Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
Int J Mol Sci ; 25(19)2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39409128

ABSTRACT

Papillary thyroid cancer (PTC) is the 8th most common cancer among women overall. Licorice contains over 300 active compounds, many of them with anti-cancer properties. Glycyrrhetinic acid (GA) is a major component of licorice. The aim of this study was to investigate the potential anti-proliferative effects of licorice and GA on PTC cell cultures. Licorice extract (LE) was produced from the root and tested on BCPAP and K1 cell lines, as well as GA and aldosterone. We used the MTT test to investigate the anti-proliferative activity, the wound healing test for the migratory activity, and finally, we analyzed cell cycle distribution, apoptosis, and oxidative stress after LE, GA, or aldosterone incubation. Both LE and GA reduced cell viability at 48 h and cell migration at 24 h in both PTC cultures. Aldosterone reduced cell migration only in K1 cells. LE and GA induced cell cycle arrest in the G0/G1 phase in the BCPAP cell line, while LE and aldosterone induced it in the K1 culture. GA but not LE increased the apoptosis rate in both cell lines, whereas LE but not GA increased oxidative stress in both cultures. This study presents the first evidence of the in vitro anti-proliferative and anti-migratory activity of LE and GA on PTC.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Glycyrrhetinic Acid , Glycyrrhiza , Plant Extracts , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Cell Movement/drug effects , Cell Proliferation/drug effects , Glycyrrhiza/chemistry , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/drug therapy , Glycyrrhetinic Acid/pharmacology , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Line, Tumor , Apoptosis/drug effects , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/drug therapy , Oxidative Stress/drug effects , Cell Survival/drug effects , Cell Cycle Checkpoints/drug effects
2.
Int J Mol Sci ; 25(19)2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39408974

ABSTRACT

Papillary thyroid cancer (PTC) is one of the most treatable forms of cancer, with many cases being fully curable. However, resistance to anticancer drugs often leads to metastasis or recurrence, contributing to the failure of cancer therapy and, ultimately, patient mortality. The mechanisms underlying molecular differences in patients with metastatic or recurrent PTC, particularly those resistant to anticancer drugs through epigenetic reprogramming, remain poorly understood. Consequently, refractory PTC presents a critical challenge, and effective therapeutic strategies are urgently needed. Therefore, this study aimed to identify small-molecule inhibitors to enhance treatment efficacy in lenvatinib-resistant PTC. We observed an increase in sarco/endoplasmic reticulum calcium ATPase (SERCA) levels in patient-derived lenvatinib-resistant PTC cells compared with lenvatinib-sensitive ones, highlighting its potential as a therapeutic target. We subsequently identified two SERCA inhibitors [candidates 40 (isoflurane) and 42 (ethacrynic acid)] through in silico screening. These candidates demonstrated significant tumor shrinkage in a xenograft tumor model and reduced cell viability in patient-derived lenvatinib-resistant PTC cells when used in combination with lenvatinib. Our findings have potential clinical value for the development of new combination therapies to effectively target highly malignant, anticancer drug-resistant cancers.


Subject(s)
Drug Resistance, Neoplasm , Phenylurea Compounds , Quinolines , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Thyroid Cancer, Papillary , Thyroid Neoplasms , Xenograft Model Antitumor Assays , Humans , Quinolines/pharmacology , Quinolines/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Animals , Drug Resistance, Neoplasm/drug effects , Mice , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Cell Line, Tumor , Female , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Mice, Nude , Cell Proliferation/drug effects
3.
Cell Death Dis ; 15(9): 708, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349421

ABSTRACT

Adiponectin, a unique adipose-derived factor, is significantly downregulated in obesity, making it a crucial target for tumor-related metabolic research. AdipoRon is a novel adiponectin receptor agonist with the advantages of a small molecular weight, high stability and a long half-life. By screening the cervical adipose tissue of papillary thyroid carcinoma (PTC) patients with adipokine antibody array, we found that adiponectin was a potential correlation factor between obesity and PTC progression. AdipoRon has oral activity and is easily absorbed and delivered to target tissues. The effects of AdipoRon on thyroid cancer have not been reported. In this study, we identified adiponectin receptor 1 (AdipoR1) and AdipoR2 on the surface of thyroid cancer cell lines. AdipoRon inhibited the proliferation and migration of thyroid cancer cells, limited energy metabolism in thyroid cancer cells, promoted differentiation of thyroid cancer cells, and induced autophagy and apoptosis. Mechanistic studies revealed that AdipoRon inhibited p-mTOR Ser2448 and p-p70S6K Thr389, and activated ULK1 and p-ULK1. ULK1 knockdown suppressed the effect of AdipoRon on LC3BII/I protein and lysosomes. AdipoR2 knockdown reduced AdipoRon-induced autophagy in thyroid cancer cells. This study is the first to demonstrate the role of AdipoRon in PTC. Our findings illustrate a previously unknown function and mechanism of the AdipoRon-AdipoR2-ULK/p-ULK1 axis in PTC and lay the foundation for clinical translation of AdipoRon to PTC. Targeting the AdipoRon-AdipoR2-ULK/p-ULK1 axis may represent a new therapeutic strategy for PTC.


Subject(s)
Autophagy-Related Protein-1 Homolog , Autophagy , Cell Proliferation , Obesity , Receptors, Adiponectin , Thyroid Neoplasms , Humans , Receptors, Adiponectin/metabolism , Receptors, Adiponectin/agonists , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , Obesity/drug therapy , Obesity/metabolism , Cell Line, Tumor , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy/drug effects , Cell Proliferation/drug effects , Female , Cell Movement/drug effects , Male , Apoptosis/drug effects , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/genetics , Signal Transduction/drug effects , Animals , Middle Aged , TOR Serine-Threonine Kinases/metabolism , Adiponectin/metabolism , Piperidines , Intracellular Signaling Peptides and Proteins
4.
J Nanobiotechnology ; 22(1): 571, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294724

ABSTRACT

Thyroid cancer is one of the most common endocrine malignancies in clinical practice. Traditional surgery and radioactive iodine ablation have poor treatment results for poorly differentiated thyroid cancer, and there is a risk of metastasis and recurrence. In this study, caffeic acid, a natural herbal extract with certain biological activity, has been as precursor to prepare new caffeic acid carbon nanodots via a one-step hydrothermal method. The caffeic acid carbon nanodots retains part of the structure and biological activity of caffeic acid, and have good biocompatibility, water solubility and stability. The construction of the carbon nanodots could effectively improve their bio-absorption rate and the efficacy. In vitro cell experiments showed that low-dose caffeic acid carbon nanodots had a significant inhibitory effect on poorly differentiated papillary thyroid carcinoma BCPAP cells. At low concentrations of 16 µg/mL, the inhibition rate of human thyroid cancer cells BCPAP was ~ 79%. The anti-tumor mechanism was predicted and verified by transcriptome, real-time quantitative PCR and western blot experiments. The caffeic acid carbon nanodots showed to simultaneously downregulate the expression of KRAS, p-BRAF, p-MEK1 and p-ERK1/2, the four continuous key proteins in a MAPK classical signaling pathway. In vivo experiments further confirmed the caffeic acid carbon nanodots could significantly inhibit the tumorigenicity of xenografts in papillary thyroid carcinoma at quite low doses. This piece of work provides a new nanomedicine and therapeutic strategy for highly resistant poorly differentiated papillary thyroid carcinoma.


Subject(s)
Caffeic Acids , Carbon , Mice, Nude , Thyroid Cancer, Papillary , Thyroid Neoplasms , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , Humans , Animals , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Cell Line, Tumor , Carbon/chemistry , Mice , Mice, Inbred BALB C , Drug Resistance, Neoplasm/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Female
6.
Comb Chem High Throughput Screen ; 27(14): 2151-2158, 2024.
Article in English | MEDLINE | ID: mdl-39099452

ABSTRACT

BACKGROUND: Thyroid Cancer (TC) is an endocrine organ malignancy that has become more common in recent decades. Vernodalin (VN), a cytotoxic sesquiterpene, has been reported to exhibit anticancer properties against human breast and liver cancer cells. However, no study has explored the efficacy of VN with respect to its antiproliferative and apoptotic action on human Papillary Thyroid Cancer cells (PTC). OBJECTIVE: The study intended to examine the antitumor and antiproliferative effects of VN and the apoptosis mechanisms underlying its action on TPC-1 human PTC cells. METHODS: In this study, we examined the VN cell viability by MTT assay; performed ROS measurement by DCFH staining method, MMP identification by Rh-123 staining method, and apoptotic morphological assay by employing AO/EB and DAPI stain method, and further, p38 MAPK/ERK/JNK cell proliferation markers were determined by western blotting technique. RESULTS: The findings showed that VN could inhibit the growth of PTC cells by increasing intracellular ROS, damaging MMP, and stimulating apoptosis in a concentration-dependent manner. The study demonstrated how VN inhibited TPC-1 cell viability by causing ROS-induced cell death via the MAPK signaling pathway. CONCLUSION: VN may serve as an agonist to impact apoptosis in PTC cells. In human PTC, VN could play an effective role in chemotherapy. More studies pertaining to animal tumor models are needed to prove its anti-cancer effectiveness in vivo.


Subject(s)
Apoptosis , Cell Proliferation , MAP Kinase Signaling System , Reactive Oxygen Species , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , MAP Kinase Signaling System/drug effects , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Drug Screening Assays, Antitumor , Cell Line, Tumor , Dose-Response Relationship, Drug
7.
Medicina (Kaunas) ; 60(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39064466

ABSTRACT

A 76-year-old female patient presented with an iodine-refractory papillary thyroid carcinoma (PTC), diagnosed eight years earlier, with several lymph node recurrences requiring successive surgeries. Fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) imaging revealed a new unresectable loco-regional recurrence. The patient was diagnosed with a somatic BRAF V600E mutation. Therefore, dabrafenib and trametinib combination therapy was introduced and closely monitored by a dedicated multidisciplinary team, involving pharmaceutical consultations. As early as six weeks after treatment initiation, the patient reported multiple adverse events (AEs) to the clinical pharmacy team, who provided advice on resolving AEs or improving tolerance. Close interprofessional collaboration among healthcare workers involved in the care pathway allowed for the identification of the most opportune times for temporary suspension of treatment (four suspensions over seven months) or dose reduction (two reductions over 3.5 months). This resulted in a total treatment duration (one year) longer than the average times reported in the literature. The patient showed a rapid and excellent response to treatment immediately after initiation, culminating in a complete metabolic response assessed by [18F]FDG PET/CT imaging at nine months. Twenty-five months after treatment discontinuation, the disease remained controlled. Overall, dabrafenib and trametinib combination could offer excellent outcomes in selected patients with refractory BRAF-mutated PTC, with additional clinical pharmacy initiatives allowing for the optimized management of AEs and prolonged treatment periods.


Subject(s)
Imidazoles , Oximes , Pyridones , Pyrimidinones , Thyroid Neoplasms , Humans , Female , Oximes/therapeutic use , Oximes/administration & dosage , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Pyridones/therapeutic use , Pyridones/administration & dosage , Aged , Pyrimidinones/therapeutic use , Pyrimidinones/administration & dosage , Thyroid Neoplasms/drug therapy , Positron Emission Tomography Computed Tomography/methods , Iodine Radioisotopes/therapeutic use , Iodine Radioisotopes/administration & dosage , Thyroid Cancer, Papillary/drug therapy , Treatment Outcome , Proto-Oncogene Proteins B-raf/genetics
8.
Phytomedicine ; 132: 155804, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38943696

ABSTRACT

BACKGROUND: Papillary thyroid carcinoma (PTC) is an endocrine malignant tumor of the head and neck. Surgery and chemotherapy are PTC treatments, but have adverse effects. Exploration of new non-toxic anti-PTC drugs for PTC treatment is an unmet need. METHODS: We aimed to identify anti-PTC drugs that could inhibit PTC-cell proliferation through high-throughput screening of a library of well-characterized naturally occurring small-molecule compounds. Then, the anti-PTC function of rhodiolin was validated by in vitro cell models and xenograft tumor models RESULTS: We initially demonstrated that rhodiolin inhibited the growth and induced the apoptosis of PTC cells significantly in vitro and in vivo. At the metabolic level, rhodiolin blocked glycolysis through glucose 6-phosphate isomerase (GPI), which suggested that glycolytic inhibition may be involved in mediating the anti-PTC function of rhodiolin. Transcriptomics analysis combined with bioinformatics analysis identified rhodiolin treatment to inhibit phosphorylation of the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway. Collectively, our findings demonstrated that rhodiolin inhibited the proliferation and induced the apoptosis of PTC cells by blocking glycolysis through the glycolytic enzyme GPI, thereby inhibiting phosphorylation of the PI3K/Akt/mTOR signaling pathway. CONCLUSION: Our study demonstrates the potential use of rhodiolin in inhibiting the proliferation and inducing the apoptosis of PTC cells. Inhibition of phosphorylation of the PI3K/Akt/mTOR signaling pathway mediated by GPI plays an extremely important part in the ant-PTC function of rhodiolin. These results suggest that rhodiolin is a promising drug in the treatment of PTC progression. Our results provide a novel target and cell signaling pathway for PTC therapy from the perspective of energy metabolism, which could provide new perspectives and new drug choices for PTC therapy. In addition to that, our study will help to make up for the lack of drug research for PTC.


Subject(s)
Apoptosis , Cell Proliferation , Glucose-6-Phosphate Isomerase , Glycolysis , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , TOR Serine-Threonine Kinases/metabolism , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Cell Proliferation/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Glycolysis/drug effects , Glucose-6-Phosphate Isomerase/metabolism , Apoptosis/drug effects , Mice, Nude , Mice , Mice, Inbred BALB C , Antineoplastic Agents, Phytogenic/pharmacology , Xenograft Model Antitumor Assays
9.
BMC Surg ; 24(1): 133, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702652

ABSTRACT

OBJECTIVES: While surgery plays a crucial role in treating papillary thyroid carcinoma (PTC), the potential effects of subsequent TSH suppression therapy on prognosis should not be overlooked. This study aims to investigate the factors that influence postoperative TSH suppression therapy in patients with PTC. METHODS: This study was a retrospective cohort study conducted at our hospital. It included 268 patients who underwent surgery and were pathologically diagnosed with PTC between February 2019 and February 2021. The selected patients received postoperative TSH suppression therapy. Based on the TSH level measured 12 months after surgery, the patients were divided into two groups: TSH level conforming group (n = 80) and non-conforming group (n = 188). We then compared the general clinical data, clinicopathological characteristics, preoperative laboratory test indicators, postoperative levothyroxine sodium tablet dosage, follow-up frequency, and thyroid function-related indicators between the two groups of patients. The correlation between the observed indicators and the success of TSH suppression therapy was further analyzed, leading to the identification of influencing factors for TSH suppression therapy. RESULTS: There were no statistically significant differences in general clinical data and clinicopathological characteristics between the two groups of patients (P > 0.05). The proportion of patients with preoperative TSH ≥ 2.0 mU/L was higher in the non-conforming group compared to the TSH level conforming group (P < 0.05), and the ROC curve analysis indicated that the area under the curve for the preoperative TSH index was 0.610 (P < 0.05). The proportion of patients in the TSH level conforming group who took oral levothyroxine sodium tablets at a dose of ≥ 1.4 µg/kg·d after surgery was higher (P < 0.05). The postoperative levels of FT3 and FT4 were higher in the TSH level conforming group (P < 0.05). The results of binary logistic regression analysis indicated that factors "Postoperative TSH level ≥ 2 mU/L", "Levothyroxine sodium tablet dose<1.4 µg/kg·d", and "Combined with Hashimoto thyroiditis" were significantly associated with an elevated risk of postoperative TSH levels failing to reach the target (P < 0.05). CONCLUSION: Optimal thyroid function in patients with PTC post-surgery is best achieved when adjusting the dose of levothyroxine sodium in a timely manner to reach the target TSH level during follow-up visits.


Subject(s)
Thyroid Cancer, Papillary , Thyroid Neoplasms , Thyroidectomy , Thyrotropin , Thyroxine , Humans , Retrospective Studies , Male , Female , Thyroid Cancer, Papillary/surgery , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Thyrotropin/blood , Thyrotropin/antagonists & inhibitors , Thyroid Neoplasms/surgery , Thyroid Neoplasms/drug therapy , Middle Aged , Thyroxine/therapeutic use , Thyroxine/administration & dosage , Adult , Treatment Outcome , Postoperative Period
10.
Med Oncol ; 41(6): 160, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38763968

ABSTRACT

Papillary thyroid carcinoma (PTC) is a common endocrine malignancy. The pathology of PTC is far from clear. As a kinase that can be targeted, the role of TNIK in PTC has not been investigated. This study was focused on the effects and molecular mechanisms of TNIK in PTC. Both public datasets and clinical specimens were used to verify TNIK expression. The effects of TNIK were investigated in both cell lines and mice models. Transcriptome analysis was used to explore the underlying mechanism of TNIK. Immunofluorescence, wound healing, and qRT-PCR assays were used to validate the mechanism of TNIK in PTC. The therapeutic effects of TNIK inhibitor NCB-0846 were evaluated by flow cytometry, western blot, and subcutaneous xenografts mice. TNIK expression was upregulated in PTC tissues. TNIK knockdown could suppress cell proliferation and tumor growth in no matter cell models or nude mice. The transcriptome analysis, GO enrichment analysis, and GSEA analysis results indicated TNIK was highly correlated with cytoskeleton, cell motility, and Wnt pathways. The mechanistic studies demonstrated that TNIK regulated cytoskeleton remodeling and promoted cell migration. NCB-0846 significantly inhibited TNIK kinase activity, induced cell apoptosis, and activated apoptosis-related proteins in a dose-dependent manner. In addition, NCB-0846 inhibited tumor growth in tumor-bearing mice. In summary, we proposed a novel regulatory mechanism in which TNIK-mediated cytoskeleton remodeling and cell migration to regulate tumor progression in PTC. TNIK is a therapeutic target in PTC and NCB-0846 would act as a novel targeted drug for PTC therapy.


Subject(s)
Cell Proliferation , Thyroid Cancer, Papillary , Thyroid Neoplasms , Animals , Female , Humans , Male , Mice , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Mice, Inbred BALB C , Mice, Nude , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/genetics , Xenograft Model Antitumor Assays
11.
Neoplasia ; 52: 100996, 2024 06.
Article in English | MEDLINE | ID: mdl-38593698

ABSTRACT

Papillary thyroid carcinoma (PTC) is the most common endocrine malignancy, and its incidence has increased rapidly in recent years. The BRAF inhibitor vemurafenib is effective against BRAFV600E-positive PTC; however, acquired resistance to single agent therapy frequently leads to tumor recurrence and metastasis, underscoring the need to develop tailored treatment strategies. We previously showed that the oncogenic kinase PIM1 was associated with the malignant phenotype and prognosis of PTC. In this study, we showed that sustained expression of the PIM1 protein in PTC was affected by the BRAFV600E mutation. Based on this regulatory mechanism, we tested the synergistic effects of inhibitors of BRAF (BRAFi) and PIM1 in BRAFV600E-positive PTC cell lines and xenograft tumors. LC-MS metabolomics analyses suggested that BRAFi/PIMi therapy acted by restricting the amounts of critical amino acids and nucleotides required by cancer cells as well as modulating DNA methylation. This study elucidates the role of BRAFV600E in the regulation of PIM1 in PTC and demonstrates the synergistic effect of a novel combination, BRAFi/PIMi, for the treatment of PTC. This discovery, along with the pathways that may be involved in the powerful efficacy of BRAFi/PIMi strategy from the perspective of cell metabolism, provides insight into the molecular basis of PTC progression and offers new perspectives for BRAF-resistant PTC treatment.


Subject(s)
Drug Synergism , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-pim-1 , Thyroid Cancer, Papillary , Thyroid Neoplasms , Animals , Humans , Mice , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Sci Rep ; 14(1): 7853, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38570592

ABSTRACT

Thyroid cancer is the most common endocrine carcinoma and, among its different subtypes, the papillary subtype (PTC) is the most frequent. Generally, PTCs are well differentiated, but a minor percentage of PTCs are characterized by a worse prognosis and more aggressive behavior. Phytochemicals, naturally found in plant products, represent a heterogeneous group of bioactive compounds that can interfere with cell proliferation and the regulation of the cell cycle, taking part in multiple signaling pathways that are often disrupted in tumor initiation, proliferation, and progression. In this work, we focused on 15,16-dihydrotanshinone I (DHT), a tanshinone isolated from Salvia miltiorrhiza Bunge (Danshen). We first evaluated DHT biological effect on PTC cells regarding cell viability, colony formation ability, and migration capacity. All of these parameters were downregulated by DHT treatment. We then investigated gene expression changes after DHT treatment by performing RNA-seq. The analysis revealed that DHT significantly reduced the Wnt signaling pathway, which plays a role in various diseases, including cancer. Finally, we demonstrate that DHT treatment decreases protein levels of ß-catenin, a final effector of canonical Wnt signaling pathway. Overall, our data suggest a possible use of this nutraceutical as an adjuvant in the treatment of aggressive papillary thyroid carcinoma.


Subject(s)
Carcinoma, Papillary , Furans , Phenanthrenes , Quinones , Thyroid Neoplasms , Humans , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , beta Catenin/genetics , beta Catenin/metabolism , Down-Regulation , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Cell Line, Tumor , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Wnt Signaling Pathway/genetics , Cell Proliferation/physiology , Cell Movement/genetics
13.
Toxicology ; 504: 153807, 2024 May.
Article in English | MEDLINE | ID: mdl-38641160

ABSTRACT

Decabromodiphenyl ether (BDE209) has been demonstrated to be associated with thyroid dysfunction and thyroid carcinoma risk as a widely used brominated flame retardants. Although dabrafenib has been confirmed to be a promising therapeutic agent for papillary thyroid carcinoma (PTC) harboring BRAFV600E mutation, the rapid acquired dabrafenib resistance has brought a great challenge to clinical improvement and the underpinning mechanisms remain poorly defined. By treating PTC-derived and normal follicular epithelial cell lines with BDE209, we assessed its impact on the MAPK pathway's activation and evaluated the resultant effects on cell viability and signaling pathways, utilizing methods such as Western blot, IF staining, and RNA-seq bioinformatic analysis. Our findings reveal that BDE209 exacerbates MAPK activation, undermining dabrafenib's inhibitory effects by triggering the EGFR pathway, thereby highlighting BDE209's potential to diminish the pharmacological efficacy of dabrafenib in treating BRAF-mutated PTC. This research underscores the importance of considering environmental factors like BDE209 exposure in the effective management of thyroid carcinoma treatment strategies.


Subject(s)
ErbB Receptors , Halogenated Diphenyl Ethers , Imidazoles , Mutation , Oximes , Proto-Oncogene Proteins B-raf , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Proto-Oncogene Proteins B-raf/genetics , Halogenated Diphenyl Ethers/toxicity , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Oximes/pharmacology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Imidazoles/pharmacology , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents/pharmacology , MAP Kinase Signaling System/drug effects , Cell Survival/drug effects
14.
Cancer Res Treat ; 56(4): 1270-1276, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38453274

ABSTRACT

PURPOSE: BRAF mutations are detected in 30%-80% of papillary thyroid cancer (PTC) cases. DaBRAFenib and trametinib showed promising antitumor activity in patients with BRAFV600E-mutated metastatic melanoma and non-small cell lung cancer. This study aimed to evaluate the efficacy and safety of daBRAFenib and trametinib in patients with metastatic BRAFV600E-mutated thyroid cancer. MATERIALS AND METHODS: This was a retrospective study to evaluate the efficacy of daBRAFenib and trametinib in patients with metastatic BRAFV600E-mutated PTC. The patients received daBRAFenib 150 mg twice daily and trametinib 2 mg once daily at the Samsung Medical Center. This study evaluated the progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR) overall survival (OS), and safety of daBRAFenib and trametinib. RESULTS: Between December 2019 and January 2022, 27 PTC patients including eight patients with poorly differentiated or anaplastic transformation, received daBRAFenib and trametinib. The median age was 73.0 years, and the median follow-up period was 19.8 months. The majority (81.5%) had undergone thyroidectomy, while 8 patients had received prior systemic treatments. ORR was 73.1%, with 19 partial responses, and DCR was 92.3%. Median PFS was 21.7 months, and median OS was 21.7 months. Treatment-related adverse events included generalized weakness (29.6%), fever (25.9%), and gastrointestinal problems (22.2%). Dose reduction due to adverse events was required in 81.5% of the patients. CONCLUSION: DaBRAFenib and trametinib demonstrated a high ORR with promising PFS; however, most patients with BRAFV600E-mutated metastatic PTC required a dose reduction.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Imidazoles , Mutation , Oximes , Proto-Oncogene Proteins B-raf , Pyridones , Pyrimidinones , Thyroid Neoplasms , Humans , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Pyridones/administration & dosage , Pyridones/therapeutic use , Pyridones/adverse effects , Oximes/administration & dosage , Oximes/therapeutic use , Pyrimidinones/administration & dosage , Pyrimidinones/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Male , Female , Aged , Retrospective Studies , Middle Aged , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/mortality , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aged, 80 and over , Adult , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Neoplasm Metastasis
15.
J Ethnopharmacol ; 326: 117912, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38387682

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Papillary thyroid carcinoma (PTC) is the predominant form of thyroid cancer with a rising global incidence. Despite favorable prognoses, a significant recurrence rate persists. Dioscorea bulbifera L. (DBL), a traditional Chinese medicine, has been historically used for thyroid-related disorders. However, its therapeutic effects and mechanisms of action on PTC remain unclear. AIM OF THE STUDY: To explore the potential therapeutic effects, principal active components, and molecular mechanisms of DBL in the treatment of PTC through network pharmacology and molecular docking, with experimental validation conducted to corroborate these findings. MATERIALS AND METHODS: The Traditional Chinese Medicine Systems Pharmacology Database (TCMSP) was utilized as a systematic tool for collecting and screening the phytochemical components of DBL, and for establishing associations between these components and molecular targets. Based on this, network data was visually processed using Cytoscape software (version 3.8.0). Concurrently, precise molecular docking studies of the principal active components of DBL and their corresponding targets were conducted using Autodock software. Additionally, PTC-related genes were selected through the GeneCards and GEO databases. We further employed the DAVID bioinformatics resources to conduct comprehensive Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on the intersecting genes between DBL and PTC. These analyses aid in predicting the potential therapeutic actions of DBL on PTC and its mechanisms of action. To validate these findings, corresponding in vitro experimental studies were also conducted. RESULTS: In this investigation, 14 bioactive compounds of DBL and 195 corresponding molecular targets were identified, with 127 common targets shared between DBL and PTC. Molecular docking revealed strong binding affinities between major bioactive compounds and target proteins. GO enrichment analysis unveiled key processes involved in DBL's action. KEGG analysis highlighted DBL's modulation of the PI3K/AKT signaling pathway. Experimental outcomes demonstrated DBL's potential in inhibiting PTC cell proliferation and migration, suppressing PI3K/AKT pathway activation, and promoting ferroptosis. CONCLUSION: In conclusion, DBL offers a multifaceted therapeutic approach for PTC, targeting multiple molecular entities and influencing diverse biological pathways. Network pharmacology and molecular docking shed light on DBL's potential utility in PTC treatment, substantiated by experimental validation. This study contributes valuable insights into using DBL as a promising therapeutic agent for PTC management.


Subject(s)
Dioscorea , Drugs, Chinese Herbal , Ferroptosis , Thyroid Neoplasms , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , Network Pharmacology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt , Molecular Docking Simulation , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use
16.
Biomed Pharmacother ; 173: 116324, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38422655

ABSTRACT

Oxidative stress (OS) is recognized as a contributing factor in the development and progression of thyroid cancer. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a pivotal transcription factor involved in against OS generated by excessive reactive oxygen species (ROS). It governs the expression of a wide array of genes implicated in detoxification and antioxidant pathways. However, studies have demonstrated that the sustained activation of Nrf2 can contribute to tumor progression and drug resistance in cancers. The expression of Nrf2 was notably elevated in papillary thyroid cancer tissues compared to normal tissues, indicating that Nrf2 may play an oncogenic role in the development of papillary thyroid cancer. Nrf2 and its downstream targets are involved in the progression of thyroid cancer by impacting the prognosis and ferroptosis. Furthermore, the inhibition of Nrf2 can increase the sensitivity of target therapy in thyroid cancer. Therefore, Nrf2 appears to be a potential therapeutic target for the treatment of thyroid cancer. This review summarized current data on Nrf2 expression in thyroid cancer, discussed the function of Nrf2 in thyroid cancer, and analyzed various strategies to inhibit Nrf2.


Subject(s)
NF-E2-Related Factor 2 , Thyroid Neoplasms , Humans , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , NF-E2-Related Factor 2/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Oxidative Stress , Antioxidants/metabolism , Reactive Oxygen Species/metabolism
17.
Oncologist ; 29(1): 84-88, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38037189

ABSTRACT

Herein, we present a misleading case of advanced papillary thyroid carcinoma with lung, node, and pleural metastases, initially diagnosed as metastatic lung adenocarcinoma with papillary features, based on the histological and immunohistochemical analysis of a pleural biopsy. Between August 2019 and August 2020, the patient received 2 ineffective lines of systemic therapy, including a first line of chemotherapy with cisplatin and pemetrexed, and a second line of immunotherapy with atezolizumab. Comprehensive genomic profiling by next-generation sequencing on the archival pleural biopsy revealed an NTRK1-TMP3 fusion and comutation of the TERT promoter, commonly found in papillary thyroid carcinoma. After palliative partial thyroidectomy that confirmed the diagnosis of papillary thyroid carcinoma, in February 2021, the patient was enrolled in the STARTRK-2 GO40782 basket trial and received entrectinib, an oral pan-TRK inhibitor specifically targeting NTRK-rearranged tumors. After initially experiencing drug-related grade 2 anorexia, dysgeusia, and neurotoxicity and grade 3 asthenia, the dose was reduced, and an excellent and durable objective response was observed.


Subject(s)
Receptor, trkA , Thyroid Neoplasms , Humans , Receptor, trkA/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/drug therapy , Protein Kinase Inhibitors/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics
18.
Nanomedicine (Lond) ; 18(27): 2039-2059, 2023 11.
Article in English | MEDLINE | ID: mdl-38131284

ABSTRACT

Aim: This study aimed to identify molecular markers associated with papillary thyroid cancer (PTC) and investigate the therapeutic potential of targeted nanoscale drugs. Materials & methods: We analyzed the effects of circICA1 and miR-486-3p on B-CPAP cells' proliferation, apoptosis, migration and invasion. The regulation of the miR-486-3p/SERPINA1 axis was explored using quantitative real-time reverse transcription PCR and western blot analyses for metastasis. In vivo, we evaluated the effects of hyperbranched polyamidoamine-RGD peptide/si-circICA1 on PTC growth and metastasis. Results: Enhanced miR-486-3p expression inhibits B-CPAP cells' proliferation and invasion. si-circICA1 delivered via hyperbranched polyamidoamine-RGD peptide nanoparticles shows potential for treating metastasis in PTC. Conclusion: This study identifies key molecular mechanisms underlying PTC invasiveness and suggests a promising therapeutic strategy for PTC using targeted nanoscale drugs.


Subject(s)
MicroRNAs , Oligopeptides , Polyamines , Thyroid Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Neoplasm Invasiveness/genetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Cell Proliferation , Cell Movement , Gene Expression Regulation, Neoplastic , alpha 1-Antitrypsin/metabolism
19.
Int J Mol Sci ; 24(22)2023 Nov 16.
Article in English | MEDLINE | ID: mdl-38003602

ABSTRACT

Thyroid cancer is the most well-known type of endocrine cancer that is easily treatable and can be completely cured in most cases. Nonetheless, anti-cancer drug-resistant metastasis or recurrence may occur and lead to the failure of cancer therapy, which eventually leads to the death of a patient with cancer. This study aimed to detect novel thyroid cancer target candidates based on validating and identifying one of many anti-cancer drug-resistant targets in patient-derived sorafenib-resistant papillary thyroid cancer (PTC). We focused on targeting the sarco/endoplasmic reticulum calcium ATPase (SERCA) in patient-derived sorafenib-resistant PTC cells compared with patient-derived sorafenib-sensitive PTC cells. We discovered novel SERCA inhibitors (candidates 33 and 36) by virtual screening. These candidates are novel SERCA inhibitors that lead to remarkable tumor shrinkage in a xenograft tumor model of sorafenib-resistant patient-derived PTC cells. These results are clinically valuable for the progression of novel combinatorial strategies that facultatively and efficiently target extremely malignant cancer cells, such as anti-cancer drug-resistant PTC cells.


Subject(s)
Antineoplastic Agents , Thyroid Neoplasms , Animals , Humans , Sorafenib/pharmacology , Sorafenib/therapeutic use , Thyroid Cancer, Papillary/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal
20.
Oral Oncol ; 147: 106625, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37948895

ABSTRACT

OBJECTIVES: To describe the first reported use of neoadjuvant dabrafenib and trametinib specifically to permit organ conservation surgery in locally advanced recurrent differentiated thyroid carcinoma. PATIENTS AND METHODS: A patient presented with locally recurrent, radioiodide-resistant DTC with a BRAF V600E mutation invading the laryngotrachea. Definitive treatment would require a total laryngectomy. She was offered neoadjuvant dabrafenib and trametinib prior to surgery. RESULTS: A significant radiographic response permitted partial laryngectomy, enabling preservation of voice, early resumption of oral feeding, and avoidance of permanent tracheostomy. At 9 months, she remained free of disease. CONCLUSION: Neoadjuvant tyrosine kinase inhibitor treatment prior to definitive surgery for locally-invasive recurrent DTC is a potential approach that may limit the degree of surgery and associated morbidity.


Subject(s)
Proto-Oncogene Proteins B-raf , Thyroid Neoplasms , Female , Humans , Thyroid Cancer, Papillary/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Neoadjuvant Therapy , Organ Preservation , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Pyridones/pharmacology , Pyridones/therapeutic use , Mutation , Antineoplastic Combined Chemotherapy Protocols
SELECTION OF CITATIONS
SEARCH DETAIL