Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
1.
Thyroid ; 34(1): 70-81, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37917101

RESUMEN

Objective: Redifferentiation therapy (RDT) can restore radioactive iodine (RAI) uptake in differentiated thyroid cancer (DTC) cells to enable salvage 131I therapy for previously RAI refractory (RAIR) disease. This study evaluated the clinical outcomes of patients who underwent RDT and identified clinicopathologic characteristics predictive of RAI restoration following RDT. Methods: This is a retrospective case series of 33 patients with response evaluation criteria in solid tumors (RECIST)-progressive metastatic RAIR-DTC who underwent RDT between 2017 and 2022 at the Mayo Clinic (Rochester, MN). All patients underwent genomic profiling and received MEK, RET or ALK inhibitors alone, or combination BRAF-MEK inhibitors for 4 weeks. At week 3, those with increased RAI avidity in metastatic foci received high-dose 131I therapy. Baseline and clinicopathologic outcomes were comprehensively reviewed. Results: Of the 33 patients, 57.6% had restored RAI uptake following RDT (Redifferentiated subgroup). 42.1% (8/19) with papillary thyroid cancers (PTC), 100% (4/4) with invasive encapsulated follicular variant PTCs (IEFV-PTCs), and 100% (7/7) with follicular thyroid cancers (FTC) redifferentiated. All (11/11) RAS mutant tumors redifferentiated compared with 38.9% (7/18) with BRAF mutant disease (6 PTC and 1 IEFV-PTC). 76.5% (13/17) of redifferentiated and 66.7% (8/12) of non-redifferentiated patients achieved a best overall RECIST response of stable disease (SD) or non-complete response/non-progressive disease. Both subgroups had a median 12% tumor shrinkage at 3 weeks on drug(s) alone. The redifferentiated subgroup, following high-dose 131I therapy, achieved an additional median 20% tumor reduction at 6 months after RDT. There were no statistically significant differences between both groups in progression free survival (PFS), time to initiation of systemic therapy, and time to any additional therapy. Of the entire cohort, 6.1% (2/33) experienced histologic transformation to anaplastic thyroid cancer, 15.1% (5/33) died, and all had redifferentiated following RDT and received 131I therapy. Conclusion: RDT has the potential to restore RAI avidity and induce RECIST responses following 131I therapy in select patients with RAIR-DTC, particularly those with RAS-driven "follicular" phenotypes. In patients with PTC, none of the evaluated clinical outcomes differed statistically between the redifferentiated and non-redifferentiated subgroups. Further studies are needed to better characterize the long-term survival and/or safety outcomes of high-dose RAI following RDT, particularly whether it could be associated with histologic anaplastic transformation.


Asunto(s)
Adenocarcinoma Folicular , Yodo , Neoplasias de la Tiroides , Humanos , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/radioterapia , Radioisótopos de Yodo/uso terapéutico , Yodo/uso terapéutico , Estudios Retrospectivos , Proteínas Proto-Oncogénicas B-raf/genética , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/genética , Adenocarcinoma Folicular/radioterapia , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/uso terapéutico
2.
Endocrine ; 83(2): 251-258, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37824045

RESUMEN

PURPOSE: This review focuses on post-operative thyroid hormone replacement and thyrotropin suppression therapy in patients with differentiated thyroid cancer. METHODS: A clinical review. RESULTS: Differentiated thyroid cancers (DTC), including papillary and follicular thyroid cancers, have an excellent prognosis and their management leverages a unique set of clinical tools arising from homology to the normal thyroid follicular cell. Surgery is the cornerstone of initial management, and post-operative care often requires thyroid hormone replacement therapy, which may be approached with the intent of physiologic normalization or used pharmacologically to suppress TSH as part of a DTC treatment. CONCLUSION: Management of DTC and approaches to TSH suppression are tailored to an individual's risk of DTC recurrence and are adjusted to a patient's clinical status and comorbidities over time with the goal of mitigating risk and maximizing benefit.


Asunto(s)
Adenocarcinoma Folicular , Antineoplásicos , Neoplasias de la Tiroides , Humanos , Tirotropina , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/tratamiento farmacológico , Terapia de Reemplazo de Hormonas , Antineoplásicos/uso terapéutico , Hormonas Tiroideas/uso terapéutico , Tiroxina/uso terapéutico
3.
Histopathology ; 83(4): 657-663, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37501641

RESUMEN

AIMS: Lenvatinib is a multikinase inhibitor used for treating unresectable or metastatic cancers, including thyroid cancer. As total thyroidectomy followed by radioactive iodine therapy is a commonly recommended initial treatment for thyroid cancer, histological findings of the thyroid after lenvatinib therapy remain unclear. Therefore, the aim of this study was to analyse in-vivo changes in patients who underwent thyroidectomy after lenvatinib therapy. METHODS AND RESULTS: We screened 167 patients with thyroid cancer [papillary thyroid cancer (PTC), n = 102; follicular thyroid cancer (FTC), n = 26; anaplastic thyroid cancer (ATC), n = 39] who underwent lenvatinib therapy. Among these patients, six underwent thyroidectomy (lenvatinib-treated group: PTC, n = 3; FTC, n = 1; ATC, n = 2), and the specimens were examined. Five patients with PTC who did not receive lenvatinib therapy were included for comparison (untreated group). Microvessel density (MVD) was evaluated in both groups. The PTC and FTC specimens showed relatively more ischaemic changes than ATC specimens. Coagulative necrosis and ischaemic changes in cancer cells were frequently observed. ATC specimens showed fibrosis and mild cell damage. As hypothyroidism is a common side effect of lenvatinib therapy, non-cancerous thyroid tissues were also examined. Histological findings included mild lymphocytic infiltration, lymphoid follicular formation, histiocytic reaction and follicular epithelial destruction. The MVD in lenvatinib-treated tissues was significantly lower than that in untreated tissues. CONCLUSIONS: Lenvatinib therapy probably induces relatively specific ischaemic changes in thyroid cancer cells. Moreover, inflammatory cell infiltration and decreased MVD occur to varying degrees in non-cancerous thyroid tissue and may be related to hypothyroidism, a side effect of lenvatinib.


Asunto(s)
Adenocarcinoma Folicular , Carcinoma Anaplásico de Tiroides , Neoplasias de la Tiroides , Humanos , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/patología , Radioisótopos de Yodo/uso terapéutico , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/patología , Compuestos de Fenilurea/efectos adversos , Cáncer Papilar Tiroideo/tratamiento farmacológico
4.
Front Endocrinol (Lausanne) ; 14: 1117001, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37324277

RESUMEN

Objective: Repeat radioiodine (RAI) treatment has been widely implemented for RAI-avid lung metastases and is clinically effective for lung metastatic differentiated thyroid cancer (DTC). We aim to investigate the association between the interval of RAI treatment and short-term response, and the side effects in patients with lung metastases from DTC and to identify predictors for non-effective response to the next RAI treatment. Methods: A total of 282 course pairs from 91 patients were established and categorized into two groups by the interval of neighboring RAI treatment (<12 and ≥12 months), and the characteristics and treatment response between the two groups were compared. Multivariate logistic regression was used to identify predictors associated with treatment response. The side effects in the former course and the latter course were compared while taking into account the interval. Results: No significant difference was found between the two groups in treatment response in the latter course (p > 0.05). In the multivariate analysis, age ≥ 55 years (OR = 7.29, 95% CI = 1.66-33.35, p = 0.008), follicular thyroid cancer (OR = 5.00, 95% CI = 1.23-22.18, p = 0.027), and a second RAI treatment as the former course (OR = 4.77, 95% CI = 1.42-18.61, p = 0.016) were significantly associated with a non-effective response. There was no significant difference in the side effects in the former and latter courses between the two groups (p > 0.05). Conclusion: The interval of RAI treatment does not affect short-term response and side effects of DTC patients with RAI-avid lung metastases. It was feasible to defer repeat evaluation and treatment with an interval of at least 12 months to obtain an effective response and reduce the risk of side effects.


Asunto(s)
Adenocarcinoma Folicular , Neoplasias Pulmonares , Neoplasias de la Tiroides , Humanos , Persona de Mediana Edad , Radioisótopos de Yodo/efectos adversos , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/radioterapia , Adenocarcinoma Folicular/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/tratamiento farmacológico
5.
Eur Thyroid J ; 12(3)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36976625

RESUMEN

Background: Treatment of advanced follicular thyroid carcinoma (FTC) is based primarily on indirect evidence obtained with multikinase inhibitors (MKI) in clinical trials in which papillary carcinomas represent the vast majority of cases. However, it should be noted that MKI have a non-negligible toxicity that may decrease the patient's quality of life. Conventional chemotherapy with GEMOX (gemcitabine plus oxaliplatin) is an off-label therapy, which seems to have some effectiveness in advanced differentiated thyroid carcinomas, with a good safety profile, although further studies are needed. Case report: We report a case of a metastatic FTC, resistant to several lines of therapy. However, with a durable response to GEMOX, the overall survival of our patient appears to have been extended significantly due to this chemotherapy. Conclusion: GEMOX may have a role in patients with thyroid cancer unresponsive to MKI.


Asunto(s)
Adenocarcinoma Folicular , Neoplasias de la Tiroides , Humanos , Gemcitabina , Oxaliplatino/uso terapéutico , Calidad de Vida , Adenocarcinoma Folicular/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico
6.
Cancer ; 129(8): 1195-1204, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36748723

RESUMEN

BACKGROUND: The authors report results from the thyroid carcinoma cohort of the multicohort phase 2 KEYNOTE-158 study (NCT02628067), which evaluated pembrolizumab monotherapy in patients with previously treated cancers. METHODS: Eligible patients had histologically and/or cytologically confirmed papillary or follicular thyroid carcinoma, failure of or intolerance to prior therapy, and measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. Patients received pembrolizumab (200 mg) every 3 weeks for up to 35 cycles. The primary end point was objective response rate (ORR) per RECIST v1.1 by independent central review. RESULTS: A total of 103 patients were enrolled and received pembrolizumab. Median duration from first dose to data cutoff (October 5, 2020) was 49.4 (range, 43.9-54.9) months. ORR was 6.8% (95% confidence interval [CI], 2.8%-13.5%), and median duration of response was 18.4 (range, 4.2-47.2+) months. ORR was 8.7% (95% CI, 2.4%-20.8%) among patients with programmed cell death ligand 1 (PD-L1) combined positive score (CPS) ≥1 (n = 46) and 5.7% (95% CI, 1.2%-15.7%) among patients with PD-L1 CPS <1 (n = 53). Median overall survival and progression-free survival were 34.5 (95% CI, 21.2 to not reached) and 4.2 (95% CI, 3.9-6.2) months, respectively. Treatment-related adverse events occurred in 69.9% of patients (grade 3-5, 14.6%). CONCLUSIONS: Pembrolizumab demonstrated manageable toxicity and durable antitumor activity in a small subset of patients with advanced thyroid cancer. These results provide evidence of modest antitumor activity in this setting regardless of tumor PD-L1 expression. Future studies evaluating immune checkpoint inhibitors in patients with differentiated thyroid cancer should focus on biomarker-driven patient selection or combination of immune checkpoint inhibitors with other agents, in order to achieve higher response rates than observed in this study.


Asunto(s)
Adenocarcinoma Folicular , Antineoplásicos Inmunológicos , Neoplasias de la Tiroides , Humanos , Inhibidores de Puntos de Control Inmunológico , Antígeno B7-H1 , Antineoplásicos Inmunológicos/efectos adversos , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/tratamiento farmacológico
7.
Endocr Relat Cancer ; 29(5): R57-R66, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35266878

RESUMEN

During the last decades, the knowledge on follicular cell-derived thyroid cancer molecular biology has led to the evolution of a number of novel therapies for these tumors, mainly tyrosine kinase inhibitors. Lenvantinib, sorafenib and recently cabozantinib have been approved for differentiated thyroid cancer (DTC), while larotrectinib and entrectinib for neurotrophic-tropomyosin receptor kinase-fusion thyroid cancer. For radioiodine (RAI) refractory DTCs ongoing research aims to identify agents that may restore RAI-avidity via redifferentiation protocols (vemurafenib or dabrafenib and trametinib) or combination treatments. These treatments are based on the tumor molecular signature. The treatment with targeted therapies has changed the therapeutic strategies and the disease prognosis, however drug resistance remains the main reason for treatment failure. Thus, the understanding of both molecular pathways implicated in tumorigenesis, and tumoral escape mechanisms, are of paramount significance for the development of new therapies for DTC. The present review focuses on the molecular landscape of DTC, the approved targeted therapies as well as the mechanisms of drug resistance. Furthermore, it points to the ongoing research and the future perspectives for the development of more efficient drugs for DTC.


Asunto(s)
Adenocarcinoma Folicular , Neoplasias de la Tiroides , Adenocarcinoma Folicular/tratamiento farmacológico , Humanos , Radioisótopos de Yodo/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Sorafenib , Neoplasias de la Tiroides/patología
8.
Cancer Treat Rev ; 106: 102380, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35305441

RESUMEN

Most malignant thyroid tumours are initially treated with surgery or a combination of surgery and radioactive iodine (RAI) therapy. However, in patients with metastatic disease, many tumours become refractory to RAI, and these patients require alternative treatments, such as locoregional therapies and/or systemic treatment with multikinase inhibitors. Improvements in our understanding of the genetic alterations that occur in thyroid cancer have led to the discovery of several targeted therapies with clinical efficacy. These alterations include NTRK (neurotrophic tyrosine receptor kinase) gene fusions, with the tropomyosin receptor kinase inhibitors larotrectinib and entrectinib both approved by the European Medicines Agency and in other markets worldwide. Inhibitors of aberrant proteins resulting from alterations in RET (rearranged during transfection) and BRAF (B-Raf proto-oncogene) have also shown promising efficacy, and so far have received approval by the US Food and Drug Administration. Selpercatinib, a RET kinase inhibitor, was approved for use in Europe in early 2021. With the discovery of multiple actionable targets, it is imperative that effective testing strategies for these genetic alterations are integrated into the diagnostic armamentarium to ensure that patients who could potentially benefit from targeted treatments are identified. In this review, we offer our recommendations on the optimal testing strategies for detecting genetic alterations in thyroid cancer that have the potential to be targeted by molecular therapy. We also discuss the future of treatments for thyroid cancers, including the use of immune checkpoint inhibitors, and new generations of targeted treatments that are being developed to counter acquired tumour resistance.


Asunto(s)
Adenocarcinoma Folicular , Neoplasias de la Tiroides , Adenocarcinoma Folicular/diagnóstico , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/genética , Humanos , Radioisótopos de Yodo , Medicina de Precisión , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/diagnóstico , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética
9.
Horm Metab Res ; 53(10): 683-691, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34607367

RESUMEN

In differentiated thyroid cancer (DTC), the standard treatment includes total thyroidectomy and lifetime levothyroxine (LT4) replacement. However, long-term exogenous LT4 has become controversial due to the adverse effects of oversuppression. The study included 191 patients (aged 18-76 years) with a prospective diagnosis of non-metastatic DTC and 79 healthy individuals. The patients with DTC were stratified into three groups according to their TSH levels: suppressed thyrotropin if TSH was below 0.1 µIU/ml, mildly suppressed thyrotropin if TSH was between 0.11 and 0.49 µIU/ml, and low-normal thyrotropin if THS was between 0.5 and 2 µIU/ml. The Beck Depression Inventory (BDI), Beck Anxiety Inventory (BAI), Anxiety Sensitivity Index (ASI), Short Symptom Inventory (SSI), and Pittsburgh Sleep Quality Index (PSQI) were administered to all participants. It was found that the BDI, BAI, SSI and PSQI scores were worse in patients with DTC (p=0.024, p=0.014, p=0.012, and p=0.001, respectively). According to theTSH levels, the mean ASI was found to be higher in the suppressed and mildly suppressed thyrotropin groups (19±14.4 vs. 10.6±11.1; 16.4±14.9 vs. 10.6±11.1, p=0.024, respectively), the mean SSI was found higher in the suppressed group (61.0±55.5 vs. 35.1±37.0, p=0.046), and the mean PSQI was higher in all three groups (7.94±3.97 vs. 5.35±4.13; 7.21±4.59 vs. 5.35±4.13; 7.13±4.62 vs. 5.35±4.13, p=0.006) when compared with the controls. No significant difference was found between the groups. A positive correlation was detected in the duration of LT4 use and BDI and SSI, and a weak, negative correlation was detected between TSH levels and ASI and PSQI. Based on our study, it was found that depression, anxiety disorders, and sleep problems were more prevalent in patients with DTC, being more prominent in the suppressed TSH group. These results were inversely correlated with TSH values and positively correlated with the duration of LT4 use. Unnecessary LT4 oversuppression should be avoided in patients with DTC.


Asunto(s)
Adenocarcinoma Folicular , Trastornos Mentales , Calidad del Sueño , Neoplasias de la Tiroides , Tirotropina/sangre , Tiroxina/efectos adversos , Adenocarcinoma Folicular/sangre , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/psicología , Adenocarcinoma Folicular/cirugía , Adolescente , Adulto , Anciano , Enfermedades Asintomáticas , Estudios de Casos y Controles , Enfermedad Crónica , Estudios Transversales , Regulación hacia Abajo/efectos de los fármacos , Femenino , Terapia de Reemplazo de Hormonas/efectos adversos , Humanos , Hipertiroidismo/sangre , Hipertiroidismo/inducido químicamente , Hipertiroidismo/fisiopatología , Hipertiroidismo/psicología , Hipotiroidismo/sangre , Hipotiroidismo/tratamiento farmacológico , Hipotiroidismo/epidemiología , Hipotiroidismo/etiología , Masculino , Trastornos Mentales/sangre , Trastornos Mentales/epidemiología , Trastornos Mentales/etiología , Persona de Mediana Edad , Neoplasias de la Tiroides/sangre , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/psicología , Neoplasias de la Tiroides/cirugía , Tiroidectomía/rehabilitación , Tirotropina/efectos de los fármacos , Tiroxina/uso terapéutico , Turquía/epidemiología , Adulto Joven
10.
Thyroid ; 31(10): 1597-1602, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34114495

RESUMEN

Background: We report the therapeutic use of K1-70™, a thyrotropin receptor (TSHR) antagonist monoclonal antibody, in a patient with follicular thyroid cancer (FTC), Graves' disease (GD), and Graves' ophthalmopathy (GO). Methods: A 51-year-old female patient, who smoked, presented in October 2014 with FTC complicated by GD, high levels of TSHR autoantibodies with high thyroid stimulating antibody (TSAb) activity, and severe GO. K1-70 was administered at 3 weekly intervals with the dose adjusted to block TSAb activity. Her cancer was managed with lenvatinib and radioiodine therapy. Results: Following initiation of K1-70 therapy, TSAb activity measured in serum decreased and GO (proptosis and inflammation) improved. On K1-70 monotherapy during the pause in lenvatinib, several metastatic lesions stabilized while others showed progression attenuation compared with that before lenvatinib therapy. Conclusions: These observations suggest that blocking TSHR stimulation with K1-70 can be an effective treatment for GO and may also benefit select patients with FTC and GD.


Asunto(s)
Adenocarcinoma Folicular/complicaciones , Adenocarcinoma Folicular/tratamiento farmacológico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Enfermedad de Graves/complicaciones , Enfermedad de Graves/tratamiento farmacológico , Oftalmopatía de Graves/complicaciones , Oftalmopatía de Graves/tratamiento farmacológico , Receptores de Tirotropina/antagonistas & inhibidores , Neoplasias de la Tiroides/complicaciones , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/inmunología , Anticuerpos Monoclonales/farmacología , Antineoplásicos/uso terapéutico , Autoanticuerpos/sangre , Femenino , Enfermedad de Graves/inmunología , Oftalmopatía de Graves/inmunología , Humanos , Inmunoglobulinas Estimulantes de la Tiroides/sangre , Radioisótopos de Yodo/uso terapéutico , Persona de Mediana Edad , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/uso terapéutico , Quinolinas/administración & dosificación , Quinolinas/uso terapéutico , Radiofármacos/uso terapéutico , Receptores de Tirotropina/inmunología , Neoplasias de la Tiroides/inmunología , Resultado del Tratamiento
11.
J Clin Endocrinol Metab ; 106(8): 2355-2366, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-33901285

RESUMEN

CONTEXT: 18F-fluoro-2-deoxy-D-glucose positron emission tomography-computed tomography ([18F]-FDG-PET/CT)-positive metastatic lesions in radioiodine-refractory differentiated thyroid cancer (RAI-R DTC) have a poor prognosis and lenvatinib represents the best therapy. OBJECTIVE: We investigated the role of [18F]-FDG-PET/CT in the evaluation of metabolic response and prediction of the outcome of RAI-R DTC patients treated with lenvatinib. METHODS: Patients (n = 33) with progressive metastatic RAI-R DTC who were treated with lenvatinib were investigated at baseline and during follow-up with biochemical (thyroglobulin and thyroglobulin antibodies), morphological (whole-body CT scan) and metabolic ([18F]-FDG-PET/CT) evaluation. RESULTS: Nineteen (57.6%) patients showed the greatest metabolic response at the first [18F]-FDG-PET/CT scan, performed after 4 weeks of lenvatinib, while 5/33 (15.1%) patients had this response later. Moreover, 66.7% of patients had both a metabolic response at the first [18F]-FDG-PET/CT scan and a morphological response at the first CT scan. We observed a correlation between the metabolic response at [18F]-FDG-PET/CT scan performed after 4 weeks of treatment and the biochemical response at the same time in 60.6% of patients. The median overall survival (OS) was significantly longer in patients with either a metabolic response at last [18F]-FDG-PET/CT (40.00 vs 8.98 months) or a morphological response at last CT scan (37.22 vs 9.53 months) than in those without response. Moreover, the OS was longer in patients with a metabolic response at [18F]-FDG-PET/CT performed after 4 weeks of treatment (36.53 vs 11.28 months). CONCLUSIONS: Our data show that [18F]-FDG-PET/CT can early predict the response to lenvatinib and correlates with the OS of RAI-R DTC patients treated with this drug.


Asunto(s)
Adenocarcinoma Folicular/diagnóstico por imagen , Antineoplásicos/uso terapéutico , Compuestos de Fenilurea/uso terapéutico , Quinolinas/uso terapéutico , Neoplasias de la Tiroides/diagnóstico por imagen , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/mortalidad , Adulto , Anciano , Femenino , Fluorodesoxiglucosa F18 , Humanos , Radioisótopos de Yodo , Masculino , Persona de Mediana Edad , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tasa de Supervivencia , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/mortalidad , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Imagen de Cuerpo Entero
12.
J Clin Endocrinol Metab ; 106(8): e3027-e3036, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-33769497

RESUMEN

CONTEXT: Radioiodine refractory differentiated thyroid cancer (RAIR-DTC) has been a global challenge due to its poor prognosis and limited treatment options. OBJECTIVE: We report here the long-term results of the phase II clinical trial of apatinib, an anti-angiogenic tyrosine kinase inhibitor, for RAIR-DTC. METHODS: This was an open-label, exploratory phase II clinical trial among progressive RAIR-DTC patients. Apatinib treatment was given once daily until disease progression, unmanageable toxicity, withdrawal, or death. The primary end points were objective response rate (ORR) and disease control rate (DCR). Progression-free survival (PFS), overall survival (OS), duration of response, long-term safety, and the association between patients with different tumor genotype (BRAFV600E and TERT promotor mutation) and their PFS rates were also assessed. RESULTS: The ORR was 80%, and the DCR was 95%. The overall median PFS was 18.4 months (95% CI, 9.2-36.8 months) and the median OS was 51.6 months (95% CI, 29.2-not reached [NR]). Patients with BRAFV600E mutation (10 of 18 evaluated) had a longer median PFS compared with patients with BRAF wild-type (NR vs 9.2 months; P = 0.002). The most common adverse events included palmar-plantar erythrodysesthesia syndrome (19/20), proteinuria (18/20), and hypertension (16/20). CONCLUSION: In this long-term evaluation, apatinib displayed sustainable efficacy and tolerable safety profile, warranting it as a promising treatment option for progressive RAIR-DTC.


Asunto(s)
Adenocarcinoma Folicular/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/uso terapéutico , Cáncer Papilar Tiroideo/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Adulto , Anciano , Antineoplásicos/efectos adversos , Femenino , Humanos , Radioisótopos de Yodo/uso terapéutico , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/efectos adversos , Piridinas/efectos adversos , Resultado del Tratamiento
13.
Surgery ; 169(1): 34-42, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32718802

RESUMEN

BACKGROUND: Patients with advanced differentiated thyroid cancer develop resistance to lenvatinib treatment from metabolic dysregulation. Heat shock protein 90 is a molecular chaperone that plays an important role in glycolysis and metabolic pathway regulation. We hypothesize that lenvatinib-resistant differentiated thyroid cancer cells will have an increased dependency on glycolysis and that a novel C-terminal heat shock protein 90 inhibitor (KU757) can effectively treat lenvatinib-resistant cells by targeting glycolysis. METHODS: Inhibitory concentration 50 values of thyroid cancer cells were determined by CellTiter-Glo assay (Promega Corp, Madison, WI). Glycolysis was measured through Seahorse experiments. Reverse transcription-polymerase chain reaction and Western blot evaluated glycolytic pathway genes/proteins. Exosomes were isolated/validated by nanoparticle tracking analysis and Western blot. Differentially expressed long non-coding ribonucleic acids in exosomes and cells were evaluated using quantitative polymerase chain reaction. RESULTS: Extracellular acidification rate demonstrated >2-fold upregulation of glycolysis in lenvatinib-resistant cells versus parent cells and was downregulated after KU757 treatment. Lenvatinib-resistant cells showed increased expression of the glycolytic genes lactic acid dehydrogenase, pyruvate kinase M1/2, and hexokinase 2. KU757 treatment resulted in downregulation of these genes and proteins. Several long non-coding ribonucleic acids associated with glycolysis were significantly upregulated in WRO-lenvatinib-resistant cells and exosomes and downregulated after KU757 treatment. CONCLUSION: Lenvatinib resistance leads to increased glycolysis, and KU757 effectively treats lenvatinib-resistant cells and overcomes this increased glycolysis by targeting key glycolytic genes, proteins, and long non-coding ribonucleic acids.


Asunto(s)
Adenocarcinoma Folicular/tratamiento farmacológico , Aminocumarinas/farmacología , Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/patología , Aminocumarinas/uso terapéutico , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Glucólisis/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Concentración 50 Inhibidora , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Quinolinas/farmacología , Quinolinas/uso terapéutico , Células Epiteliales Tiroideas , Neoplasias de la Tiroides/patología
14.
J Biochem Mol Toxicol ; 35(1): e22608, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32886819

RESUMEN

Thyroid cancer is the most frequent endocrine malignancy, which accounts for nearly 1% of all the cancer worldwide. Crocin has a diverse biological function, such as anti-cancer, anti-inflammatory and antioxidant functions, specifically in the respiratory related diseases. Using in vitro techniques, this work was intended to illuminate the anti-cancer effect of crocin in follicular thyroid carcinoma (FTC) (FT 133 cells), and the potential molecular mechanism convoluted. The outcome of the present work showed that crocin was able to prevent the proliferation and triggering the apoptosis in a dose-dependent mode, of FTC-133 cells by methyl thiazolyldiphenyl-tetrazolium bromide and staining assay (acridine orange/propiduim iodide [PI], 4',6-diamidino-2-phenylindole, and PI dye). Crocin did not show toxicity to the normal thyroid (PCCL3) cells. Crocin-induced reactive oxygen species, mitochondrial membrane potential activity, caspase-8 and -9, lipid peroxidation (thiobarbituric acid reactive substances) activity while suppressing antioxidant activity (superoxide dismutase, catalase, and glutathione) in FTC-133 cells. In addition, crocin was also participated in a halting of the proteins related to cell cycle, cyclin D1, and pro-apoptotic proteins; Bax and caspase-3 expression, together with the elevation of anti-apoptotic factor Bcl-2. Further, crocin have a dual inhibition of two major pathways, nuclear factor-κB, extracellular signal-regulated kinase, and janus kinase-signal transducer and activator of transcription signaling pathways. In conclusion, crocin can inhibit follicular thyroid carcinoma proliferation and promote cell apoptosis.


Asunto(s)
Adenocarcinoma Folicular , Apoptosis/efectos de los fármacos , Carotenoides/farmacología , Quinasas Janus/metabolismo , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción STAT/metabolismo , Neoplasias de la Tiroides , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/metabolismo , Adenocarcinoma Folicular/patología , Línea Celular Tumoral , Humanos , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología
15.
Front Endocrinol (Lausanne) ; 12: 798653, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069445

RESUMEN

Patients with bone metastases are treated with long-term bone resorption inhibitors such as bisphosphonates and denosumab. However, resorption inhibitors have been known to cause fractures, such as atypical femoral fractures (AFFs). In recent years, there have been an increasing number of reports of atypical ulna fractures (AUFs) caused by bone resorption inhibitor usage. Treatment of AUFs is complicated, especially when they occur in patients with bone metastases, because it is difficult to discontinue bone resorption inhibitor treatment without the risk of aggravating metastatic lesions. Prophylactic surgery is recommended in AFFs when fractures are predicted, but there are few reports of prophylactic surgery for AUFs. Here, we report a case of incomplete AUF in a 74-year-old woman which was surgically treated with prophylactic plate fixation. The patient had been using denosumab for 6 years to treat bone metastases due to thyroid cancer. After surgery, no fractures were observed for 2 years without discontinuing denosumab, and her forearm function was adequate. AUFs are rare and difficult to treat, so oncologists who treat bone metastases need to pay special attention to diagnose this incomplete AUF before the fracture worsens. We believe that detection of a possible fracture and prophylactic surgery can improve prognosis.


Asunto(s)
Adenocarcinoma Folicular/tratamiento farmacológico , Conservadores de la Densidad Ósea/efectos adversos , Neoplasias Óseas/tratamiento farmacológico , Placas Óseas , Denosumab/efectos adversos , Fijación de Fractura/métodos , Procedimientos Quirúrgicos Profilácticos/métodos , Neoplasias de la Tiroides/patología , Fracturas del Cúbito/prevención & control , Adenocarcinoma Folicular/secundario , Anciano , Neoplasias Óseas/secundario , Femenino , Humanos , Fracturas del Cúbito/inducido químicamente , Fracturas del Cúbito/diagnóstico por imagen , Fracturas del Cúbito/cirugía
16.
Drug Deliv ; 27(1): 1063-1072, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32672077

RESUMEN

GLOBOCAN estimates 36 types of cancers in 185 countries based on the incidence, mortality, and prevalence in the year 2019. Nowadays, chemotherapy is the most widely used cancer treatment among immune, radio, hormone, and gene therapies. Here, we describe a very simple yet cost-effective approach that synergistically combines drug reconstitution, supramolecular nano-assembly, and tumor-specific targeting to address the multiple challenges posed by the delivery of the chemotherapeutic Gemcitabine (GEM) drug. The GEM prodrugs were gifted to impulsively self-assemble into excellent steady nanoparticles size on covalent conjugation of linoleic acid hydrophobic through amide group with ∼100 nm. Newly synthesized GEM-NPs morphology was confirmed by various electron microscopic techniques. After successful synthesis, we have evaluated the anticancer property of GEM and GEM-NPs against B-CPAP (papillary thyroid carcinoma) and FTC-133 (human follicular thyroid carcinoma) cancer cell lines. Further studies such as AO-EB (acridine orange-ethidium bromide), nuclear staining and flow cytometry analyses on cell death mechanism signified that the cytotoxicity was associated with apoptosis in thyroid cancer cells. GEM-NPs show excellent biocompatibility compared to GEM. The present study explained that GEM-NPs as a safe and hopeful strategy for chemotherapeutics of thyroid cancer therapy and deserve for further clinical evaluations.


Asunto(s)
Adenocarcinoma Folicular/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Ácido Linoleico , Nanoconjugados , Polímeros , Profármacos/administración & dosificación , Cáncer Papilar Tiroideo/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/secundario , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Portadores de Fármacos , Humanos , Microscopía Electrónica , Nanopartículas/ultraestructura , Profármacos/farmacología , Cáncer Papilar Tiroideo/secundario , Neoplasias de la Tiroides/patología , Gemcitabina
17.
Cancer Biomark ; 28(2): 255-268, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32390600

RESUMEN

PURPOSE: DNA methylation plays an important role in thyroid oncogenesis. The aim of this study was to investigate the connection between global and local DNA methylation status and to establish the levels of important DNA methylation regulators (TET family and DNMT1) in thyroid tumours: follicular adenoma-FA, papillary thyroid carcinoma-PTC (classic papillary thyroid carcinoma-cPTC and papillary thyroid carcinoma follicular variant fvPTC). METHODS: Global DNA methylation profile in thyroid tumours tissue (41 paired samples) was assessed by 5-methylcytosine and 5-hydroxymethylcytosine levels evaluation (ELISA), along with TETs and DNMT1 genes expression quantification. Also, it was investigated for the first time TET1 and TET2 promoter's methylation in thyroid tumours. BRAF V600E mutation and RET/PTC translocation testing were performed on all investigated samples. In vitro studies upon DNA methylation in K1 thyroid cancer cells were performed with demethylating agents (5-AzaC and vitamin C). RESULTS: TET1 and TET2 displayed a significantly reduced gene expression level in PTC, while DNMT1 gene presented a high level of expression. PTC samples presented increased levels of 5-methylcytosine and low levels of 5-hydroxymethylcytosine. 5-methylcytosine levels were associated with TET1/TET2 expression levels. TET1 gene expression was significantly lower in patients positive for BRAF mutation and with RET/PTC rearrangement. TET2 gene was found hypermethylated in thyroid carcinoma patients overall, especially in PTC-follicular variant samples (p= 0.0002), where TET2 gene expression levels were significantly reduced (p= 0.0031). Furthermore, the data indicate for all thyroid cancer patients a good sensitivity (81.08%) and specificity (86.49%) regarding the use of TET1 (p< 0.0001), and TET2 (71.79%, 64.10%, p= 0.0001) hypermethylation as biomarkers for thyroid oncogenesis. CONCLUSIONS: These results suggest that TET1/TET2 gene expression and methylation may serve as potential diagnostic tools for thyroid neoplasia. Our study showed that the methylation of TET1 increases in malignant thyroid tumours. fvPTC patients presented lower methylation levels compared to cPTC and could be a discriminatory factor between two cancer types and benign lesions. TET2 is a poorer discriminator between FA and fvPTC, but it can be useful for cPTC identification. K1-cells treated with demethylating agents showed a demethylation effect, especially upon TET2 gene. The cumulative effect of L-AA and 5-AzaC proved to have a potent combined demethylating effect on genes promoter's activation and could open new perspectives for thyroid cancer therapy.


Asunto(s)
Adenocarcinoma Folicular/genética , Biomarcadores de Tumor/genética , Metilación de ADN/genética , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética , Adenocarcinoma Folicular/diagnóstico , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/patología , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Azacitidina/farmacología , Azacitidina/uso terapéutico , Carcinogénesis/genética , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1/genética , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Dioxigenasas , Ensayos de Selección de Medicamentos Antitumorales , Epigénesis Genética/efectos de los fármacos , Estudios de Factibilidad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Oxigenasas de Función Mixta/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Sensibilidad y Especificidad , Cáncer Papilar Tiroideo/diagnóstico , Cáncer Papilar Tiroideo/tratamiento farmacológico , Cáncer Papilar Tiroideo/patología , Glándula Tiroides/patología , Neoplasias de la Tiroides/diagnóstico , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/patología , Adulto Joven
18.
Exp Cell Res ; 393(2): 112095, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32442537

RESUMEN

Aberrant protein glycosylation is involved in many diseases including cancer. This study investigated the role of fucosyltransferase VII (FUT7) in the progression of follicular thyroid carcinoma (FTC). FUT7 expression was found to be upregulated in FTC compared to paracancerous thyroid tissue, and in FTC with T2 stage of TMN classification compared to FTC with T1 stage. FUT7 overexpression promoted cell proliferation, epithelial-mesenchymal transition (EMT), and the migration and invasion of primary FTC cell line FTC-133. Consistently, FUT7 knock-down inhibited cell proliferation, EMT, as well as the migration and invasion of the metastatic FTC cell line FTC-238. Mechanistic investigation revealed that FUT7 catalyzed the α1,3-fucosylation of epidermal growth factor receptor (EGFR) in FTC cells. The extent of glycan α1,3-fucosylation on EGFR was positively correlated with the activation of EGFR in the presence/absence of epidermal growth factor (EGF) treatment. Furthermore, FUT7 was shown to enhance EGF-induced progression of FTC cells through mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathways. These findings provide a new perspective on FUT7 that may be a novel diagnostic and therapeutic target of FTC.


Asunto(s)
Adenocarcinoma Folicular/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Fucosiltransferasas/metabolismo , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/patología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Regulación hacia Arriba/efectos de los fármacos
19.
Clin Cancer Res ; 26(11): 2524-2534, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32144134

RESUMEN

PURPOSE: In this phase I study (NCT01307267), we evaluated safety, pharmacokinetics, clinical activity, and pharmacodynamics of treatment with utomilumab plus rituximab in patients with relapsed/refractory follicular lymphoma (FL) and other CD20+ non-Hodgkin lymphomas (NHL). PATIENTS AND METHODS: Primary objectives were to assess treatment safety and tolerability for estimating the MTD, using a modified time-to-event continual reassessment method, and selecting the recommended phase II dose (RP2D). RESULTS: Sixty-seven patients received utomilumab (0.03-10.0 mg/kg every 4 weeks) and rituximab (375 mg/m2 weekly) in the dose-escalation groups or utomilumab (1.2 mg/kg every 4 weeks) plus rituximab in the dose-expansion cohort. No patient experienced dose-limiting toxicity. The MTD for utomilumab in combination with rituximab was not reached and estimated to be ≥10 mg/kg every 4 weeks. The majority of the utomilumab treatment-related adverse events (AE) were grade 1 to 2; the most common AE was fatigue (16.4%). The pharmacokinetics of utomilumab in combination with rituximab was linear in the 0.03 to 10 mg/kg dose range. A low incidence (1.5%) of treatment-induced antidrug antibodies against utomilumab was observed. The objective response rate was 21.2% (95% CI, 12.1%-33.0%) in all patients with NHL, including four complete and 10 partial responses. Analysis of paired biopsies from a relapsed/refractory FL patient with complete response showed increased T-cell infiltration and cytotoxic activity in tumors. Biomarker correlations with outcomes suggested that clinical benefit may be contingent on patient immune function. CONCLUSIONS: Utomilumab in combination with rituximab demonstrated clinical activity and a favorable safety profile in patients with CD20+ NHLs.


Asunto(s)
Adenocarcinoma Folicular/tratamiento farmacológico , Antígenos CD20/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfoma no Hodgkin/tratamiento farmacológico , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Adenocarcinoma Folicular/inmunología , Adenocarcinoma Folicular/patología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/administración & dosificación , Femenino , Estudios de Seguimiento , Humanos , Inmunoglobulina G/administración & dosificación , Linfoma no Hodgkin/inmunología , Linfoma no Hodgkin/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Pronóstico , Rituximab/administración & dosificación , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...