Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
JCI Insight ; 9(8)2024 Mar 07.
Article En | MEDLINE | ID: mdl-38478516

Both anaplastic thyroid cancer (ATC) and papillary thyroid cancer (PTC) originate from thyroid follicular epithelial cells, but ATC has a significantly worse prognosis and shows resistance to conventional therapies. However, clinical trials found that immunotherapy works better in ATC than late-stage PTC. Here, we used single-cell RNA sequencing (scRNA-Seq) to generate a single-cell atlas of thyroid cancer. Differences in ATC and PTC tumor microenvironment components (including malignant cells, stromal cells, and immune cells) leading to the polarized prognoses were identified. Intriguingly, we found that CXCL13+ T lymphocytes were enriched in ATC samples and might promote the development of early tertiary lymphoid structure (TLS). Last, murine experiments and scRNA-Seq analysis of a treated patient's tumor demonstrated that famitinib plus anti-PD-1 antibody could advance TLS in thyroid cancer. We displayed the cellular landscape of ATC and PTC, finding that CXCL13+ T cells and early TLS might make ATC more sensitive to immunotherapy.


Chemokine CXCL13 , Immunotherapy , Thyroid Cancer, Papillary , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Tumor Microenvironment , Tumor Microenvironment/immunology , Humans , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Carcinoma, Anaplastic/therapy , Thyroid Carcinoma, Anaplastic/immunology , Animals , Mice , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/immunology , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/therapy , Thyroid Neoplasms/pathology , Thyroid Neoplasms/immunology , Thyroid Neoplasms/therapy , Thyroid Neoplasms/genetics , Immunotherapy/methods , Chemokine CXCL13/metabolism , Chemokine CXCL13/genetics , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/pathology , Single-Cell Analysis , Prognosis , T-Lymphocytes/immunology , Female , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Male
2.
Gland Surg ; 12(11): 1500-1507, 2023 Nov 24.
Article En | MEDLINE | ID: mdl-38107486

Background: Despite the high incidence of lateral neck lymph node (LN) metastasis in papillary thyroid cancer (PTC), the management of the lateral neck remains controversial. We aimed to map the draining LNs in the lateral neck using carbon nanoparticles and explore its potential in neck evaluation. Methods: We conducted a multicenter, prospective study in PTC patients who had non-palpable yet suspicious metastatic lateral LNs on ultrasound and/or computed tomography (CT) but could not be confirmed by fine needle aspiration. Carbon nanoparticle suspension was injected peritumorally into the thyroid and modified lateral neck dissection was subsequently performed. Results: A total of 154 patients were enrolled for analysis. And 5,070 lateral LNs were removed, of which 1,079 (21.3%) were dyed. The median of dyed LNs was 6 per case (range, 1-33). The distribution of dyed LNs in neck compartments was IV > III > IIA > IIB/V, independent of tumor size, location, multifocality or microscopic extra-thyroidal extension (ETE). Compared with undyed LNs, the probabilities of metastasis in dyed LNs were significantly increased in compartment III, IV, V, and II-V (III: 29.3% vs. 15.4%, P<0.001; IV: 26.3% vs. 14.5%, P<0.001; V: 16.7% vs. 3.3%, P=0.005; II-V: 26.3% vs. 10.0%, P<0.001). The relative risks of metastasis in dyed LNs compared with undyed LNs were 1.90, 1.82, 5.04 and 2.62 in compartment III, IV, V, and II-V, respectively. Conclusions: It was the first prospective multicenter study to map the lateral neck LNs with carbon nanoparticles, which could help surgeons visualize the suspicious LNs during surgery. Instead of unguided LN biopsy, this method has a potential role in lateral neck assessment for indeterminate lateral LNs in PTC.

3.
Ann Surg Oncol ; 30(12): 7172-7180, 2023 Nov.
Article En | MEDLINE | ID: mdl-37543550

BACKGROUND: Surgery is the primary treatment for locally advanced differentiated thyroid cancer (DTC). However, some locally advanced patients are not candidates for R0/1 resection. There is limited evidence of neoadjuvant treatment in locally advanced DTC. Surufatinib targets multiple kinases, which is efficient, tolerable, and safe in patients with radioiodine-refractory DTC. In addition, surufatinib plus toripalimab (an anti-PD-1 antibody) showed encouraging antitumor activity in advanced solid tumors. This study was designed to evaluate the efficacy and safety of surufatinib plus toripalimab in locally advanced DTC in the neoadjuvant setting. METHODS: In this single-arm, phase II study, patients with pathologically confirmed unresectable or borderline resectable DTC were eligible and received a combination of 250 mg of surufatinib (orally daily) with 240 mg of toripalimab (intravenous, every 3 weeks). Treatment continued until satisfied for curative surgery, disease progression, withdrawal of consent, unacceptable toxicity, or investigator decision. Primary endpoint was objective response rate (ORR). Secondary endpoints included R0/1 resection rate, adverse events (AEs), etc. RESULTS: Ten patients were enrolled and received at least 4 cycles of treatment. The ORR was 60%. Nine patients received R0/1 resections after neoadjuvant treatment. The median best percentage change in the sum of the target lesion diameter was 32%. Most adverse events (AEs) were grade 1 or 2. CONCLUSIONS: Surufatinib in combination with toripalimab as neoadjuvant therapy for locally advanced DTC was feasible, and the majority of patients achieved R0/1 resection. It represents a new option for locally advanced DTC and needs further investigation.

4.
J Oral Pathol Med ; 52(5): 389-401, 2023 May.
Article En | MEDLINE | ID: mdl-36153671

BACKGROUND: Lymph node metastasis can independently predict oral squamous cell carcinoma patients' survival. This study would investigate the genetic and cellular differences between oral squamous cell carcinoma with positive and negative lymph node metastases. METHODS: We gathered single-cell RNA sequencing and bulk gene expression data from the Cancer Genome Atlas and Gene Expression Omnibus databases. Sixty lymph node-metastasis-related genes were discovered with refined single-cell RNA sequencing data analysis, and consensus clustering provided three molecular subtypes of oral squamous cell carcinoma. Least absolute shrinkage and selection operator analyses were then utilized to establish a five-gene risk model. CIBERSORT analysis revealed the immune infiltration profile of different risk subgroups. RESULTS: Oral squamous cell carcinoma patients were classified into three subtypes based on the 60 lymph node-metastasis-related key genes identified by single-cell RNA sequencing data. Patients in Subtype 3 showed a tendency for lymph node metastasis and poorer prognosis. Moreover, five biomarkers were selected from the 60 genes to construct a five-gene risk model evaluating the risk of lymph node metastasis. A lower probability of lymph node metastasis and a better prognosis was observed in the low-risk group. The immune infiltration of three different risk groups was explored with CIBERSORT. Besides, further analysis implied different sensitivities of anticancer drugs, including immunotherapy drugs and targeted compounds, in the three risk groups. CONCLUSION: In view of intratumoral heterogeneity, we found 60 genes associated with lymph node metastasis of oral squamous cell carcinoma. Subsequently, we constructed a five-gene signature that could improve the prediction of lymph node metastasis, clinical outcome, and promote individualized treatment strategies for oral squamous cell carcinoma.


Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Lymphatic Metastasis/genetics , Prognosis , RNA-Seq
5.
Head Neck ; 44(3): 805-809, 2022 03.
Article En | MEDLINE | ID: mdl-34862825

The objective of this study is to demonstrate a novel method for the reconstruction of right recurrent laryngeal nerve (RLN) by transforming into nonrecurrent RLN: the end-to-free vagal laryngeal branch end anastomosis. Here we report a case of locally advanced thyroid carcinoma. The patient underwent radical thyroid surgery with inevitably partial RLN resection and immediate right RLN reconstruction at our institution. With the guidance of intraoperative neuromonitoring (IOMN), we completed a novel end-to-free vagal laryngeal branch end anastomosis. The whole procedure was deliberately monitored by IOMN. Surgeons can procure adequate free nerve for tension-free anastomosis by transforming the right RLN into nonrecurrent nerve. Follow-up laryngoscope showed improved adductory movement of the right arytenoid. The end-to-free vagal end anastomosis is an effective way to reconstruct segmental nerve resection of right RLN. Its long-term postoperative result needs to be further warranted.


Recurrent Laryngeal Nerve Injuries , Recurrent Laryngeal Nerve , Anastomosis, Surgical , Humans , Laryngeal Nerves/surgery , Recurrent Laryngeal Nerve/physiology , Recurrent Laryngeal Nerve/surgery , Recurrent Laryngeal Nerve Injuries/prevention & control , Recurrent Laryngeal Nerve Injuries/surgery , Thyroidectomy/methods , Vagus Nerve/physiology , Vagus Nerve/surgery
6.
Clin Endocrinol (Oxf) ; 96(3): 402-412, 2022 03.
Article En | MEDLINE | ID: mdl-34592016

BACKGROUND: Limited studies have focused on the associated clinicopathologic features and short-term prognostic impacts of metastatic patterns at initial diagnosis in differentiated thyroid cancer (DTC). METHODS: Overall, 530 individuals with distant DTC diagnosed between 2010 and 2014 were identified from Surveillance, Epidemiology, and End Results (SEER) database. Multinomial logistic regression model was used to assess the clinicopathologic factors influencing the pattern of distant metastasis. Kaplan-Meier method and multivariable Cox regression were used to estimate the short-term effects of metastatic patterns on overall (OS) and thyroid cancer-specific survival (TCSS). RESULTS: Fifty, 111, 263, 59 and 47 patients presented with distant lymph node (LN)-only, bone-only, lung-only, bone plus lung, and liver and/or brain metastases (Mets), respectively. Regional lymph node metastasis (LNM) and follicular histotype were the only confirmed risk factors for distant LN-only Mets and bone-only Mets, respectively. Larger tumour size, extrathyroidal extension (ETE) and papillary histotype were associated with lung-only Mets. Synchronous bone and lung Mets were more likely to occur in older patients. In addition, patients with distant LN-only Mets had hardly any negative effect on OS and TCSS, whereas those with synchronous bone and lung or liver/brain Mets predicted unfavourable short-term outcomes, regardless of whether they received total thyroidectomy and radioisotopes. CONCLUSIONS: Different clinicopathologic factors predispose to different patterns of metastases with profound short-term survival differences among DTC patients. Our findings may help to determine effective pretreatment screening for aggressive metastatic patterns at initial diagnosis, and thus to provide additional treatment or access of clinical trials for these patients.


Lung Neoplasms , Thyroid Neoplasms , Aged , Humans , Lymphatic Metastasis , Prognosis , Retrospective Studies , Thyroid Neoplasms/pathology , Thyroidectomy/methods
7.
Thyroid ; 31(12): 1808-1813, 2021 12.
Article En | MEDLINE | ID: mdl-34610756

Background: Surgery is the primary treatment for locally advanced thyroid cancer. For some cases, R0/R1 resection could not be achieved at initial diagnosis and neoadjuvant treatment would be an option. Anlotinib is a multitarget tyrosine kinase inhibitor, which demonstrated antitumor activity in radioiodine-refractory differentiated thyroid cancer and medullary thyroid cancer. We aimed to evaluate the efficacy and safety of anlotinib in locally advanced thyroid cancer in the neoadjuvant setting. Methods: This single-arm phase II study investigated the efficacy and safety of anlotinib (12 mg orally daily, 2 weeks on/1 week off) for 2-6 cycles in patients with locally advanced thyroid cancer in the neoadjuvant setting. The key eligibility criteria included age 14-80 years old; locally advanced thyroid cancer that would benefit from surgery, and at least one measurable lesion. Operable patients received surgery after neoadjuvant treatment. The primary endpoint was objective response rate (ORR). Results: A total of 13 patients were enrolled and received an average of 3.5 cycles of anlotinib treatment. The ORR of anlotinib was 76.9% (95% confidence interval: 46.2-95.0%). The R0/R1 resection rate in the intent-to-treat population was 61.5% and in the per-protocol population was 72.7%. The median time to response was 61.5 days, and the disease control rate at 18 weeks was 92.3%. No patients had blood transfusion or tracheotomy. Most adverse events (AEs) were grade 1 or 2 and tended to discontinue when neoadjuvant treatment ceased. Common AEs of all grades were hypertension (76.9%), hypertriglyceridemia (69.2%), proteinuria (53.8%), thyrotropin increase (53.8%), cholesterol elevation (53.8%), and hand-foot syndrome (38.5%). Conclusions: Anlotinib demonstrated antitumor activity in the neoadjuvant treatment and the majority of patients achieved R0/R1 resection. AEs were consistent with the known anlotinib AE profile. These results suggest that anlotinib neoadjuvant treatment represents a new option for locally advanced thyroid cancer. Clinical Trial Registration Number: NCT04309136.


Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Quinolines/therapeutic use , Thyroid Neoplasms/therapy , Aged , Female , Humans , Male , Middle Aged , Neck Dissection , Neoadjuvant Therapy , Thyroidectomy
8.
Front Oncol ; 11: 647034, 2021.
Article En | MEDLINE | ID: mdl-34017682

Thyroid cancer is the most common type of endocrine malignancy. Although the general prognosis is good, the treatment of advanced disease is still challenging. Exosomes are vesicle units containing specific components that transmit information between cells. In order to explore its role in papillary thyroid cancer (PTC), our study screened exosome enriched lncRNA SNHG9 by lncRNA chip and explored its biological function. We used lncRNA chips combined with bioinformatics analysis to screen lncRNA SNHG9 enriched in exosomes. GO analysis suggested its relationship with autophagy and apoptosis. Quantitative PCR showed SNHG9 was highly expressed in PTC cells and exosomes and its correlation with PTC tumor size was analyzed by clinical characteristics. SNHG9 could inhibit the protective cell autophagy induced by starvation of human normal thyroid epithelial cell line Nthy-ori-3 and promote its apoptosis through PTC cell exosomes. RNA-pull down combined with protein spectrum showed that SNHG9 could interact with YBOX3. Western blot and RNA immunoprecipitation further confirmed their interaction. Western blot showed that SNHG9 could induce degradation of YBOX3, thus interfering with the stability of P21 mRNA and inducing cell apoptosis. In conclusion, our study identified SNHG9 as a PTC cell exosome-enriched lncRNA. SNHG9 could inhibit cell autophagy and promote apoptosis of Nthy-ori-3 cell through YBOX3/P21 pathway.

9.
J Clin Endocrinol Metab ; 106(8): e3208-e3220, 2021 07 13.
Article En | MEDLINE | ID: mdl-33656532

CONTEXT: The role of immune-related genes (IRGs) in thyroid cancer dedifferentiation and accompanying immune exhaustion remains largely unexplored. OBJECTIVE: To construct a significant IRG-based signature indicative of dedifferentiation and immune exhaustion in thyroid cancer. DESIGN AND SETTINGS: One exploratory cohort and 2 validation cohorts were used to identify stably dysregulated IRGs in dedifferentiated thyroid cancer (DDTC) and to obtain independent risk factors for dedifferentiation. The IRGs formed a gene signature, whose predictive value was tested by the receiver operating characteristic curve. Correlations between the signature and differentiation-related genes, immune checkpoints, and prognosis were analyzed. Gene set enrichment analyses were performed to identify related signaling pathways. RESULTS: Four IRGs (PRKCQ, PLAUR, PSMD2, and BMP7) were found to be repeatedly dysregulated in DDTC, and they formed an IRG-based signature with a satisfactory predictive value for thyroid cancer dedifferentiation. Correlation analyses revealed that immune checkpoints were closely related to the 4 IRGs and the IRG-based signature, which was significantly associated with the histological subtype (P = 0.026), lymph node metastasis (P = 0.001), and BRAFV600E mutation (P < 0.001). The downregulated expression of PRKCQ shortened the disease-free survival for patients with thyroid cancer. Furthermore, we identified several signaling pathways inherently associated with the IRG-based signature. CONCLUSIONS: This study suggests that IRGs participate in the dedifferentiation and immune exhaustion process of thyroid cancer and are potential biomarkers for DDTC.


Cell Dedifferentiation/genetics , Gene Expression Regulation, Neoplastic , Thyroid Neoplasms/genetics , Biomarkers, Tumor/genetics , Bone Morphogenetic Protein 7/genetics , Cell Line, Tumor , Gene Expression Profiling , Humans , Mutation , Prognosis , Protein Kinase C-theta/genetics , Receptors, Urokinase Plasminogen Activator/genetics , TNF Receptor-Associated Factor 2/genetics , Thyroid Neoplasms/immunology , Thyroid Neoplasms/pathology , Transcriptome
10.
J Clin Endocrinol Metab ; 106(1): 120-132, 2021 01 01.
Article En | MEDLINE | ID: mdl-33000173

CONTEXT: Programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte antigen 4 (CTLA-4), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), lymphocyte activation gene-3 (LAG-3), and T-cell immunoglobulin and ITIM domain (TIGIT) are considered major immune co-inhibitory receptors (CIRs) and the most promising immunotherapeutic targets in cancer treatment, but they are largely unexplored in medullary thyroid carcinoma (MTC). OBJECTIVE: We aimed to provide the first evidence regarding the expression profiles and clinical significance of CIRs in a large cohort of MTC patients. DESIGN AND PATIENTS: In total, 200 MTC patients who received initial surgery in our hospital were included. Immunohistochemistry was performed to evaluate CIR expressions in tissue microarrays (TMAs). Combined with the results of our previous programmed cell death ligand-1 (PD-L1) study, clinicopathologic and prognostic correlations of these proteins were retrospectively analyzed. RESULTS: TIM-3, PD-1, CTLA-4, LAG-3, and TIGIT positivity was detected in 96 (48.0%), 27 (13.5%), 25 (12.5%), 6 (3.0%), and 6 (3.0%) patients, respectively, in whom TIM-3, PD-1, and CTLA-4 expressions were positively correlated. Log-rank tests and multivariate Cox analyses both indicated that TIM-3, CTLA-4 expression, and PD-1/PD-L1 coexpression were associated with worse structural recurrence-free survival. In addition, among 20 patients who developed advanced disease during follow-up, 12 (60%) showed TIM-3 positivity, among whom 6 cases also had concurrent moderate to strong PD-1, PD-L1, or CTLA-4 expression. CONCLUSIONS: Using the currently largest TMA cohort of this rare cancer, we delineated the CIR expression profiles in MTC, and identified TIM-3, CTLA-4 expression, and PD-1/PD-L1 coexpression as promising biomarkers for tumor recurrence. Furthermore, a subset of advanced MTCs are probably immunogenic, for which single or combined immunotherapy including TIM-3, PD-1, PD-L1, or CTLA-4 blockade may be potential therapeutic approaches in the future.


Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/metabolism , Immune Checkpoint Proteins/metabolism , Thyroid Neoplasms/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD/analysis , Antigens, CD/metabolism , Biomarkers, Tumor/analysis , CTLA-4 Antigen/analysis , CTLA-4 Antigen/metabolism , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/pathology , Child , China/epidemiology , Cohort Studies , Disease Progression , Female , Hepatitis A Virus Cellular Receptor 2/analysis , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immune Checkpoint Proteins/analysis , Immunohistochemistry , Male , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/analysis , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/analysis , Receptors, Immunologic/metabolism , Retrospective Studies , Survival Analysis , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Tissue Array Analysis , Young Adult , Lymphocyte Activation Gene 3 Protein
11.
Oral Oncol ; 115: 105092, 2021 04.
Article En | MEDLINE | ID: mdl-33189578

Mammary analog secretory carcinoma (MASC), or secretory carcinoma of the thyroid is an extremely rare disease harboring ETV6-NTRK3 gene fusion with TRK activation. Here we report the twelfth case of MASC of the thyroid worldwide. A 36-year-old female was diagnosed with poor-differentiated thyroid carcinoma (PDTC). Pathology consultant and immunochemical workups showed the tumor cells were negative for TTF1, TG, PAX8, positive for S100, Vimentin, GATA-3, and focally positive for mammaglobin. Fluorescence in situ hybridization (FISH) assay using a dual-color break-apart probe showed ETV6 translocation t(12p13) (ETV6) was present and established the diagnosis of MASC. Next-generation sequencing (NGS) of a 47-gene panel identified exon 1-5 of ETV6 gene were fused with exons 15-19 of NTRK3 gene. The patient experienced three loco-regional recurrences within 12 months and eventually developed inoperable local disease as well as bilateral lung metastasis. She is currently receiving anti-TRK treatment with a follow-up time of 33 months. A literature review of MASC in the thyroid was also conducted.


Biomarkers, Tumor/metabolism , Oncogene Proteins/metabolism , Thyroid Neoplasms/diagnosis , Adult , Female , Humans , Mammary Analogue Secretory Carcinoma/diagnosis , Mammary Analogue Secretory Carcinoma/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/pathology
12.
Ann Transl Med ; 8(19): 1226, 2020 Oct.
Article En | MEDLINE | ID: mdl-33178758

BACKGROUND: Patients with anaplastic thyroid cancer (ATC), which is among the deadliest of all cancers, often have a poor response to traditional therapies. Currently, the role of long non-coding RNAs (lncRNAs) in ATC carcinogenesis is unclear. In this study, we analyzed the lncRNA expression profile of ATC with the aim of identifying potential molecular targets for treatment of the disease. METHODS: Whole transcriptome sequencing of three ATC and two normal thyroid (NT) samples was performed, and the lncRNA expression profile of ATC was analyzed. Original data as well as datasets deposited in the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) were used for clinical validation. Cell proliferation, Transwell, and apoptosis assays were performed using ATC cell lines. Gene Ontology (GO) and gene set enrichment analyses (GSEA) were performed to determine the dysregulated pathways. RESULTS: Whole transcriptome sequencing revealed 182 lncRNAs to be differentially expressed in ATC. One of the lncRNAs, mitotically associated long non-coding RNA (MANCR; LINC00704), was significantly overexpressed in ATC cell lines and patient samples compared with NT and papillary thyroid cancer (PTC). MANCR depletion in ATC cells significantly inhibited cancer cell proliferation and invasion, and induced apoptosis. By further analyzing the transcriptome data, we identified 451 genes co-expressed with MANCR. GO and GSEA showed that the top dysregulated pathways were related to mitosis and cell cycle. CONCLUSIONS: MANCR is a tumor promoter in ATC, and its role in carcinogenesis is possibly associated with cell cycle regulation. Because MANCR expression is minimal in most normal tissues, it may serve as a potential target in the future treatment of ATC.

13.
J Cancer ; 11(14): 4250-4260, 2020.
Article En | MEDLINE | ID: mdl-32368308

The impact of Hashimoto's thyroiditis (HT) on the progression of papillary thyroid cancer (PTC) is still unclear. Interleukin-2 (IL-2) is a growth factor and crucial for HT development. This study aimed at investigating the effect of IL-2 on MHC class I expression in PTC cells and immune activation with experimental treatment for PTC using PTC cell lines. We assessed the expression of IL-2, HLA class I, PD-L1, CD3, CD8 and CD16 molecules in paired PTC tissues and HLA-ABC and PD-L1 expression in IL-2 pre-treated K1, TPC-1 and BCPAP cells by immunohistochemistry, qPCR, flow cytometry and Western blotting. The effect of IL-2 on immunogenicity of PTC cells to stimulate activated human T cells was determined for the percentages of activated CD8+ T cells and their cytokine production as well as PD-1 and PD-L1 expression. Compared with non-tumor tissues, we found that IL-2 expression was up-regulated in PTC tissues, particularly in PTC+HT tissues and correlated positively with HLA-class I, CD3 and CD8 expression in PTC+HT tissues. Conversely, PD-L1 expression decreased in PTC+HT tissues. Treatment with IL-2 significantly up-regulated HLA-class I expression, but down-regulated PD-L1 expression in PTC cells. Co-culture with IL-2-pre-treated PTC cells significantly promoted the proliferation of activated CD8+ T cells and their IL-2 secretion, but decreased their PD-1 expression, accompanied by decreased PD-L1 expression in IL-2-treated PTC cells in vitro. In conclusion, IL-2 up-regulated HLA-class I expression and enhanced anti-tumor T cell immunity during the development of PTC and HT. IL-2 may be a promising immunotherapy for PTC.

14.
Endocrinology ; 161(10)2020 10 01.
Article En | MEDLINE | ID: mdl-32348468

The incidence of papillary thyroid cancer (PTC) with concomitant Hashimoto thyroiditis (HT) is increasing. Interleukin (IL)-10 is a cytokine previously reported to be elevated in this condition. Evidence from multiple human malignancies showed IL-10 participated in tumor immunity and exhibited therapeutic potential. The aim of this study is to investigate whether IL-10 interferes with tumor immunity in PTC with concomitant HT. Expression of IL-10 and major histocompatibility complex (MHC) class Ⅰ were compared with PTC tissues with or without concomitant HT. PTC cell lines K1 and TPC-1 were stimulated with IL-10 and analyzed for MHC class Ⅰ expression afterward. T-cell activation, production of IL-2 and interferon (IFN)-γ and programmed death-1 (PD-1) expression were assessed by coculture of donor peripheral blood lymphocytes (PBLs) with IL-10-pretreated PTC cells. Programmed death-ligand 1 (PD-L1) expression was measured in PTC tissues and IL-10-pretreated cells of K1 and TPC-1. Increased levels of IL-10 and MHC class Ⅰ were observed in PTC with concomitant HT. IL-10 stimulation increased MHC class Ⅰ expression of PTC cells in vitro. Coculture of PBLs with IL-10-pretreated PTC cells enhanced T-cell activation (% cluster of differentiation [CD]25+ of CD3+T cells) and increased IL-2 production along with decreased IFN-γ secretion and PD-1 expression. Reduced PD-L1 expression was seen in PTC + HT tissue samples and IL-10-stimulated PTC cell lines. Elevated IL-10 expression in PTC with concomitant HT restores MHC class Ⅰ expression and interferes with tumor immunity. The potential mechanism of IL-10 in tumor immunity needs further investigation.


Genes, MHC Class I/drug effects , Hashimoto Disease/genetics , Interleukin-10/pharmacology , Lymphocyte Activation/drug effects , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Adult , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cells, Cultured , Cohort Studies , Female , Gene Expression Regulation, Neoplastic/drug effects , Hashimoto Disease/complications , Hashimoto Disease/immunology , Hashimoto Disease/pathology , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocyte Activation/genetics , Male , Middle Aged , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Thyroid Cancer, Papillary/complications , Thyroid Cancer, Papillary/immunology , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/complications , Thyroid Neoplasms/immunology , Thyroid Neoplasms/pathology , Tumor Escape/drug effects , Tumor Escape/genetics , Young Adult
15.
Oncol Rep ; 43(3): 783-794, 2020 03.
Article En | MEDLINE | ID: mdl-31922231

Endeavors towards identifying key molecular markers for early diagnosis and treatment are driving the clinical study of papillary thyroid carcinoma (PTC). Recent studies have indicated that protein phosphatase, Mg2+/Mn2+ dependent, 1D (PPM1D) exerts an oncogenic function by increasing cell proliferation, migration and invasion in various cancer types. In addition, PPM1D has a high frequency of genetic alterations and has been proposed as a tumor driver in thyroid cancer, making PPM1D an attractive potential oncotarget for cancer treatment. The aims of the present study were to investigate the downstream targets of PPM1D and the potential molecular mechanisms of its oncogenic activities, as well as its clinical significance in PTC. As anticipated, PPM1D overexpression was confirmed in PTC clinical specimens. Furthermore, knockdown of PPM1D in thyroid cancer cell lines significantly suppressed the proliferation, migration and invasion but facilitated cell apoptosis. The protein levels of phosphorylated p38 mitogen­activated protein kinase (MAPK), p53 and Bax were increased in PPM1D­knockdown cells, while inhibition of p38 phosphorylation restored cell migration, proliferation and cell apoptosis. In addition, silencing of PPM1D expression induced nuclear translocation of p53 in K­1 and TPC­1 cells. The present results demonstrated that PPM1D regulated p38 MAPK and p53 signaling pathways to promote thyroid cancer progression. Collectively with the clinical results, these data qualified PPM1D as a potential diagnostic biomarker and therapeutic target in human thyroid cancer.


Biomarkers, Tumor/genetics , Protein Phosphatase 2C/genetics , Thyroid Cancer, Papillary/genetics , Tumor Suppressor Protein p53/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Adult , Aged , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Thyroid Cancer, Papillary/pathology , bcl-2-Associated X Protein/genetics
16.
Eur Arch Otorhinolaryngol ; 276(12): 3425-3434, 2019 Dec.
Article En | MEDLINE | ID: mdl-31511971

PURPOSE: The role of lymph node ratio (LNR, ratio of metastatic to examined nodes) in the staging of multiple human malignancies has been reported. We aim to evaluate its value in salivary gland cancer (SGC). METHODS: Records of SGC patients from Surveillance, Epidemiology, and End Results database (SEER, training set, N = 4262) and Fudan University Shanghai Cancer Center (FUSCC, validating set, N = 154) were analyzed for the prognostic value of LNR. Kaplan-Meier survival estimates, the Log-rank χ2 test and Cox proportional hazards model were used for univariate and multivariate analysis. Optimal LNR cutoff points were identified by X-tile. RESULTS: Optimal LNR cutoff points classified patients into four risk groups, R0, R1 (≤ 0.17), R2 (0.17-0.56) and R3 (> 0.56), corresponding to 5-year cause-specific survival in SEER patients of 88.6%, 57.2%, 53.1% and 39.7%, disease-free survival in FUSCC patients of 69.2%, 63.3%, 34.6% and 0%, and disease-specific survival in FUSCC patients of 92.3%, 90.0%, 71.4% and 0%, respectively. Compared with TNM staging, TNM + R staging showed smaller AIC values and higher C-index values in the Cox regression model in both patient sets. CONCLUSIONS: LNR classification should be considered as a complementary system to TNM staging and LNR classification based clinical trials deserve further research.


Lymph Node Ratio , Neoplasm Staging/methods , Adult , Aged , China/epidemiology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging/standards , Prognosis , Proportional Hazards Models , Risk Factors , SEER Program , Salivary Gland Neoplasms/pathology
17.
J Cancer ; 10(18): 4380-4388, 2019.
Article En | MEDLINE | ID: mdl-31413758

Background: The aim of this study was to develop and validate prognostic nomograms predicting overall (OS) and cancer-specific survival (CSS) of patients with major salivary gland (MaSG) mucoepidermoid carcinoma (MEC). Methods: 1398 MaSG-MEC patients were identified from the Surveillance, Epidemiology and End Results (SEER) database. They were randomly and equally divided into a training cohort (n=699) and a validation cohort (n=699). The best subsets of covariates were identified to develop nomograms predicting OS and CSS based on the smallest Akaike Information Criterion (AIC) value in the multivariate Cox models. The nomograms were internally and externally validated by the bootstrap resampling method. The predictive ability was evaluated by Harrell's Concordance Index (C-index). Results: For the training cohort, eight (age at diagnosis, tumor grade, primary site, surgery, radiation, T, N and M classification) and seven predictors (all the above factors except primary site) were selected to create the nomograms estimating the 3- and 5- year OS and CSS, respectively. C-index indicated better predictive performance of the nomograms than the 7th AJCC staging system, which was confirmed by both internal (via the training cohort: OS: 0.888 vs 0.785, CSS: 0.938 vs 0.821) and external validation (via the validation cohort: OS: 0.844 vs 0.743, CSS: 0.882 vs 0.787). The calibration plots also revealed good agreements between the nomogram-based prediction and observed survival. Conclusions: We have proposed and validated the nomograms predicting OS and CSS of MaSG-MEC. They are proved to be of higher predictive value than the AJCC staging system and may be adopted in future clinical practice.

18.
Diagn Pathol ; 14(1): 52, 2019 Jun 04.
Article En | MEDLINE | ID: mdl-31159823

BACKGROUND: The incidence of coexisting papillary thyroid cancer (PTC) and Hashimoto's thyroiditis (HT) is increasing. The impact of HT on PTC prognosis and its possible mechanism remains controversial. Interleukin-17A (IL-17A) has been reported to participate in the pathogenesis of multiple autoimmune diseases and cancers. The aim of this study is to investigate the role of IL-17A in PTC with coexistent HT and evaluate the changes in tumor antigenicity. METHODS: Expression of IL-17A and major histocompatibility complex (MHC) class I molecules were compared on PTC tissue samples with or without HT. PTC cell lines K1 and TPC-1 were stimulated with IL-17A and analyzed for MHC class I expression afterwards. Cluster of differentiation (CD) 8+T cell activation, production of Interleukin-2 (IL-2) and Interferon-gamma (IFN-γ) as well as the programmed death-1 (PD-1) expression on lymphocytes were assessed by coculture of donor peripheral blood lymphocytes (PBLs) with IL-17A pretreated PTC cells. RESULTS: Elevated IL-17A and MHC class I expression were observed in PTC tissue samples with coexistent HT. Stimulation of PTC cells with IL-17A effectively increased MHC class I expression in vitro. Coculture of PBLs with IL-17A pretreated PTC cells resulted in enhanced T cell activation (%CD25+ of CD3+T cells) and increased IL-2 production along with decreased IFN-γ secretion and PD-1 expression of the lymphocytes. CONCLUSIONS: Papillary thyroid cancer with coexisting Hashimoto's thyroiditis presents elevated MHC class I expression, which may be the result of IL-17A secretion. T cell activation is enhanced in vitro by IL-17A and may provide future utility in PTC immunotherapy.


Hashimoto Disease/immunology , Histocompatibility Antigens Class I/metabolism , Interleukin-17/metabolism , Thyroid Cancer, Papillary/immunology , Thyroid Neoplasms/immunology , Aged , Antigenic Variation/immunology , Cell Line, Tumor , Cohort Studies , Female , Hashimoto Disease/pathology , Humans , Immunohistochemistry , Interferon-gamma/metabolism , Male , Middle Aged , Prognosis , Thyroid Cancer, Papillary/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Tumor Escape
19.
Eur J Surg Oncol ; 45(11): 2143-2150, 2019 Nov.
Article En | MEDLINE | ID: mdl-31253544

BACKGROUND: We aimed to explore whether the anatomic extent of lymph node metastases (AE-LNM) could independently predict prognosis of node-positive major salivary gland carcinoma (MaSGC). METHODS: A total of 376 pathologically node-positive MaSGC patients were identified from the Surveillance, Epidemiology and End Results database and constituted the training cohort. Using the X-Tile program, these patients were divided into three groups based on AE-LNM degrees. Discrimination of overall survival (OS) and disease-specific survival (DSS) was evaluated and compared with the 8th American Joint Committee on Cancer (AJCC) pN classification. The results were externally validated by 220 patients in a Chinese multicenter cohort (Validation cohort). RESULTS: Using the training cohort, AE-LNM was divided into Extent 1 (spread to parotid LNs or level I), Extent 2 (spread to level II-IV) and Extent 3 (spread to level V or bilateral LNs or rare LNs). Regarding both OS and DSS, the AE-LNM model revealed clear separation of survival curves, while the pN classification failed to discriminate the prognosis of pN1 and pN2 patients. When we incorporated both the AE-LNM model and AJCC pN classification into the same multivariate Cox analyses, AE-LNM was still an independent prognostic factor, while the AJCC pN classification lost its significance. These results were externally validated by the validation cohort. CONCLUSION: AE-LNM is an independent nodal prognosticator for node-positive MaSGC and may have improved discriminative ability over the current AJCC pN classification. Integration of anatomic extent of LNM into the current AJCC N classification could be considered.


Adenocarcinoma/pathology , Lymph Nodes/pathology , Salivary Gland Neoplasms/pathology , Adult , Aged , Carcinoma, Acinar Cell/pathology , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Ductal/pathology , Carcinoma, Mucoepidermoid/pathology , China , Cohort Studies , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Parotid Neoplasms/pathology , Prognosis , SEER Program , Survival Rate , United States
20.
Int J Endocrinol ; 2019: 8428547, 2019.
Article En | MEDLINE | ID: mdl-30915116

BACKGROUND: Anaplastic thyroid cancer (ATC) responds poorly to conventional therapies and requires a multidisciplinary approach to manage. The aim of the current study is to explore whether aggressive treatment is beneficial, especially the appropriate extent of surgery in ATC. METHODS: Patients diagnosed with ATC from 2004 to 2014 were identified from the Surveillance, Epidemiology, and End Results (SEER) database and included in our study. RESULTS: A total of 735 ATC patients were identified. The two-year overall survival (OS) rates for stage IVA, IVB, and IVC patients were 36.5%, 15.6%, and 1.4%, respectively. By directly comparing eight treatment modalities, we found that surgery + radiotherapy (RT) ± chemotherapy was the most effective treatment strategy. surgery + chemotherapy and RT + chemotherapy had comparable results (hazard ratio (HR) = 1.461, 95% confidential interval (CI): 0.843-2.531, P = 0.177). Multivariate Cox regression analysis also showed increased mortality risk in patients with increased age (HR = 1.022, P < 0.001), tumor extension to adjacent structures (HR = 1.649, P = 0.013), and distant metastasis (HR = 2.041, P < 0.001), while surgery + RT (HR = 0.600, P = 0.004) and chemotherapy (HR = 0.692, P = 0.010) were independently associated with improved OS. Further analysis revealed that patients undergoing total/near-total thyroidectomy (TT) had superior OS to those receiving less than TT (P < 0.001). In subgroup analysis, the benefit of TT remained significant in patients with tumors larger than 4.0 cm (HR = 0.776, 95% CI: 0.469-0.887, P = 0.007), with adjacent structure extension (HR = 0.642, 95% CI: 0.472-0.877, P = 0.005), including trachea and major vessels, but not in patients with early phase local disease such as tumor ≤ 4.0 cm or tumor within the thyroid or with minimal extrathyroidal extension. Patients with very locally advanced disease or distant metastasis could not benefit from TT as well. CONCLUSIONS: In operable cases, surgery + RT ± chemotherapy was the optimal treatment modality. Otherwise, RT + chemotherapy was the appropriate strategy. However, TT was not beneficial for very early stage or metastatic ATC.

...