Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Horm Metab Res ; 54(12): 795-812, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35878617

RESUMEN

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.


Asunto(s)
Tumores Neuroendocrinos , Humanos , Tumores Neuroendocrinos/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico
2.
Cytopathology ; 33(3): 344-349, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34957617

RESUMEN

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.


Asunto(s)
Neoplasias de la Tiroides , Nódulo Tiroideo , Análisis Mutacional de ADN , Humanos , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/diagnóstico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Nódulo Tiroideo/patología
3.
Lancet Oncol ; 21(11): e528-e537, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33152312

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de los Tejidos Conjuntivo y Blando , Neoplasias de la Tiroides , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Toma de Decisiones Clínicas , Humanos , Neoplasias de los Tejidos Conjuntivo y Blando/química , Neoplasias de los Tejidos Conjuntivo y Blando/genética , Neoplasias de los Tejidos Conjuntivo y Blando/patología , Neoplasias de los Tejidos Conjuntivo y Blando/terapia , Valor Predictivo de las Pruebas , Pronóstico , Neoplasias de la Tiroides/química , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/terapia
4.
Mol Pharmacol ; 97(1): 2-8, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31704717

RESUMEN

The thyrotropin (TSH) receptor (TSHR) signals via G proteins of all four classes and ß-arrestin 1. Stimulation of TSHR leads to increasing cAMP production that has been reported as a monotonic dose-response curve that plateaus at high TSH doses. In HEK 293 cells overexpressing TSHRs (HEK-TSHR cells), we found that TSHR activation exhibits an "inverted U-shaped dose-response curve" with increasing cAMP production at low doses of TSH and decreased cAMP production at high doses (>1 mU/ml). Since protein kinase A inhibition by H-89 and knockdown of ß-arrestin 1 or ß-arrestin 2 did not affect the decreased cAMP production at high TSH doses, we studied the roles of TSHR downregulation and of Gi/Go proteins. A high TSH dose (100 mU/ml) caused a 33% decrease in cell-surface TSHR. However, because inhibiting TSHR downregulation with combined expression of a dominant negative dynamin 1 and ß-arrestin 2 knockdown had no effect, we concluded that downregulation is not involved in the biphasic cAMP response. Pertussis toxin, which inhibits activation of Gi/Go, abolished the biphasic response with no statistically significant difference in cAMP levels at 1 and 100 mU/ml TSH. Concordantly, co-knockdown of Gi/Go proteins increased cAMP levels stimulated by 100 mU/ml TSH from 55% to 73% of the peak level. These data show that biphasic regulation of cAMP production is mediated by Gs and Gi/Go at low and high TSH doses, respectively, which may represent a mechanism to prevent overstimulation in TSHR-expressing cells. SIGNIFICANCE STATEMENT: We demonstrate biphasic regulation of TSH-mediated cAMP production involving coupling of the TSH receptor (TSHR) to Gs at low TSH doses and to Gi/o at high TSH doses. We suggest that this biphasic cAMP response allows the TSHR to mediate responses at lower levels of TSH and that decreased cAMP production at high doses may represent a mechanism to prevent overstimulation of TSHR-expressing cells. This mechanism could prevent chronic stimulation of thyroid gland function.


Asunto(s)
AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Receptores de Tirotropina/metabolismo , Transducción de Señal/efectos de los fármacos , Tirotropina/administración & dosificación , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Dinamina I/genética , Dinamina I/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Toxina del Pertussis/administración & dosificación , Receptores de Tirotropina/genética , Transducción de Señal/genética , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
5.
Oncologist ; 25(4): 290-300, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32297436

RESUMEN

Immune checkpoint inhibitors have proven to be effective for various advanced neoplasia. Immune-related adverse events (irAEs) as a result of increased T cell activation are unique and potentially life-threating toxicities associated with the use of immune checkpoint inhibitors. Multiple endocrine irAEs, including primary hyperthyroidism and hypothyroidism, thyroiditis, primary adrenal insufficiency, type 1 diabetes mellitus, and hypophysitis, have been reported with the use of various immune checkpoint inhibitors. In some cases, these irAEs can lead to discontinuation of treatment. Here we propose for the general oncologist algorithms for managing endocrine irAEs to aid in the clinical care of patients receiving immunotherapy. KEY POINTS: There is a relative high risk of endocrine immune-related adverse events (irAEs) during therapy with checkpoint inhibitors, particularly when combination therapy is implemented. Patients treated with anti-CTLA-4 antibodies have an increased risk of hypophysitis, whereas patients treated with anti-PD-1/PD-L1 antibodies have a higher risk of primary thyroid dysfunction. Rarely, patients develop T1DM and central diabetes insipidus, and hypoparathyroidism is a rare occurrence. A growing clinical understanding of endocrine irAEs has led to effective treatment strategies with hormone replacement.


Asunto(s)
Hipofisitis , Neoplasias , Algoritmos , Humanos , Hipofisitis/inducido químicamente , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia/efectos adversos , Neoplasias/tratamiento farmacológico
6.
Horm Metab Res ; 52(8): 562-577, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32040962

RESUMEN

The diagnostic modalities, stratification tools, and treatment options for patients with thyroid cancer have rapidly evolved since the development of the American Thyroid Association (ATA) guidelines in 2015. This review compiles newer concepts in diagnosis, stratification tools and treatment options for patients with differentiated thyroid cancer (DTC), medullary thyroid carcinoma (MTC) and anaplastic thyroid cancer (ATC). Newer developments apply precision medicine in thyroid cancer patients to avoid over-treatment in low risk disease and under-treatment in high risk disease. Among novel patient-tailored therapies are selective RET inhibitors that have shown efficacy in the treatment of MTC with limited systemic toxicity compared with non-specific tyrosine kinase inhibitors. The combination of BRAF and MEK inhibitors have revolutionized management of BRAF V600E mutant ATC. Several immunotherapeutic agents are being actively investigated in the treatment of all forms of thyroid cancer. In this review, we describe the recent advances in the diagnosis and management of DTC, MTC, and ATC, with an emphasis on novel treatment modalities.


Asunto(s)
Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Neoplasias de la Tiroides/terapia , Tiroidectomía/métodos , Terapia Combinada , Manejo de la Enfermedad , Humanos , Comunicación Interdisciplinaria , Pronóstico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
7.
Clin Endocrinol (Oxf) ; 89(4): 481-488, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29972703

RESUMEN

OBJECTIVE: Pretreatment with lithium (Li) is associated with an increased residence time of radioactive iodine (RAI) in differentiated thyroid cancer (DTC) metastases. There are no data translating this observation into long-term outcomes. The study goal was to compare the efficacy of three methods of preparation for RAI therapy in metastatic DTC-thyroid hormone withdrawal (THW), THW with pretreatment with Li (THW+Li), and recombinant human TSH (rhTSH). DESIGN/PATIENTS/MEASUREMENTS: We performed a cohort study comparing overall survival (OS) and progression-free survival (PFS) between the three groups: THW (n = 52), THW+Li (n = 41) and rhTSH (n = 42). Kaplan-Meier analyses were performed to compare OS and PFS between the groups. Cox proportional hazards regression model with a stepwise variable selection was performed to study the contribution of age, gender, histology, TNM status, a location of distant metastases and RAI dose. RESULTS: During the follow-up of median 5.1 (IQR = 3.0-8.1) years, 52% of patients had disease progression and 12.6% died. Although THW+Li group was characterized by the longest OS (P = 0.007), only age (HR 1.05, CI 1.01-1.09, P = 0.01) and widespread disease (HR 3.8, CI 1.2-11.8, P = 0.02) were found to affect OS in a multivariate model. There was no difference in PFS between the groups (P = 0.47). Presence of distant metastases limited to the lungs only was associated with longer PFS (PFS HR 0.35, CI 0.20-0.60, P = 0.0002). CONCLUSION: The older age is associated with shorter OS, while disease burden affects OS and PFS in patients with metastatic thyroid cancer. The method of preparation for RAI therapy does not affect the outcome.


Asunto(s)
Radioisótopos de Yodo/uso terapéutico , Litio/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Adulto , Estudios de Cohortes , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pruebas de Función de la Tiroides , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/mortalidad , Tiroidectomía
10.
Thyroid ; 34(6): 674-686, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38717947

RESUMEN

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.


Asunto(s)
Neoplasias de la Tiroides , Tirotropina , Humanos , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/mortalidad , Neoplasias de la Tiroides/patología , Tirotropina/sangre , Supervivencia sin Enfermedad , Supervivencia sin Progresión , Resultado del Tratamiento
11.
Artículo en Inglés | MEDLINE | ID: mdl-38421044

RESUMEN

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.

12.
Front Endocrinol (Lausanne) ; 14: 1275813, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37886645

RESUMEN

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.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Feocromocitoma , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Anciano , Feocromocitoma/radioterapia , Estudios Retrospectivos , Prolactina , Hidrocortisona , Hormona Adrenocorticotrópica , Hormona Folículo Estimulante , Catecolaminas , Tirotropina
13.
Clin Thyroidol ; 34(7): 286-289, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36937987

RESUMEN

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.

14.
Clin Thyroidol ; 34(4): 161-164, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36969797

RESUMEN

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.

15.
Pol Arch Intern Med ; 132(3)2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35243857

RESUMEN

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.


Asunto(s)
Enfermedad de Hashimoto , Hipotiroidismo , Nacimiento Prematuro , Femenino , Enfermedad de Hashimoto/diagnóstico , Enfermedad de Hashimoto/terapia , Humanos , Recién Nacido , Masculino , Embarazo , Nacimiento Prematuro/tratamiento farmacológico , Hormonas Tiroideas , Tiroxina/uso terapéutico , Triyodotironina/uso terapéutico
16.
Front Endocrinol (Lausanne) ; 13: 896287, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35712243

RESUMEN

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.


Asunto(s)
Carcinoma Neuroendocrino , Neoplasias de la Tiroides , Carcinoma Neuroendocrino/patología , Humanos , Radioisótopos de Yodo/uso terapéutico , Receptores de Somatostatina/uso terapéutico , Neoplasias de la Tiroides/patología
17.
Thyroid ; 32(1): 90-96, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34714162

RESUMEN

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.


Asunto(s)
Autoanticuerpos/inmunología , Fibroblastos/inmunología , Oftalmopatía de Graves/complicaciones , Células Epiteliales Tiroideas/inmunología , Adulto , Autoanticuerpos/análisis , Femenino , Fibroblastos/metabolismo , Enfermedad de Graves/sangre , Enfermedad de Graves/inmunología , Oftalmopatía de Graves/sangre , Oftalmopatía de Graves/patología , Humanos , Masculino , Persona de Mediana Edad , Células Epiteliales Tiroideas/metabolismo , Tirotropina/metabolismo
18.
J Clin Transl Endocrinol ; 27: 100285, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34900602

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen responsible for coronavirus disease 2019 (COVID-19) has been a major cause of morbidity and mortality globally. Older age, and the presence of certain components of metabolic syndrome, including hypertension have been associated with increased risk for severe disease and death in COVID-19 patients. The role of antihypertensive agents in the pathogenesis of COVID-19 has been extensively studied since the onset of the pandemic. This review discusses the potential pathophysiologic interactions between hypertension and COVID-19 and provides an up-to-date information on the implications of newly emerging SARS-CoV-2 variants, and vaccines on patients with hypertension.

19.
Cell Signal ; 90: 110212, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34896620

RESUMEN

The TSH receptor (TSHR) is the major regulator of thyroid hormone biosynthesis in human thyrocytes by regulating the transcription of a number of genes including thyroglobulin (TG) and thyroperoxidase (TPO). Until recently, it was thought that TSHR initiated signal transduction pathways only at the cell-surface and that internalization was primarily involved in TSHR desensitization and downregulation. Studies primarily in mouse cells showed that TSHR internalization regulates gene transcription at an intracellular site also. However, this has not been shown for genes involved in thyroid hormone biosynthesis in human thyrocytes. We used human thyrocytes in primary culture. In these cells, the dose-response to TSH for gene expression is biphasic with low doses upregulating gene expression and higher doses decreasing gene expression. We used two approaches to inhibit internalization. In the first, we used inhibitors of dynamins, dynasore and dyngo-4a. Pretreatment with dynasore or dyngo-4a markedly inhibited TSH upregulation of TG and TPO mRNAs, as well as TG secretion. In the second, we used knockdown of dynamin 2, which is the most abundant dynamin in human thyrocytes. We showed that dynamin 2 knockdown inhibited TSHR internalization and decreased the TSH-stimulated levels of TG and TPO mRNAs and proteins. Lastly, we showed that the level of the activatory transcription factor phosphorylated cAMP response element binding protein (pCREB) in the cell nuclei was reduced by 68% when internalization was inhibited. We conclude that upregulation of genes involved in thyroid hormone synthesis in human thyrocytes is, in part, dependent on internalization leading to nuclear localization of an activated transcription factor(s).


Asunto(s)
Yoduro Peroxidasa , Tiroglobulina , Animales , Humanos , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Ratones , Receptores de Tirotropina/genética , Receptores de Tirotropina/metabolismo , Tiroglobulina/genética , Tiroglobulina/metabolismo , Tirotropina/genética , Tirotropina/farmacología , Transcripción Genética
20.
Front Pharmacol ; 13: 791710, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36249761

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA