the diagnosis of PTC was 22.4 years. The exposure estimates (250 mGy on average and up to 8800 mGy) were based on direct measurements of the thyroid-absorbed dose within 8 weeks after the accident in 53 individuals, while for the remaining cohort, they were imputed from direct measurements in individuals living in a similar area.The molecular drivers were identified in the vast majority of the tumors (98.4%), mainly as a low mutation burden consisting of single candidates in the mitogen-activated protein kinase (MAPK) pathway. The most common driver was the BRAF V600E mutation, but fusions in RET proto-oncogene, receptor tyrosine kinase (RTK), such as NTRK1, NTRK3, ALK, as well as BRAF, PPRAG, and IGF2/IGF2BP3 accounted for the majority of the remaining drivers (41% of the tumors). Moreover, there was a significant association between fusion drivers and radiation dose, after adjustment for age at diagnosis and sex. Same significant association with radiation was observed for small deletions and balanced structural variants resulting from the nonhomologous end-joining repair of double-stranded DNA damage. In contrast, transcriptome, methylome, or telomere length were not significantly associated with the radiation dose, and all tumors were microsatellite-stable. No novel molecular signature unique for radiation-associated PTC has been identified. Conclusions: The study reveals potential mechanisms behind radiation-associated PTC, consisting of DNA double-stranded breaks leading to nonhomologous end-joining repair mechanisms, that result in pathogenic gene fusions responsible for clonal growth. There is no unique signature of radiation-associated PTC that could serve as a biomarker of radiation-induced malignancy.

14.
Thyroid ; 32(1): 90-96, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34714162

RESUMO

Background: The pathogenesis of Graves' hyperthyroidism (GH) and associated Graves' orbitopathy (GO) appears to involve stimulatory autoantibodies (thyrotropin receptor [TSHR]-stimulating antibodies [TSAbs]) that bind to and activate TSHRs on thyrocytes and orbital fibroblasts. In general, measurement of circulating TSHR antibodies by clinical assays correlates with the status of GH and GO. However, most clinical measurements of TSHR antibodies use competitive binding assays that do not distinguish between TSAbs and antibodies that bind to but do not activate TSHRs. Moreover, clinical assays for TSAbs measure stimulation of only one signaling pathway, the cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) pathway, in engineered cells that are not thyrocytes or orbital fibroblasts. We determined whether measuring TSAbs by a cAMP-PKA readout in engineered cells accurately reveals the efficacies of stimulation by these antibodies on thyrocytes and orbital fibroblasts. Methods: We measured TSAb stimulation of normal human thyrocytes and orbital fibroblasts from patients with GO in primary cultures in vitro. In thyrocytes, we measured secretion of thyroglobulin (TG) and in orbital fibroblasts secretion of hyaluronan (hyaluronic acid [HA]). We also measured stimulation of cAMP production in engineered TSHR-expressing cells in an assay similar to clinical assays. Furthermore, we determined whether there were differences in stimulation of thyrocytes and orbital fibroblasts by TSAbs from patients with GH alone versus from patients with GO understanding that patients with GO have accompanying GH. Results: We found a positive correlation between TSAb stimulation of cAMP production in engineered cells and TG secretion by thyrocytes as well as HA secretion by orbital fibroblasts. However, TSAbs from GH patients stimulated thyrocytes more effectively than TSAbs from GO patients, whereas TSAbs from GO patients were more effective in activating orbital fibroblasts than TSAbs from GH patients. Conclusions: Clinical assays of stimulation by TSAbs measuring activation of the cAMP-PKA pathway do correlate with stimulation of thyrocytes and orbital fibroblasts; however, they do not distinguish between TSAbs from GH and GO patients. In vitro, TSAbs exhibit selectivity in activating TSHRs since TSAbs from GO patients were more effective in stimulating orbital fibroblasts and TSAbs from GH patients were more effective in stimulating thyrocytes.


Assuntos
Autoanticorpos/imunologia , Fibroblastos/imunologia , Oftalmopatia de Graves/complicações , Células Epiteliais da Tireoide/imunologia , Adulto , Autoanticorpos/análise , Feminino , Fibroblastos/metabolismo , Doença de Graves/sangue , Doença de Graves/imunologia , Oftalmopatia de Graves/sangue , Oftalmopatia de Graves/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Células Epiteliais da Tireoide/metabolismo , Tireotropina/metabolismo
15.
Front Endocrinol (Lausanne) ; 12: 653401, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34326811

RESUMO

Familial non-medullary thyroid cancer (FNMTC) is a form of endocrine malignancy exhibiting an autosomal dominant mode of inheritance with largely unknown germline molecular mechanism. Hereditary nonpolyposis colorectal cancer syndrome (HNPCC) is another hereditary autosomal dominant cancer syndrome which, if proven to be caused by germline mutations in mismatch repair genes (MMR)-MLHL, MSH2, MSH6, PMS2, and EPCAM-is called Lynch syndrome (LS). LS results in hereditary predisposition to a number of cancers, especially colorectal and endometrial cancers. Tumors in LS are characterized by microsatellite instability (MSI) and/or loss of MMR protein expression in immunohistochemistry (IHC). MSI is a rare event in thyroid cancer (TC), although it is known to occur in up to 2.5% of sporadic follicular TC cases. There are limited data on the role of germline MMR variants FNMTC. The goal of this study was to analyze the potential clinical and molecular association between HNPCC and FNMTC. We performed a cohort study analyzing the demographic, clinical, and pathologic data of 43 kindreds encompassing 383 participants (104 affected, 279 unaffected), aged 43.5 [7-99] years with FNMTC, and performed high-throughput whole-exome sequencing (WES) of peripheral blood DNA samples of selected 168 participants (54 affected by FNMTC and 114 unaffected). Total affected by thyroid cancer members per family ranged between 2 and 9 patients. FNMTC was more prevalent in women (68.3%) and characterized by a median tumor size of 1.0 [0.2-5.0] cm, multifocal growth in 44%, and gross extrathyroidal extension in 11.3%. Central neck lymph node metastases were found in 40.3% of patients at presentation, 12.9% presented with lateral neck lymph node metastases, and none had distant metastases. Family history screening revealed one Caucasian family meeting the clinical criteria for FNMTC and HNPCC, with five members affected by FNMTC and at least eight individuals reportedly unaffected by HNPCC-associated tumors. In addition, two family members were affected by melanoma. Genome Analysis Tool Kit (GATK) pipeline was used in variant analysis. Among 168 sequenced participants, a heterozygous missense variant in the MSH2 gene (rs373226409; c.2120G>A; p.Cys707Tyr) was detected exclusively in FNMTC- HNPCC- kindred. In this family, the sequencing was performed in one member affected by FNMTC, HPNCC-associated tumors and melanoma, one member affected solely by HNPCC-associated tumor, and one member with FNMTC only, as well as seven unaffected family members. The variant was present in all three affected adults, and in two unaffected children of the affected member, under the age of 18 years, and was absent in non-affected adults. This variant is predicted to be damaging/pathogenic in 17/20 in-silico models. However, immunostaining performed on the thyroid tumor tissue of two affected by FNMTC family members revealed intact nuclear expression of MSH2, and microsatellite stable status in both tumors that were tested. Although the MSH2 p.Cys707Tyr variant is rare with a minor allele frequency (MAF) of 0.00006 in Caucasians; it is more common in the South Asian population at 0.003 MAF. Therefore, the MSH2 variant observed in this family is unlikely to be an etiologic factor of thyroid cancer and a common genetic association between FNMTC and HNPCC has not yet been identified. This is the first report known to us on the co-occurrence of FNMTC and HNPCC. The co-occurrence of FNMTC and HNPCC-associated tumors is a rare event and although presented in a single family in our large FNMTC cohort, a common genetic background between the two comorbidities could not be established.


Assuntos
Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Câncer Papilífero da Tireoide/diagnóstico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Estudos de Coortes , Neoplasias Colorretais Hereditárias sem Polipose/complicações , Neoplasias Colorretais Hereditárias sem Polipose/genética , Comorbidade , Técnicas Citológicas , Reparo de Erro de Pareamento de DNA , Exoma , Saúde da Família , Feminino , Predisposição Genética para Doença , Variação Genética , Mutação em Linhagem Germinativa , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Linhagem , Estudos Prospectivos , Proteínas Proto-Oncogênicas B-raf/genética , Câncer Papilífero da Tireoide/complicações , Câncer Papilífero da Tireoide/genética , Neoplasias da Glândula Tireoide/complicações , Neoplasias da Glândula Tireoide/genética , Adulto Jovem
17.
Thyroid ; 31(8): 1160-1170, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34042535

RESUMO

Background: Biotin has been reported to interfere with several commonly used laboratory assays resulting in misleading values and possible erroneous diagnosis and treatment. This report describes a prospective study of possible biotin interference in thyroid-related laboratory assays, with a comparison of different commonly used assay platforms. Materials and Methods: Thirteen adult subjects (mean age 45 ± 13 years old) were administered biotin 10 mg/day for eight days. Blood specimens were collected at three time points on day 1 and on day 8 (baseline, two, and five hours after biotin ingestion). Thyrotropin (TSH), free triiodothyronine (fT3), free thyroxine (fT4), total triiodothyronine (TT3), total thyroxine (TT4), thyroxine binding globulin (TBG), and thyroglobulin (Tg) levels were analyzed with four different platforms: Abbott Architect, Roche Cobas 6000, Siemens IMMULITE 2000, and liquid chromatography with tandem mass spectrometry (LC-MS/MS). TSH, fT3, fT4, TT3, and TT4 were measured with Abbott Architect and Roche Cobas 6000. fT3, fT4, TT3, and TT4 were also measured by LC-MS/MS. Tg was measured by Siemens IMMULITE 2000. TBG was assessed with Siemens IMMULITE 2000. Results: Significant changes in TSH, fT4, and TT3 measurements were observed after biotin exposure when the Roche Cobas 6000 platform was used. Biotin intake resulted in a falsely lower Tg level when measurements were performed with Siemens IMMULITE 2000. At the time points examined, maximal biotin interference was observed two hours after biotin exposure both on day 1 and day 8. Conclusions: A daily dose of 10 mg was shown to interfere with specific assays for TSH, fT4, TT3, and Tg. Physicians must be aware of the potential risk of erroneous test results in subjects taking biotin supplements. Altered test results for TSH and Tg can be particularly problematic in patients requiring careful titration of levothyroxine therapy such as those with thyroid cancer.


Assuntos
Biotina/análise , Biotina/farmacologia , Tireoglobulina/análise , Hormônios Tireóideos/análise , Tireotropina/análise , Adulto , Idoso , Cromatografia Líquida de Alta Pressão , Reações Falso-Negativas , Feminino , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Estudos Prospectivos , Testes de Função Tireóidea
18.
Clin Cancer Res ; 27(5): 1399-1409, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33355247

RESUMO

PURPOSE: The goal of this study was to analyze the role of somatostatin receptor type 2 (SSTR2) as a molecular target for the imaging and treatment of thyroid cancer through analysis of SSTR2 expression and its epigenetic modulation and testing tumor uptake of different radiolabeled SSTR2 analogues. EXPERIMENTAL DESIGN: We analyzed SSTR2 expression by immunostaining of 92 thyroid cancer tissue samples and quantified standard uptake values (SUVmax) of SSTR2 analogue, 68Ga-DOTA-TATE, by PET/CT imaging in 25 patients with metastatic thyroid cancer. We utilized human thyroid cancer cell lines characterized by differential SSTR2 expression (TT, BCPAP, and FTC133) and rat pancreatic cell line (AR42J) with intrinsically high SSTR2 expression for functional in vitro studies. SSTR2-high (AR42J) and SSTR2-low (FTC133) xenograft mouse models were used to test the uptake of radiolabeled SSTR2 analogues and their therapeutic efficacy in vivo. RESULTS: Thyroid cancer had a higher SSTR2 expression than normal thyroid. Hurthle cell thyroid cancer was characterized by the highest 68Ga-DOTA-TATE uptake [median SUVmax, 16.5 (7.9-29)] than other types of thyroid cancers. In vivo studies demonstrated that radiolabeled DOTA-EB-TATE is characterized by significantly higher tumor uptake than DOTA-TATE (P < 0.001) and DOTA-JR11 (P < 0.001). Treatment with 177Lu-DOTA-EB-TATE extended survival and reduced tumor size in a mouse model characterized by high somatostatin (SST) analogues uptake (SUVmax, 15.16 ± 4.34), but had no effects in a model with low SST analogues uptake (SUVmax, 4.8 ± 0.27). CONCLUSIONS: A novel SST analogue, 177Lu-DOTA-EB-TATE, has the potential to be translated from bench to bedside for the targeted therapy of patients characterized by high uptake of SST analogues in metastatic lesions.


Assuntos
Compostos Radiofarmacêuticos/administração & dosagem , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Neoplasias da Glândula Tireoide/tratamento farmacológico , Adulto , Animais , Apoptose , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Pessoa de Meia-Idade , Metástase Neoplásica , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Prognóstico , Compostos Radiofarmacêuticos/metabolismo , Receptores de Somatostatina/química , Somatostatina/administração & dosagem , Somatostatina/metabolismo , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Lancet Oncol ; 21(11): e528-e537, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33152312

RESUMO

Most primary thyroid tumours are of epithelial origin. Primary thyroid mesenchymal tumours are rare but are being increasingly detected. A vast majority of thyroid mesenchymal tumours occur between the fourth and seventh decades of life, presenting as progressively enlarging thyroid nodules that often yield non-diagnostic results or spindle cells on fine needle aspiration biopsy. Surgery is the preferred mode of treatment, with adjuvant chemoradiotherapy used for malignant thyroid mesenchymal tumours. Benign thyroid mesenchymal tumours have excellent prognosis, whereas the outcome of malignant thyroid mesenchymal tumours is variable. Each thyroid mesenchymal tumour is characterised by its unique histopathology and immunohistochemistry. Because of the rarity and aggressive nature of malignant thyroid mesenchymal tumours, a multidisciplinary team-based approach should ideally be used in the management of these tumours. Comprehensive guidelines on the management of thyroid mesenchymal tumours are currently lacking. In this Review, we provide a detailed description of thyroid mesenchymal tumours, their clinical characteristics and tumour behaviour, and provide recommendations for the optimal management of these tumours.


Assuntos
Biomarcadores Tumorais , Neoplasias de Tecido Conjuntivo e de Tecidos Moles , Neoplasias da Glândula Tireoide , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Tomada de Decisão Clínica , Humanos , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/química , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/genética , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/patologia , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/terapia , Valor Preditivo dos Testes , Prognóstico , Neoplasias da Glândula Tireoide/química , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA