Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Toxicology ; 509: 153937, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39216546

ABSTRACT

For many years overnight fasting of rats before the collection of clinical pathology blood samples and necropsy has been a common procedure in toxicological studies for regulatory purposes. The fasting was thought to minimize the intragroup variability for clinical pathology and organ weights. However, depriving rats of food overnight will impact animal welfare by interfering with the general metabolism and may result in physiological and behavioural changes. The effects of overnight fasting in comparison to rats that were not fasted prior to necropsy was investigated in lactating rats based on an evaluation of organ weights, haematological, and clinical biochemical parameters. The results of 92 OECD 422 studies were analysed (i.e., Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test) of which approximately half of the studies incorporated fasting prior to necropsy and the other half did not. Terminal body and organ weights from all 92 studies were evaluated. Clinical pathology was included in 78 of these 92 studies. Decreased glucose levels following fasting had been reported in the literature but were not observed when comparing 39 studies with fasted conditions versus 39 studies with non-fasted conditions. Both literature and the analysed database exhibited a reduction in liver weight, alanine aminotransferase, and alkaline phosphatase levels in overnight fasted groups. These differences between fasted and non-fasted states are considered of little account as study results are always interpreted based on the differences in parameter values between treated animals compared to control animals within a study. Contrarily to previously suggested, intragroup variability was lower in the majority of parameters in non-fasted animals. According to laboratory historical data, clinical pathology and organ weight parameters are found to be very similar in male and female rats, indicating that the results of this study may be extrapolatable to non-lactating female and male rats. Based on these comparisons, it is recommended not to fast rats prior to necropsy but to continue feeding all rats ad libitum, to minimize physiological changes in these animals, to reduce variability, improve animal welfare and thereby improve the scientific value of study results.

2.
Oncol Lett ; 22(2): 590, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34149901

ABSTRACT

Differentiated thyroid cancer (DTC) is the most frequent endocrine tumor with a good prognosis after primary treatment in most cases. By contrast, 30-40% of patients with metastatic DTC are unresponsive to 131I radioactive iodide (RAI) treatment due to tumor dedifferentiation. Currently, underlying molecular mechanisms of dedifferentiation remain elusive and predictive biomarkers are lacking. Therefore, the present study aimed to identify molecular biomarkers in primary tumors associated with RAI refractoriness. A retrospective cohort was gathered consisting of RAI-sensitive patients with DTC and RAI-refractory patients with poorly DTC. In all patients, extensive intratumoral mutation profiling, gene fusions analysis, telomerase reverse transcriptase (TERT) promoter mutation analysis and formalin-fixed paraffin-embedded-compatible RNA sequencing were performed. Genetic analyses revealed an increased mutational load in RAI-refractory DTC, including mutations in AKT1, PTEN, TP53 and TERT promoter. Transcriptomic analyses revealed profound differential expression of insulin-like growth factor 2 (IGF2), with up to 100-fold higher expression in RAI-refractory DTC compared with in RAI-sensitive DTC cases. ELISA revealed significant lower IGF2 plasma concentrations after surgery and subsequent 131I RAI therapy in patients with DTC compared with pretreatment baseline. Overall, the current findings suggested that the tumor-promoting growth factor IGF2 may have a potential role in acquiring RAI refractoriness.

3.
Cell Oncol (Dordr) ; 44(3): 611-625, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33534128

ABSTRACT

PURPOSE: Non-medullary thyroid cancer (NMTC) treatment is based on the ability of thyroid follicular cells to accumulate radioactive iodide (RAI). However, in a subset of NMTC patients tumor dedifferentiation occurs, leading to RAI resistance. Digoxin has been demonstrated to restore iodide uptake capacity in vitro in poorly differentiated and anaplastic NMTC cells, termed redifferentiation. The aim of the present study was to investigate the in vivo effects of digoxin in TPO-Cre/LSL-BrafV600E mice and digoxin-treated NMTC patients. METHODS: Mice with thyroid cancer were subjected to 3D ultrasound for monitoring tumor growth and 124I PET/CT for measurement of intratumoral iodide uptake. Post-mortem analyses on tumor tissues comprised gene expression profiling and measurement of intratumoral autophagy activity. Through PALGA (Dutch Pathology Registry), archived tumor material was obtained from 11 non-anaplastic NMTC patients who were using digoxin. Clinical characteristics and tumor material of these patients were compared to 11 matched control NMTC patients never treated with digoxin. RESULTS: We found that in mice, tumor growth was inhibited and 124I accumulation was sustainably increased after short-course digoxin treatment. Post-mortem analyses revealed that digoxin treatment increased autophagy activity and enhanced expression of thyroid-specific genes in mouse tumors compared to vehicle-treated mice. Digoxin-treated NMTC patients exhibited significantly higher autophagy activity and a higher differentiation status as compared to matched control NMTC patients, and were associated with favourable clinical outcome. CONCLUSIONS: These in vivo data support the hypothesis that digoxin may represent a repositioned adjunctive treatment modality that suppresses tumor growth and improves RAI sensitivity in patients with RAI-refractory NMTC.


Subject(s)
Digoxin/therapeutic use , Iodine Radioisotopes/therapeutic use , Radiation Tolerance/drug effects , Thyroid Cancer, Papillary/therapy , Thyroid Neoplasms/therapy , Aged , Aged, 80 and over , Animals , Autophagy/drug effects , Cell Proliferation/drug effects , Female , Humans , Male , Mice , Radiation-Sensitizing Agents/therapeutic use
4.
Ann Transl Med ; 8(16): 1028, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32953828

ABSTRACT

Tumor associated macrophages (TAMs) are important components of the tumor microenvironment (TME). They are characterized by a remarkable functional plasticity, thereby mostly promoting cancer progression. Changes in immune cell metabolism are paramount for this functional adaptation. Here, we review the functional consequences of the metabolic programming of TAMs and the influence of local and systemic targeted therapies on the metabolic characteristics of the TME that shape the functional phenotype of the TAMs. Understanding these metabolic changes within the context of the cross-talk between the different components of the TME including the TAMs and the tumor cells is an essential step that can pave the way towards identifications of ways to improve responses to different treatments, to overcome resistance to treatments, tumor progression and reduce treatment-specific toxicity.

5.
Endocr Connect ; 9(11): 1065-1074, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33112820

ABSTRACT

The PI3K-Akt-mTOR pathway plays a central role in the development of non-medullary thyroid carcinoma (NMTC). Although somatic mutations have been identified in these genes in NMTC patients, the role of germline variants has not been investigated. Here, we selected frequently occurring genetic variants in AKT1, AKT2, AKT3, PIK3CA and MTOR and have assessed their effect on NMTC susceptibility, progression and clinical outcome in a Dutch discovery cohort (154 patients, 188 controls) and a Romanian validation cohort (159 patients, 260 controls). Significant associations with NMTC susceptibility were observed for AKT1 polymorphisms rs3803304, rs2494732 and rs2498804 in the Dutch discovery cohort, of which the AKT1 rs3803304 association was confirmed in the Romanian validation cohort. No associations were observed between PI3K-Akt-mTOR polymorphisms and clinical parameters including histology, TNM staging, treatment response and clinical outcome. Functionally, cells bearing the associated AKT1 rs3803304 risk allele exhibit increased levels of phosphorylated Akt protein, potentially leading to elevated signaling activity of the oncogenic Akt pathway. All together, germline encoded polymorphisms in the PI3K-Akt-mTOR pathway could represent important risk factors in development of NMTC.

6.
J Nucl Med ; 59(5): 780-786, 2018 05.
Article in English | MEDLINE | ID: mdl-29242405

ABSTRACT

Anaplastic thyroid cancer (ATC) is a rare malignancy that accounts for 1%-2% of all thyroid cancers. ATC is one of the most aggressive human cancers, with rapid growth, tumor invasion, and development of distant metastases. The median survival is only 5 mo, and the 1-y survival is less than 20%. Moreover, as a result of severe dedifferentiation, including the loss of human sodium iodide symporter (hNIS) expression, radioactive iodide (RAI) therapy is ineffective. Recently, we have demonstrated beneficial effects of autophagy-activating digitalislike compounds (DLCs) on redifferentiation and concomitant restoration of iodide uptake in RAI-refractory papillary and follicular thyroid cancer cell lines. In the current study, the effects of DLCs on differentiation and proliferation of ATC cell lines were investigated. Methods: Autophagy activity was assessed in ATC patient tissues by immunofluorescent staining for the autophagy marker microtubule-associated protein 1A/1B-light chain 3 (LC3). In addition, the effect of autophagy-activating DLCs on the proliferation, gene expression profile, and iodide uptake capacity of ATC cell lines was studied. Results: Diminished autophagy activity was observed in ATC tissues, and in vitro treatment of ATC cell lines with DLCs robustly restored hNIS and thyroglobulin expression and iodide uptake capacity. In addition, proliferation was strongly reduced by induction of cell cycle arrest and, to some extent, cell death. Mechanistically, reactivation of functional hNIS expression could be attributed to activation of the transcription factors activating transcription factor 3 and protooncogene c-fosConclusion: DLCs could represent a promising adjunctive therapy for restoring iodide avidity within the full spectrum from RAI-refractory dedifferentiated to ATC.


Subject(s)
Digitalis/chemistry , Iodides/metabolism , Symporters/metabolism , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Carcinoma, Anaplastic/metabolism , Activating Transcription Factor 3/metabolism , Adenocarcinoma, Follicular/drug therapy , Adenocarcinoma, Follicular/metabolism , Autophagy , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Humans , Iodine Radioisotopes , Microscopy, Fluorescence , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fos/metabolism , Thyroglobulin/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism
7.
Mol Cancer Ther ; 16(1): 169-181, 2017 01.
Article in English | MEDLINE | ID: mdl-27837029

ABSTRACT

Up to 20%-30% of patients with metastatic non-medullary thyroid cancer have persistent or recurrent disease resulting from tumor dedifferentiation. Tumor redifferentiation to restore sensitivity to radioactive iodide (RAI) therapy is considered a promising strategy to overcome RAI resistance. Autophagy has emerged as an important mechanism in cancer dedifferentiation. Here, we demonstrate the therapeutic potential of autophagy activators for redifferentiation of thyroid cancer cell lines. Five autophagy-activating compounds, all known as digitalis-like compounds, restored hNIS expression and iodide uptake in thyroid cancer cell lines. Upregulation of hNIS was mediated by intracellular Ca2+ and FOS activation. Cell proliferation was inhibited by downregulating AKT1 and by induction of autophagy and p21-dependent cell-cycle arrest. Digitalis-like compounds, also designated as cardiac glycosides for their well-characterized beneficial effects in the treatment of heart disease, could therefore represent a promising repositioned treatment modality for patients with RAI-refractory thyroid carcinoma. Mol Cancer Ther; 16(1); 169-81. ©2016 AACR.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/drug effects , Calcium/metabolism , Digitalis/chemistry , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/drug effects , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Activating Transcription Factor 3/metabolism , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cluster Analysis , Gene Expression Profiling , Humans , Symporters/metabolism , Thyroid Neoplasms/genetics , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL