1
|
Zou Z, Zhong L. Anaplastic thyroid cancer: Genetic roles, targeted therapy, and immunotherapy. Genes Dis 2025; 12:101403. [PMID: 40271195 PMCID: PMC12018003 DOI: 10.1016/j.gendis.2024.101403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/02/2024] [Accepted: 08/02/2024] [Indexed: 04/25/2025] Open
Abstract
Anaplastic thyroid cancer (ATC) stands as the most formidable form of thyroid malignancy, presenting a persistent challenge in clinical management. Recent years have witnessed a gradual unveiling of the intricate genetic underpinnings governing ATC through next-generation sequencing. The emergence of this genetic landscape has paved the way for the exploration of targeted therapies and immunotherapies in clinical trials. Despite these strides, the precise mechanisms governing ATC pathogenesis and the identification of efficacious treatments demand further investigation. Our comprehensive review stems from an extensive literature search focusing on the genetic implications, notably the pivotal MAPK and PI3K-AKT-mTOR signaling pathways, along with targeted therapies and immunotherapies in ATC. Moreover, we screen and summarize the advances and challenges in the current diagnostic approaches for ATC, including the invasive tissue sampling represented by fine needle aspiration and core needle biopsy, immunohistochemistry, and 18F-fluorodeoxyglucose positron emission tomography/computed tomography. We also investigate enormous studies on the prognosis of ATC and outline independent prognostic factors for future clinical assessment and therapy for ATC. By synthesizing this literature, we aim to encapsulate the evolving landscape of ATC oncology, potentially shedding light on novel pathogenic mechanisms and avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zhao Zou
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linhong Zhong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
2
|
Pan Z, Tan Z, Xu N, Yao Z, Zheng C, Shang J, Xie L, Xu J, Wang J, Jiang L, Zhu X, Yu D, Li Y, Che Y, Gong Y, Qin Z, Zhang Y, Zou X, Xu T, Guo Z, Jin T, Guo T, Wang W, Chen W, Sun Y, Wang W, Peng X, Yin C, Ding C, Huang P, Ge M. Integrative proteogenomic characterization reveals therapeutic targets in poorly differentiated and anaplastic thyroid cancers. Nat Commun 2025; 16:3601. [PMID: 40234451 PMCID: PMC12000556 DOI: 10.1038/s41467-025-58910-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC) present major challenges in treatment owing to extreme aggressiveness and high heterogeneity. In this study, deep-scale analyses spanning genomic, proteomic, and phosphoproteomic data are performed on 348 thyroid-cancer and 119 tumor-adjacent samples. TP53 (48%), TERT promoter (36.5%), and BRAF (23%) are most frequently mutated in PDTC and ATC. Ribosome biogenesis is identified as a common hallmark of ATC, and RRP9 silencing dramatically inhibits tumor growth. Proteomic clustering identified three ATC/PDTC subtypes. Pro-I subtype is characterized with aberrant insulin signaling and low immune cell infiltration, and Pro-II is featured with DNA repair signaling, while Pro-III harbors high frequency of TP53 and BRAF mutation and intensive C5AR1+ myeloid infiltration. Targeting C5AR1 synergistically improves antitumor effect of PD-1 blockade against ATC cell-derived tumors. These findings provide systematic insights into tumor biology and opportunities for drug discovery, accelerating precision therapy for virulent thyroid cancers.
Collapse
Affiliation(s)
- Zongfu Pan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ning Xu
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zhenmei Yao
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Chuanming Zheng
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Jinbiao Shang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, China
| | - Lei Xie
- Department of Head and Neck Surgery, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiajie Xu
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Jiafeng Wang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Liehao Jiang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xuhang Zhu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, China
| | - Dingyi Yu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ying Li
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yulu Che
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yingying Gong
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Zhaoyu Qin
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xiaozhou Zou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Zhenying Guo
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Tiefeng Jin
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Tiannan Guo
- School of Medicine, Westlake University, Hangzhou, China
| | - Wei Wang
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Wanyuan Chen
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yaoting Sun
- School of Medicine, Westlake University, Hangzhou, China
| | - Weixin Wang
- Hangzhou Cosmos Wisdom Biotechnology Co. Ltd, Hangzhou, China
| | - Xiaojun Peng
- Hangzhou Cosmos Wisdom Biotechnology Co. Ltd, Hangzhou, China
| | - Changtian Yin
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Chen Ding
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, China.
| |
Collapse
|
3
|
Fang X, Luo P, Kai Z, Li P, Chen X, Huang J. Multigene Detection Analysis of Multifocal Papillary Thyroid Carcinoma. Clin Endocrinol (Oxf) 2025. [PMID: 40235071 DOI: 10.1111/cen.15251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/22/2025] [Accepted: 04/04/2025] [Indexed: 04/17/2025]
Abstract
OBJECTIVE To analyze the relationship between clinical characteristics and pathogenic gene mutations in multifocal papillary thyroid carcinoma (MPTC) and to investigate the proportion of independent primary tumors (IP) versus intrathyroidal metastases (ITM) in MPTC. Additionally, to explore the correlation between specific gene mutations and clinical features such as multifocality, tumor size, and lymph node metastasis. METHODS Patients with multifocal thyroid tumor meeting inclusion criteria were consecutively enrolled. Two lesions per case were selected for preoperative ultrasound-guided fine-needle aspiration biopsy (FNAB). All lesions were pathologically confirmed as papillary thyroid carcinoma postoperatively. FNAB samples were subjected to multi-gene panel testing and classified into three groups based on mutational profiles of 26 thyroid-related genes: (1) identical mutations in both lesions (intrathyroidal metastasis), (2) completely discordant mutations (independent primary tumors), and (3) shared mutations with an additional mutation in one lesion (uncertain origin). RESULTS Among 58 initially enrolled MPTC patients, 8 were excluded due to noncompliant specimens. The final cohort included 50 patients (37 females, 13 males) with a mean age of 42.64 ± 11.12 years. The median tumor diameter was 12.5 (IQR: 7.6, 20.0) mm, with 38.0% (19/50) classified as papillary thyroid microcarcinoma. A total of 128 mutations, 4 gene fusions, and 3 gene amplifications were detected across 100 qualified FNAB samples. BRAF V600E was the most prevalent mutation (84.0%, 84/100), followed by DICER1 (7.0%), PTEN (6.0%), and RET (4.0%). Identical mutational profiles (intrathyroidal metastasis) were observed in 64.0% (32/50) of cases, while 18.0% (9/50) exhibited completely discordant mutations (independent primary tumor). The remaining 18.0% (9/50) showed shared mutations with an additional mutation, predominantly in smaller lesions. The incidence of completely different mutations in ipsilateral lesions and bilateral lesions was different (p = 0.030). No significant correlation between BRAF V600E and clinical characteristics such as tumor size, multifocality, capsular invasion or lymph node metastasis (p > 0.05). CONCLUSION Multi-gene panel testing of preoperative FNAB samples effectively discriminates clonal relationships in MPTC, revealing distinct molecular profiles between ITM and IP. The high prevalence of BRAF V600E mutations and frequent clonal homogeneity underscore the necessity of comprehensive genetic profiling to guide personalized management. Routine multi-lesion sampling is advocated for optimizing risk stratification and surgical decision-making.
Collapse
Affiliation(s)
- Xiaolian Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Peng Luo
- Department of Research and Development, Zhejiang Shaoxing Topgen Biomedical Technology Co. Ltd., Shanghai, 201321, China
| | - Zhentian Kai
- Department of Research and Development, Zhejiang Shaoxing Topgen Biomedical Technology Co. Ltd., Shanghai, 201321, China
| | - Pingdong Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xiaohong Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Junwei Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| |
Collapse
|
4
|
Su Y, Mei L, Wu Y, Li C, Jiang T, Zhao Y, Feng X, Sun T, Li Y, Wang Z, Ji Y. Xenotropic and polytropic retrovirus receptor 1 (XPR1) promotes progression of papillary thyroid carcinoma via the BRAF-ERK1/2-P53 signaling pathway. J Endocrinol Invest 2025; 48:633-652. [PMID: 39487939 DOI: 10.1007/s40618-024-02481-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. Xenotropic and polytropic retrovirus receptor 1 (XPR1), identified as a cellular receptor, plays roles in many pathophysiological processes. However, the underlying function and molecular mechanisms of XPR1 in PTC remain unclear. Therefore, this study aimed to elucidate the role of XPR1 in the process of PTC and the potential mechanisms. METHODS RNA-sequencing was performed for gene differential expression analysis in PTC patients' tissues. Immunohistochemical assay, real-time PCR, and western blotting assay were used to determine the expression of XPR1, BRAF, and P53 in PTC tissues. The function of XPR1 on the progression of PTC was explored using in vitro and in vivo experiments. The molecular mechanism of XPR1 was investigated using gene silencing, ELISA, immunofluorescence, western blotting, and real-time PCR assays. RESULTS We found that XPR1 was markedly upregulated in PTC tissues compared to adjacent noncancerous tissues, suggesting that high expression of XPR1 could be correlated with poor patient disease-free survival in PTC. In addition, the expression of BRAF and P53 in PTC tissues was substantially higher than in adjacent noncancerous tissues. Silencing of XPR1 reduced the proliferation, migration, and invasion capacities of TPC-1 cells in vitro and effectively inhibited the tumorigenecity of PTC in vivo. More importantly, silencing of XPR1 in TPC-1 cells significantly decreased the expression of XPR1, BRAF, and P53 both in vitro and in vivo. Interestingly, we demonstrated that XPR1 may positively activate the BRAF-ERK-P53 signaling pathway, further promoting PTC progression. CONCLUSION The findings reveal a crucial role of XPR1 in PTC progression and prognosis via the BRAF-ERK1/2-P53 signaling pathway, providing potential therapeutic targets for treating PTC.
Collapse
Affiliation(s)
- Yuanhao Su
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Lin Mei
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Yongke Wu
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Cheng Li
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Tiantian Jiang
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Yiyuan Zhao
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Xin Feng
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Tingkai Sun
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Yunhao Li
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Zhidong Wang
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China.
| | - Yuanyuan Ji
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
5
|
Houl JH, Bagheri-Yarmand R, Kunnimalaiyaan M, Miranda Mendez P, Kidd JL, Dadbin A, Jurado Ruiz A, Parekh PA, Henderson YC, Chari NS, Thennavan AT, Powell RT, Stephan CC, Zhao X, Maniakas A, Nurieva R, Busaidy NL, Cabanillas ME, Dadu R, Zafereo M, Wang JR, Lai SY, Hofmann MC. Role of the ETV5/p38 signaling axis in aggressive thyroid cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.637322. [PMID: 40027641 PMCID: PMC11870521 DOI: 10.1101/2025.02.17.637322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Patients with poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC) face a much poorer prognosis than those with differentiated thyroid cancers. Around 25% of PDTCs and 35% of ATCs carry the BRAFV600E mutation, which constitutively activates the MAPK pathway, a key driver of cell growth. Although combining BRAF and MEK inhibitors can shrink tumors, resistance often develops. The exact cause of this resistance remains unclear. We previously found that in PDTC and ATC cells the BRAFV600E mutation is strongly linked to the expression of ETV5, a transcription factor downstream of the MAPK pathway. In the current study, we observed a significant association between ETV5 expression and the activation of p38, a central component of the MAPK14 pathway. Upon reduction of ETV5 levels, p38 expression and activation decreased, along with its upstream regulators MKK3/MKK6. This suggests that the MAPK and p38/MAPK14 pathways are interconnected and that p38 has oncogenic properties in these cancers. Using high-throughput screening, we established that combining p38 inhibitors with the BRAF inhibitor dabrafenib showed strong synergy in vitro, including in cells resistant to dabrafenib and trametinib that had acquired a secondary TP53 mutation. We then tested this combination in a genetically engineered mouse model (GEMM) of ATC. Overall, our findings suggest an oncogenic link between the MAPK and p38/MAPK14 pathways and that combining p38 pathway inhibitors with dabrafenib-targeted therapy could improve treatment outcomes for aggressive thyroid cancers. However, more specific and effective p38 inhibitors are required to fully harness this potential.
Collapse
|
6
|
Sun D, Zhang X, Jin X, Shi C, Sun Y, Zhang Y, Liang J, Lin Y. BRAF V600E mutation is associated with better prognoses in radioactive iodine refractory thyroid cancer patients treated with multi-kinase inhibitors: a retrospective analysis of registered clinical trials. Thyroid Res 2025; 18:5. [PMID: 39924483 PMCID: PMC11808998 DOI: 10.1186/s13044-025-00223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND The antiangiogenic multi-kinase inhibitors (MKIs) apatinib, donafenib, and anlotinib have demonstrated satisfactory efficacy in radioactive iodine refractory differentiated thyroid cancer (RAIR-DTC) in their phase II/III trials. However, the potential impact factors on the efficacy of these MKIs remain unclear. METHODS RAIR-DTC patients enrolled in clinical trials of apatinib, donafenib, and anlotinib in our center were retrospectively reviewed. The Kaplan-Meier method was used to examine the relationship between clinicopathological variables and progression-free survival (PFS) and overall survival (OS), followed by a multivariate Cox analysis on PFS. RESULTS A total of 71 progressive RAIR-DTC patients were reviewed, of which 26.7% were treated by anlotinib, 45.1% by apatinib, and 28.2% by donafenib. The median follow-up time was 44.1 months, the median PFS was 21.1 months, and the estimated median OS was 47.7 months. PFS and OS showed no significant differences in patients treated with apatinib, donafenib, or anlotinib. In the univariate analyses, patients with BRAFV600E mutation showed longer PFS (HR 0.345, 95% CI 0.187-0.636, p < 0.001) and OS (HR 0.382, 95% CI 0.166-0.878, p = 0.019) compared with patients with wild-type BRAF. Patients with follicular thyroid cancer and bone metastases had shorter PFS, and patients with worse Eastern Cooperative Oncology Group performance status, bone metastases, and a larger tumor burden had shorter OS. In the multivariate Cox analysis, BRAFV600E mutation was the only independent predictor of longer PFS (HR 0.296, 95% CI 0.138-0.638, p = 0.002). The overall response rate and disease control rate didn't differ between BRAFV600E mutation status. Subgroup analysis of PFS in papillary thyroid cancer patients stratified by BRAFV600E mutation status showed that BRAFV600E mutation was associated with longer PFS in all clinicopathological subgroups (hazard ratio < 1). CONCLUSION RAIR-DTC patients with BRAFV600E mutation treated with apatinib, donafenib, or anlotinib achieved better prognoses compared with patients with wild-type BRAF, indicating that the genetic background may play a role in predicting the efficacy of MKIs therapies. TRIAL REGISTRATION This retrospective cohort included patients in our center from clinical trials of apatinib (NCT02731352, NCT03048877), donafenib (NCT02870569, NCT03602495), and anlotinib (NCT05007093).
Collapse
Affiliation(s)
- Di Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Xin Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Xiaona Jin
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Cong Shi
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Yuqing Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Yingqiang Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Peking University, No. 1 Life Park Road, Zhongguancun Life Science Park, Changping District, Beijing, 102206, China.
- Department of Medical Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Yansong Lin
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China.
| |
Collapse
|
7
|
Pan Z, Chen J, Xu T, Cai A, Han B, Li Y, Fang Z, Yu D, Wang S, Zhou J, Gong Y, Che Y, Zou X, Cheng L, Tan Z, Ge M, Huang P. VSIG4 + tumor-associated macrophages mediate neutrophil infiltration and impair antigen-specific immunity in aggressive cancers through epigenetic regulation of SPP1. J Exp Clin Cancer Res 2025; 44:45. [PMID: 39920772 PMCID: PMC11803937 DOI: 10.1186/s13046-025-03303-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
V-set and immunoglobulin domain-containing 4 (VSIG4) positive tumor-associated macrophage (VSIG4+ TAM) is an immunosuppressive subpopulation newly identified in aggressive cancers. However, the mechanism how VSIG4+ TAMs mediate immune evasion in aggressive cancers have not been fully elucidated. In our study, we found targeting VSIG4+ TAMs by VSIG4 deficiency or blockade remarkably limited tumor growth and metastasis, especially those derived from anaplastic thyroid cancer (ATC) and pancreatic cancer, two extremely aggressive types. Moreover, the combination of VSIG4 blockade with a BRAF inhibitor synergistically enhanced anti-tumor activity in ATC-tumor bearing mice. VSIG4 deficiency recovered the antigen presentation (B2m, H2-k1, H2-d1) of TAMs and activated antigen-specific CD8+ T cells by promoting their in vivo proliferation and intratumoral infiltration. Notably, loss of VSIG4 in TAMs significantly reduced the production of lactate and histone H3 lysine 18 lactylation, resulting the decreased transcription of SPP1 mediated by STAT3, which collectively disrupted the cell-cell interactions between TAMs and neutrophils. Further combination of VSIG4 with SPP1 blockade synergistically boosted anti-tumor activity. Overall, our studies demonstrate the epigenetic regulation function of VSIG4 confers on TAMs an alternative pattern, beyond the checkpoint role of VSIG4, to shape the immunosuppressive tumor microenvironment and impair antigen-specific immunity against aggressive cancers.
Collapse
Affiliation(s)
- Zongfu Pan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Jinming Chen
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Anqi Cai
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Bing Han
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Ying Li
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Ziwen Fang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Dingyi Yu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Shanshan Wang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Junyu Zhou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Yingying Gong
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Yulu Che
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Xiaozhou Zou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Lei Cheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
| |
Collapse
|
8
|
Dutta S, Knauf JA. Development of animal models to study aggressive thyroid cancers. Eur Thyroid J 2025; 14:e240361. [PMID: 39874138 PMCID: PMC11825169 DOI: 10.1530/etj-24-0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 01/30/2025] Open
Abstract
The development of mouse models for thyroid cancer has significantly advanced over the years, enhancing our understanding of thyroid tumorigenesis, molecular pathways and treatment responses. The earliest mouse models of thyroid cancer relied on hormone, radiation or chemical carcinogenesis to induce tumors. However, as our understanding of the genetic alterations driving thyroid cancer has expanded, more sophisticated genetic engineering techniques have been employed to create models with thyroid-specific expression of these driver mutations. While driver mutations can initiate tumorigenesis, they are often insufficient to sustain cancer progression and invasion, which significantly limits their usefulness in studying advanced thyroid cancers. Recent studies exploring the genomic landscape of advanced thyroid cancer have identified several cooperating mutations, which are secondary genetic alterations that work alongside driver mutations to promote thyroid tumor progression. Indeed, mice with a combination of oncogenic drivers and common cooperating alterations have been developed, demonstrating that these alterations function in conjunction with the oncogenic driver to promote the progression to advanced thyroid cancer. These models provide important preclinical tools to explore how cooperating alterations influence the response to therapies, particularly those targeting the oncogenic driver. This review will focus on recent publications that broaden the scope of advanced thyroid cancer models by combining thyroid-specific oncogenic driver expression with various cooperating mutations.
Collapse
Affiliation(s)
- Shovan Dutta
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jeffrey A Knauf
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Otolaryngology-Head & Neck Surgery, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Qu J, Fang Y, Tao R, Zhao J, Xu T, Chen R, Zhang J, Meng K, Yang Q, Zhang K, Yan X, Sun D, Chen X. Advancing thyroid disease research: The role and potential of zebrafish model. Life Sci 2024; 357:123099. [PMID: 39374770 DOI: 10.1016/j.lfs.2024.123099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/11/2024] [Accepted: 09/28/2024] [Indexed: 10/09/2024]
Abstract
Thyroid disorders significantly affect human metabolism, cardiovascular function, skeletal health, and reproductive systems, presenting a complex challenge due to their multifactorial nature. Understanding the underlying mechanisms and developing novel therapeutic approaches require appropriate models. Zebrafish, with their genetic tractability, short life cycle, and physiological relevance, have emerged as a valuable model for investigating thyroid diseases. This review provides a comprehensive analysis of the zebrafish thyroid gland's structure and function, explores its application in modeling thyroid pathologies such as hypothyroidism, hyperthyroidism, and thyroid cancer, and discusses current limitations and possible improvements. Furthermore, it outlines future directions for zebrafish-based research, focusing on enhancing the model's relevance to human thyroid disease and its potential to expedite the development of clinical therapies.
Collapse
Affiliation(s)
- Junying Qu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Runchao Tao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jing Zhao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Ting Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Rongbing Chen
- Department of Biomedical, City university of Hong Kong, Kowloon 999077, Hong Kong
| | - Junbei Zhang
- Department of Endocrinology, Yiwu Central Hospital, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| | - Kaikai Meng
- Department of Endocrinology, Yiwu Central Hospital, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Xiaoqing Yan
- The Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China; Department of Endocrinology, Yiwu Central Hospital, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China.
| | - Xia Chen
- Department of Endocrinology, Yiwu Central Hospital, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China.
| |
Collapse
|
10
|
Cirello V, Gambale C, Nikitski AV, Masaki C, Roque J, Colombo C. Poorly differentiated thyroid carcinoma: molecular, clinico-pathological hallmarks and therapeutic perspectives. Panminerva Med 2024; 66:155-173. [PMID: 38576304 DOI: 10.23736/s0031-0808.23.05040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Poorly differentiated thyroid carcinoma (PDTC) is a rare and extremely aggressive tumor, accounting for about 2-15% of all thyroid cancer. PDTC has a distinct biological behavior compared to well-differentiated and anaplastic thyroid carcinoma and, in last years, it has been classified as a separate entity from both anatomopathological and clinical points of view. Nevertheless, there is still a lack of consensus among clinicians regarding inclusion criteria and definition of PDTC that affects its diagnosis and clinical management. Due to its rarity and difficulty in classification compared to other tumors, very few studies are available to date and series often include different histotypes in addition to PDTC. This review focuses on main studies concerning PDTC summarizing the evolution in the definition of its diagnosis criteria, clinicopathological features, management, and outcome. The data available confirm that the pathological evaluation and classification of PDTC are crucial and should therefore be standardized. Since the clinical presentation and prognosis of PDTC may vary widely depending on the different stage of the disease at diagnosis, the patient's management may differ in treatment and should be tailored to each patient. Finally, this review discusses advances in molecular insights of PDTC that, together with the implementation of both in vitro and in vivo models, will provide valuable insights into biological mechanisms of progression, metastasis, and invasion of this aggressive thyroid carcinoma. Further studies on larger, carefully selected series are needed to better assess the peculiar features of PDTC and to better define its management by focusing on the best diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Valentina Cirello
- Endocrine Oncology Unit, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Carla Gambale
- Department of Clinical and Experimental Medicine, Endocrine Unit, University Hospital of Pisa, Pisa, Italy
| | - Alyaksandr V Nikitski
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Chie Masaki
- Department of Surgery, Ito Hospital, Tokyo, Japan
| | - João Roque
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
| | - Carla Colombo
- Endocrine Oncology Unit, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy -
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Bao L, Li Y, Hu X, Gong Y, Chen J, Huang P, Tan Z, Ge M, Pan Z. Targeting SIGLEC15 as an emerging immunotherapy for anaplastic thyroid cancer. Int Immunopharmacol 2024; 133:112102. [PMID: 38652971 DOI: 10.1016/j.intimp.2024.112102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Anaplastic thyroid carcinoma (ATC) is the most aggressive subtype of thyroid cancer with few effective therapies. Though immunotherapies such as targeting PD-1/PD-L1 axis have benefited patients with solid tumor, the druggable immune checkpoints are quite limited in ATC. In our study, we focused on the anti-tumor potential of sialic acid-binding Ig-like lectins (Siglecs) in ATC. Through screening by integrating microarray datasets including 216 thyroid-cancer tissues and single-cell RNA-sequencing, SIGLEC family members CD33, SIGLEC1, SIGLEC10 and SIGLEC15 were significantly overexpressed in ATC, among which SIGLEC15 increased highest and mainly expressed on cancer cells. SIGLEC15high ATC cells are characterized by high expression of serine protease PRSS23 and cancer stem cell marker CD44. Compared with SIGLEC15low cancer cells, SIGLEC15high ATC cells exhibited higher interaction frequency with tumor microenvironment cells. Further study showed that SIGLEC15high cancer cells mainly interacted with T cells by immunosuppressive signals such as MIF-TNFRSF14 and CXCL12-CXCR4. Notably, treatment of anti-SIGLEC15 antibody profoundly increased the cytotoxic ability of CD8+ T cells in a co-culture model and zebrafish-derived ATC xenografts. Consistently, administration of anti-SIGLEC15 antibody significantly inhibited tumor growth and prolonged mouse survival in an immunocompetent model of murine ATC, which was associated with increase of M1/M2, natural killer (NK) cells and CD8+ T cells, and decrease of myeloid-derived suppressor cells (MDSCs). SIGLEC15 inhibited T cell activation by reducing NFAT1, NFAT2, and NF-κB signals. Blocking SIGLEC15 increased the secretion of IFN-γ and IL-2 in vitro and in vivo. In conclusion, our finding demonstrates that SIGLEC15 is an emerging and promising target for immunotherapy in ATC.
Collapse
Affiliation(s)
- Lisha Bao
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China
| | - Ying Li
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Xiaoping Hu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China
| | - Yingying Gong
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China
| | - Jinming Chen
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China.
| | - Zongfu Pan
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China.
| |
Collapse
|
12
|
Tahara M, Kiyota N, Imai H, Takahashi S, Nishiyama A, Tamura S, Shimizu Y, Kadowaki S, Ito KI, Toyoshima M, Hirashima Y, Ueno S, Sugitani I. A Phase 2 Study of Encorafenib in Combination with Binimetinib in Patients with Metastatic BRAF-Mutated Thyroid Cancer in Japan. Thyroid 2024; 34:467-476. [PMID: 38343359 DOI: 10.1089/thy.2023.0547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2024]
Abstract
Background: Driver mutations at BRAF V600 are frequently identified in papillary thyroid cancer and anaplastic thyroid cancer (ATC), in which BRAF inhibitors have shown clinical effectiveness. This Japanese phase 2 study evaluated the efficacy and safety of a BRAF inhibitor, encorafenib, combined with an MEK inhibitor, binimetinib, in patients with BRAF V600-mutated thyroid cancer. Methods: This phase 2, open-label, uncontrolled study was conducted at 10 institutions targeted patients with BRAF V600-mutated locally advanced or distant metastatic thyroid cancer not amenable to curative treatment who became refractory/intolerant to ≥1 previous vascular endothelial growth factor receptor-targeted regimen(s) or were considered ineligible for those. The primary endpoint was centrally assessed objective response rate (ORR). The secondary endpoints included duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. Results: We enrolled 22 patients with BRAFV600E-mutated thyroid cancer: 17 had differentiated thyroid cancer (DTC), and 5 had ATC. At data cutoff (October 26, 2022), the median follow-up was 11.5 (range = 3.4-19.0) months. The primary endpoint of centrally assessed ORR was 54.5% (95% confidence interval [CI] 32.2-75.6; partial response in 12 patients and stable disease in 10). The ORRs in patients with DTC and ATC were 47.1% (8 of 17) and 80.0% (4 of 5), respectively. The medians for DOR and PFS by central assessment and for OS were not reached in the overall population, the DTC subgroup, or the ATC subgroup. At 12 months, the rate of ongoing response was 90.9%, and the PFS and OS rates were 78.8% and 81.8%, respectively. All patients developed ≥1 adverse events (AEs): grade 3 AEs in 6 patients (27.3%). No patients developed grade 4-5 AEs. The most common grade 3 AE was lipase increased (4 patients [18.2%]). Those toxicities were mostly manageable with appropriate monitoring and dose adjustment. Conclusions: Treatment with encorafenib plus binimetinib met the primary endpoint criteria and demonstrated clinical benefit in patients with BRAFV600E-mutated thyroid cancer regardless of its histological type, such as DTC or ATC, with no new safety concerns identified. Encorafenib plus binimetinib could thus be a new treatment option for BRAF V600-mutated thyroid cancer. Clinical Trial Registration number: Japan Registry of Clinical Trials: jRCT2011200018.
Collapse
Affiliation(s)
- Makoto Tahara
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Naomi Kiyota
- Department of Medical Oncology and Hematology, Cancer Center, Kobe University Hospital, Kobe, Hyogo, Japan
| | - Hiroo Imai
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shunji Takahashi
- Department of Medical Oncology, The Cancer Institute Hospital of JFCR, Koto-ku, Tokyo, Japan
| | - Akihiro Nishiyama
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Shingo Tamura
- Department of Medical Oncology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Fukuoka, Japan
| | - Yasushi Shimizu
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Ken-Ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | | | | | | | - Iwao Sugitani
- Department of Endocrinology, Nippon Medical School Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
13
|
Han PZ, Ye WD, Yu PC, Tan LC, Shi X, Chen XF, He C, Hu JQ, Wei WJ, Lu ZW, Qu N, Wang Y, Ji QH, Ji DM, Wang YL. A distinct tumor microenvironment makes anaplastic thyroid cancer more lethal but immunotherapy sensitive than papillary thyroid cancer. JCI Insight 2024; 9:e173712. [PMID: 38478516 PMCID: PMC11141884 DOI: 10.1172/jci.insight.173712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 03/06/2024] [Indexed: 04/23/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Pei-Zhen Han
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Dong Ye
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng-Cheng Yu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li-Cheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xu-Feng Chen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cong He
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jia-Qian Hu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Jun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong-Wu Lu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Qu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing-Hai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dong-Mei Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu-Long Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Hurst ZA, Liyanarachchi S, Brock P, He H, Nabhan F, Veloski C, Toland AE, Ringel MD, Jhiang SM. Presumed Pathogenic Germ Line and Somatic Variants in African American Thyroid Cancer. Thyroid 2024; 34:378-387. [PMID: 38062767 PMCID: PMC10951570 DOI: 10.1089/thy.2023.0487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background: African American (AA) thyroid cancer patients have worse prognoses than European Americans (EA), which has been attributed to both health care disparities and possible genetic differences. We investigated the impact of both germ line and somatic variants on clinical outcome in a cohort of AA nonmedullary thyroid cancer (NMTC) patients who had received therapeutic intervention from cancer centers. Methods: Whole-exome sequencing was performed on DNA from available blood/normal tissues (N = 37) and paired tumor samples (N = 32) collected from 37 and 29 AA NMTC patients, respectively. Variants with Combined Annotation Depletion Dependent (CADD) score of ≥20 and VarSome Clinical classification of likely pathogenic or pathogenic were classified as presumed pathogenic germ line or somatic variants (PPGVs/PPSVs). PPGVs/PPSVs in cancer-related genes and PPGVs in cardiovascular risk genes were further investigated, and PPGVs/PPSVs associated with African (AFR) ancestry were identified. Results: Among 17 PPGVs identified in 16 cancer predisposition or known cancer-related genes, only WRN was previously known to associate with NMTC predisposition. Among PPSVs, BRAFV600E was most the prevalent and detected in 12 of the 29 (41%) tumors. Examining PPGVs/PPSVs among three patients who died from NMTC, one patient who died from papillary thyroid carcinoma/anaplastic thyroid carcinoma (PTC/ATC) led us to speculate that the PPGV ERCC4R799W may have increased the risk of PPSV TP53R273H acquisition. Among PPGVs identified in 18 cardiovascular risk genes, PPGVs in SC5NA, GYG1, CBS, CFTR, and SI are known to have causal and pathogenic implications in cardiovascular disease. Conclusion: In this cohort, most AA-NMTC patients exhibit favorable outcomes after therapeutic intervention given at cancer centers, suggesting that health care disparity is the major contributor for worse prognoses among AA-NMTC patients. Nevertheless, the clinical impact of PPGVs that might facilitate the acquisition of TP53 tumor mutations, and/or PPGVs that predispose individuals to adverse cardiovascular events, which could be exacerbated by therapy-induced cardiotoxicity, needs to be further explored. Integrated analysis of PPGV/PPSV profiles among NMTC patients with different stages of disease may help to identify NMTC patients who require close monitoring or proactive intervention.
Collapse
Affiliation(s)
- Zachary A. Hurst
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Sandya Liyanarachchi
- Department of Molecular Medicine and Therapeutics, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Pamela Brock
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Huiling He
- Department of Molecular Medicine and Therapeutics, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Fadi Nabhan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Colleen Veloski
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Matthew D. Ringel
- Department of Molecular Medicine and Therapeutics, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Sissy M. Jhiang
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
15
|
Landa I, Cabanillas ME. Genomic alterations in thyroid cancer: biological and clinical insights. Nat Rev Endocrinol 2024; 20:93-110. [PMID: 38049644 DOI: 10.1038/s41574-023-00920-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 12/06/2023]
Abstract
Tumours can arise from thyroid follicular cells if they acquire driver mutations that constitutively activate the MAPK signalling pathway. In addition, a limited set of additional mutations in key genes drive tumour progression towards more aggressive and less differentiated disease. Unprecedented insights into thyroid tumour biology have come from the breadth of thyroid tumour sequencing data from patients and the wide range of mutation-specific mechanisms identified in experimental models, in combination with the genomic simplicity of thyroid cancers. This knowledge is gradually being translated into refined strategies to stratify, manage and treat patients with thyroid cancer. This Review summarizes the biological underpinnings of the genetic alterations involved in thyroid cancer initiation and progression. We also provide a rationale for and discuss specific examples of how to implement genomic information to inform both recommended and investigational approaches to improve thyroid cancer prognosis, redifferentiation strategies and targeted therapies.
Collapse
Affiliation(s)
- Iñigo Landa
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
16
|
Yang J, Cheng Y, Nie Y, Tian B, Huang J, Gong R, Li Z, Zhu J, Gong Y. TRPC5 expression promotes the proliferation and invasion of papillary thyroid carcinoma through the HIF-1α/Twist pathway. Transl Oncol 2024; 39:101809. [PMID: 37918167 PMCID: PMC10638037 DOI: 10.1016/j.tranon.2023.101809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/30/2023] [Accepted: 10/15/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the effect of TRPC5 on PTC (papillary thyroid carcinoma) proliferation and invasion. METHODS Immunofluorescence and western blot were used to evaluate the expression of TRPC5 in paraffin sections and clinical tissues. Overexpression and silencing of TRPC5 to generate the cells for in vitro experiments. Wound-healing assay, transwell invasion assay, MTT assay, and in vivo tumorigenicity assay were used to determine cell proliferation and cell migration in vitro and in vivo. Real-time PCR was used to test the expression of TRPC5. Western blot was used to test the expression of downstream factors: E-cadherin, Vimentin, MMP-9, MMP-2, TRPC5, ZEB, Snail, and Twist. RESULTS The level of TRPC5 protein expression was higher in PTC than in adjacent normal thyroid tissue. TPC-1 cells overexpressing TRPC5 were more proliferative, had longer migration distances, and increased the number of invading cells. TPC-1 cells silenced with TRPC5 had a weaker proliferation capacity, shorter migration distances, and a reduced number of invading cells. Overexpression and silencing of TRPC5 modulated E-cadherin, Vimentin, MMP-9, MMP-2, TRPC5, and Twist, but did not affect ZEB and Snail. The results of tumor formation experiments in nude mice showed that inhibition of TRPC5 expression suppressed the volume and weight of transplanted tumors. CONCLUSION TRPC5 induced papillary thyroid cancer metastasis and progression via up-regulated HIF-1α signaling in vivo and in vitro. High TRPC5 expression is a biomarker for lymph node metastasis at its early stages.
Collapse
Affiliation(s)
- Jing Yang
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yue Cheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Sichuan Electric Power Hospital, China
| | - Yan Nie
- West China School of Medicine, Sichuan University, China
| | - Bole Tian
- Department of pancreatic Surgery, West China Hospital, Sichuan University, China
| | - Jing Huang
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rixiang Gong
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhihui Li
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingqiang Zhu
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanping Gong
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
17
|
Li L, Wang Z, Guo H, Lin Q. Nanomaterials: a promising multimodal theranostics platform for thyroid cancer. J Mater Chem B 2023; 11:7544-7566. [PMID: 37439780 DOI: 10.1039/d3tb01175e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Thyroid cancer is the most prevalent malignant neoplasm of the cervical region and endocrine system, characterized by a discernible upward trend in incidence over recent years. Ultrasound-guided fine needle aspiration is the current standard for preoperative diagnosis of thyroid cancer, albeit with limitations and a certain degree of false-negative outcomes. Although differentiated thyroid carcinoma generally exhibits a favorable prognosis, dedifferentiation is associated with an unfavorable clinical course. Anaplastic thyroid cancer, characterized by high malignancy and aggressiveness, remains an unmet clinical need with no effective treatments available. The emergence of nanomedicine has opened new avenues for cancer theranostics. The unique features of nanomaterials, including multifunctionality, modifiability, and various detection modes, enable non-invasive and convenient thyroid cancer diagnosis through multimodal imaging. For thyroid cancer treatment, nanomaterial-based photothermal therapy or photodynamic therapy, combined with chemotherapy, radiotherapy, or gene therapy, holds promise in reducing invasiveness and prolonging patient survival or alleviating pain in individuals with anaplastic thyroid carcinoma. Furthermore, nanomaterials enable simultaneous diagnosis and treatment of thyroid cancer. This review aims to provide a comprehensive survey of the latest developments in nanomaterials for thyroid cancer diagnosis and treatment and encourage further research in developing innovative and effective theranostic approaches for thyroid cancer.
Collapse
Affiliation(s)
- Lei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
- Department of Endocrinology, Lequn Branch, The First Hospital of Jilin University, Changchun, 130031, China.
| | - Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Hui Guo
- Department of Endocrinology, Lequn Branch, The First Hospital of Jilin University, Changchun, 130031, China.
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| |
Collapse
|
18
|
Hu H, Ng TS, Kang M, Scott E, Li R, Quintana JM, Matvey D, Vantaku VR, Weissleder R, Parangi S, Miller MA. Thyroid Cancers Exhibit Oncogene-Enhanced Macropinocytosis that Is Restrained by IGF1R and Promote Albumin-Drug Conjugate Response. Clin Cancer Res 2023; 29:3457-3470. [PMID: 37289199 PMCID: PMC10527034 DOI: 10.1158/1078-0432.ccr-22-2976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
PURPOSE Oncogene-driven macropinocytosis fuels nutrient scavenging in some cancer types, yet whether this occurs in thyroid cancers with prominent MAPK-ERK and PI3K pathway mutations remains unclear. We hypothesized that understanding links between thyroid cancer signaling and macropinocytosis might uncover new therapeutic strategies. EXPERIMENTAL DESIGN Macropinocytosis was assessed across cells derived from papillary thyroid cancer (PTC), follicular thyroid cancer (FTC), non-malignant follicular thyroid, and aggressive anaplastic thyroid cancer (ATC), by imaging fluorescent dextran and serum albumin. The impacts of ectopic BRAFV600E and mutant RAS, genetic PTEN silencing, and inhibitors targeting RET, BRAF, and MEK kinases were quantified. BrafV600E p53-/- ATC tumors in immunocompetent mice were used to measure efficacy of an albumin-drug conjugate comprising microtubule-destabilizing monomethyl auristatin E (MMAE) linked to serum albumin via a cathepsin-cleavable peptide (Alb-vc-MMAE). RESULTS FTC and ATC cells showed greater macropinocytosis than non-malignant and PTC cells. ATC tumors accumulated albumin at 8.8% injected dose per gram tissue. Alb-vc-MMAE, but not MMAE alone, reduced tumor size by >90% (P < 0.01). ATC macropinocytosis depended on MAPK/ERK activity and nutrient signaling, and increased by up to 230% with metformin, phenformin, or inhibition of IGF1Ri in monoculture but not in vivo. Macrophages also accumulated albumin and express the cognate IGF1R ligand, IGF1, which reduced ATC responsiveness to IGF1Ri. CONCLUSIONS These findings identify regulated oncogene-driven macropinocytosis in thyroid cancers and demonstrate the potential of designing albumin-bound drugs to efficiently treat them.
Collapse
Affiliation(s)
- Huiyu Hu
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
- Department of General Surgery, Xiangya Hospital, Central South University, China
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Mikyung Kang
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ella Scott
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Jeremy M. Quintana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Dylan Matvey
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Venkata R. Vantaku
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
- Department of Systems Biology, Harvard Medical School, United States
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| |
Collapse
|
19
|
Yu P, Qu N, Zhu R, Hu J, Han P, Wu J, Tan L, Gan H, He C, Fang C, Lei Y, Li J, He C, Lan F, Shi X, Wei W, Wang Y, Ji Q, Yu FX, Wang YL. TERT accelerates BRAF mutant-induced thyroid cancer dedifferentiation and progression by regulating ribosome biogenesis. SCIENCE ADVANCES 2023; 9:eadg7125. [PMID: 37647391 PMCID: PMC10468137 DOI: 10.1126/sciadv.adg7125] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/27/2023] [Indexed: 09/01/2023]
Abstract
TERT reactivation occurs frequently in human malignancies, especially advanced cancers. However, in vivo functions of TERT reactivation in cancer progression and the underlying mechanism are not fully understood. In this study, we expressed TERT and/or active BRAF (BRAF V600E) specifically in mouse thyroid epithelium. While BRAF V600E alone induced papillary thyroid cancer (PTC), coexpression of BRAF V600E and TERT resulted in poorly differentiated thyroid carcinoma (PDTC). Spatial transcriptome analysis revealed that tumors from mice coexpressing BRAF V600E and TERT were highly heterogeneous, and cell dedifferentiation was positively correlated with ribosomal biogenesis. Mechanistically, TERT boosted ribosomal RNA (rRNA) expression and protein synthesis by interacting with multiple proteins involved in ribosomal biogenesis. Furthermore, we found that CX-5461, an rRNA transcription inhibitor, effectively blocked proliferation and induced redifferentiation of thyroid cancer. Thus, TERT promotes thyroid cancer progression by inducing cancer cell dedifferentiation, and ribosome inhibition represents a potential strategy to treat TERT-reactivated cancers.
Collapse
Affiliation(s)
- Pengcheng Yu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Qu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rui Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiaqian Hu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peizhen Han
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiahao Wu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Licheng Tan
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Cong He
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chuantao Fang
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yubin Lei
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Li
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenxi He
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fei Lan
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Long Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Fagin JA, Krishnamoorthy GP, Landa I. Pathogenesis of cancers derived from thyroid follicular cells. Nat Rev Cancer 2023; 23:631-650. [PMID: 37438605 PMCID: PMC10763075 DOI: 10.1038/s41568-023-00598-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
The genomic simplicity of differentiated cancers derived from thyroid follicular cells offers unique insights into how oncogenic drivers impact tumour phenotype. Essentially, the main oncoproteins in thyroid cancer activate nodes in the receptor tyrosine kinase-RAS-BRAF pathway, which constitutively induces MAPK signalling to varying degrees consistent with their specific biochemical mechanisms of action. The magnitude of the flux through the MAPK signalling pathway determines key elements of thyroid cancer biology, including differentiation state, invasive properties and the cellular composition of the tumour microenvironment. Progression of disease results from genomic lesions that drive immortalization, disrupt chromatin accessibility and cause cell cycle checkpoint dysfunction, in conjunction with a tumour microenvironment characterized by progressive immunosuppression. This Review charts the genomic trajectories of these common endocrine tumours, while connecting them to the biological states that they confer.
Collapse
Affiliation(s)
- James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Gnana P Krishnamoorthy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iñigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
21
|
Tasoulas J, Srivastava S, Xu X, Tarasova V, Maniakas A, Karreth FA, Amelio AL. Genetically engineered mouse models of head and neck cancers. Oncogene 2023; 42:2593-2609. [PMID: 37474617 PMCID: PMC10457205 DOI: 10.1038/s41388-023-02783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
The head and neck region is one of the anatomic sites commonly afflicted by cancer, with ~1.5 million new diagnoses reported worldwide in 2020 alone. Remarkable progress has been made in understanding the underlying disease mechanisms, personalizing care based on each tumor's individual molecular characteristics, and even therapeutically exploiting the inherent vulnerabilities of these neoplasms. In this regard, genetically engineered mouse models (GEMMs) have played an instrumental role. While progress in the development of GEMMs has been slower than in other major cancer types, several GEMMs are now available that recapitulate most of the heterogeneous characteristics of head and neck cancers such as the tumor microenvironment. Different approaches have been employed in GEMM development and implementation, though each can generally recapitulate only certain disease aspects. As a result, appropriate model selection is essential for addressing specific research questions. In this review, we present an overview of all currently available head and neck cancer GEMMs, encompassing models for head and neck squamous cell carcinoma, nasopharyngeal carcinoma, and salivary and thyroid gland carcinomas.
Collapse
Affiliation(s)
- Jason Tasoulas
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sonal Srivastava
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Xiaonan Xu
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Valentina Tarasova
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Anastasios Maniakas
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florian A Karreth
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Antonio L Amelio
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
22
|
Leandro-García LJ, Landa I. Mechanistic Insights of Thyroid Cancer Progression. Endocrinology 2023; 164:bqad118. [PMID: 37503738 PMCID: PMC10403681 DOI: 10.1210/endocr/bqad118] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
Differentiated thyroid cancers (DTCs) are primarily initiated by mutations that activate the MAPK signaling cascade, typically at BRAF or RAS oncoproteins. DTCs can evolve to more aggressive forms, specifically, poorly differentiated (PDTC) and anaplastic thyroid cancers (ATC), by acquiring additional genetic alterations which deregulate key pathways. In this review, we focused on bona fide mutations involved in thyroid cancer progression for which consistent mechanistic data exist. Here we summarized the relevant literature, spanning approximately 2 decades, highlighting genetic alterations that are unquestionably enriched in PDTC/ATC. We describe the relevant functional data obtained in multiple in vitro and in vivo thyroid cancer models employed to study genetic alterations in the following genes and functional groups: TP53, effectors of the PI3K/AKT pathway, TERT promoter, members of the SWI/SNF chromatin remodeling complex, NF2, and EIF1AX. In addition, we briefly discuss other genetic alterations that are selected in aggressive thyroid tumors but for which mechanistic data is still either limited or nonexistent. Overall, we argue for the importance conveyed by preclinical studies for the clinical translation of genomic knowledge of thyroid cancers.
Collapse
Affiliation(s)
- Luis Javier Leandro-García
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Iñigo Landa
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
23
|
Xu Z, Shin HS, Kim YH, Ha SY, Won JK, Kim SJ, Park YJ, Parangi S, Cho SW, Lee KE. Modeling the tumor microenvironment of anaplastic thyroid cancer: an orthotopic tumor model in C57BL/6 mice. Front Immunol 2023; 14:1187388. [PMID: 37545523 PMCID: PMC10403231 DOI: 10.3389/fimmu.2023.1187388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/23/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Securing a well-established mouse model is important in identifying and validating new therapeutic targets for immuno-oncology. The C57BL/6 mouse is one of the most fully characterised immune system of any animal and provides powerful platform for immuno-oncology discovery. An orthotopic tumor model has been established using TBP3743 (murine anaplastic thyroid cancer [ATC]) cells in B6129SF1 hybrid mice, this model has limited data on tumor immunology than C57BL/6 inbred mice. This study aimed to establish a novel orthotopic ATC model in C57BL/6 mice and characterize the tumor microenvironment focusing immunity in the model. Methods Adapted TBP3743 cells were generated via in vivo serial passaging in C57BL/6 mice. Subsequently, the following orthotopic tumor models were established via intrathyroidal injection: B6129SF1 mice injected with original TBP3743 cells (original/129), B6129SF1 mice injected with adapted cells (adapted/129), and C57BL/6 mice injected with adapted cells (adapted/B6). Results The adapted TBP3743 cells de-differentiated but exhibited cell morphology, viability, and migration/invasion potential comparable with those of original cells in vitro. The adapted/129 contained a higher Ki-67+ cell fraction than the original/129. RNA sequencing data of orthotopic tumors revealed enhanced oncogenic properties in the adapted/129 compared with those in the original/129. In contrast, the orthotopic tumors grown in the adapted/B6 were smaller, with a lower Ki-67+ cell fraction than those in the adapted/129. However, the oncogenic properties of the tumors within the adapted/B6 and adapted/129 were similar. Immune-related pathways were enriched in the adapted/B6 compared with those in the adapted/129. Flow cytometric analysis of the orthotopic tumors revealed higher cytotoxic CD8+ T cell and monocytic-myeloid-derived suppressor cell fractions in the adapted/B6 compared with the adapted/129. The estimated CD8+ and CD4+ cell fractions in the adapted/B6 were similar to those in human ATCs but negligible in the original/B6. Conclusion A novel orthotopic tumor model of ATC was established in C57BL/6 mice. Compared with the original B6129SF1 murine model, the novel model exhibited more aggressive tumor cell behaviours and strong immune responses. We expect that this novel model contributes to the understanding tumor microenvironment and provides the platform for drug development.
Collapse
Affiliation(s)
- Zhen Xu
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Surgery, YanBian University Hospital, Yanji, Jilin, China
| | - Hyo Shik Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoo Hyung Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seong Yun Ha
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pathology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Su-jin Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Division of Surgery, Thyroid Center, Seoul National University Cancer Hospital, Seoul, Republic of Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyu Eun Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Division of Surgery, Thyroid Center, Seoul National University Cancer Hospital, Seoul, Republic of Korea
| |
Collapse
|
24
|
Choi HR, Kim K. Mouse Models to Examine Differentiated Thyroid Cancer Pathogenesis: Recent Updates. Int J Mol Sci 2023; 24:11138. [PMID: 37446316 DOI: 10.3390/ijms241311138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Although the overall prognosis of differentiated thyroid cancer (DTC), the most common endocrine malignancy, is favorable, a subset of patients exhibits aggressive features. Therefore, preclinical models that can be utilized to investigate DTC pathogenesis and novel treatments are necessary. Various mouse models have been developed based on advances in thyroid cancer genetics. This review focuses on recent progress in mouse models that have been developed to elucidate the molecular pathogenesis of DTC.
Collapse
Affiliation(s)
- Hye Ryeon Choi
- Department of Surgery, Eulji Medical Center, Eulji University School of Medicine, Seoul 01830, Republic of Korea
| | - Kwangsoon Kim
- Department of Surgery, College of Medicine, Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
25
|
Bai F, Liu X, Zhang X, Mao Z, Wen H, Ma J, Pei XH. p18INK4C and BRCA1 inhibit follicular cell proliferation and dedifferentiation in thyroid cancer. Cell Cycle 2023; 22:1637-1653. [PMID: 37345432 PMCID: PMC10361144 DOI: 10.1080/15384101.2023.2225938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/13/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023] Open
Abstract
Only 3% of thyroid cancers are medullary thyroid carcinomas (MTCs), the rest are follicular epithelial cell derived non-MTCs (NMTCs). A dysfunctional INK4-CDK4-RB pathway is detected in most of NMTCs. DNA repair defects and genome instability are associated with NMTC dedifferentiation and aggressiveness. Whether inactivation of the INK4-CDK4-RB pathway induces NMTCs and how differentiation of NMTC cells is controlled remain elusive. In this study, we generated p18Ink4c and Brca1 singly and doubly deficient mice as well as p16Ink4a and Brca1 singly and doubly deficient mice. By using these mice and human thyroid carcinoma cell lines, we discovered that loss of p18Ink4c, not p16Ink4a, in mice stimulated follicular cell proliferation and induced NMTCs. Depletion of Brca1 alone or both p16Ink4a and Brca1 did not induce thyroid tumor. Depletion of Brca1 in p18Ink4c null mice results in poorly differentiated and aggressive NMTCs with epithelial-mesenchymal transition (EMT) features and enhanced DNA damage. Knockdown of BRCA1 in thyroid carcinoma cells activated EMT and promoted tumorigenesis whereas overexpression of BRCA1 inhibited EMT. BRCA1 and EMT marker expression were inversely related in human thyroid cancers. Our finding, for the first time, demonstrates that inactivation of INK4-CDK4-RB pathway induces NMTCs and that Brca1 deficiency promotes dedifferentiation of NMTC cells. These results suggest that BRCA1 and p18INK4C collaboratively suppress thyroid tumorigenesis and progression and CDK4 inhibitors will be effective for treatment of INK4-inactivated or cyclin D-overexpressed thyroid carcinomas.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| | - Xu Zhang
- Department of Pathology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Zhuo Mao
- Department of Physiology, Shenzhen University Health Science Center, Shenzhen, China
| | - He Wen
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Jinshan Ma
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Thoracic Surgery, Xinjiang Uigur Autonomous Region People’s Hospital, Xinjiang, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
26
|
Limberg J, Egan CE, Gray KD, Singh M, Loewenstein Z, Yang Y, Riascos MC, Al Asadi H, Safe P, El Eshaky S, Liang H, Ullmann TM, Wang W, Li W, Zhang T, Xiang J, Stefanova D, Jin MM, Zarnegar R, Fahey TJ, Min IM. Activation of the JAK/STAT Pathway Leads to BRAF Inhibitor Resistance in BRAFV600E Positive Thyroid Carcinoma. Mol Cancer Res 2023; 21:397-410. [PMID: 36790391 PMCID: PMC10159921 DOI: 10.1158/1541-7786.mcr-21-0832] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/25/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023]
Abstract
A subset of thyroid cancers, recurrent differentiated thyroid cancers and anaplastic thyroid cancer (ATC), are difficult to treat by thyroidectomy and systemic therapy. A common mutation in thyroid cancer, BRAFV600E, has targetable treatment options; however, the results have been disappointing in thyroid cancers compared with BRAFV600E melanoma, as thyroid cancers quickly become resistant to BRAFV600E inhibitor (BRAFi). Here, we studied the molecular pathway that is induced in BRAFV600E thyroid cancer cells and patient-derived tumor samples in response to BRAFi, vemurafenib, using RNA-sequencing and molecular analysis. Both inducible response to BRAFi and acquired BRAFi resistance in BRAFV600E thyroid cancer cells showed significant activation of the JAK/STAT pathway. Functional analyses revealed that the combination of BRAFi and inhibitors of JAK/STAT pathway controlled BRAFV600E thyroid cancer cell growth. The Cancer Genome Atlas data analysis demonstrated that potent activation of the JAK/STAT signaling was associated with shorter recurrence rate in patients with differentiated thyroid cancer. Analysis of tumor RNA expression in patients with poorly differentiated thyroid cancer and ATC also support that enhanced activity of JAK/STAT signaling pathway is correlated with worse prognosis. Our study demonstrates that JAK/STAT pathway is activated as BRAFV600E thyroid cancer cells develop resistance to BRAFi and that this pathway is a potential target for anticancer activity and to overcome drug resistance that commonly develops to treatment with BRAFi in thyroid cancer. IMPLICATIONS Dual inhibition of BRAF and JAK/STAT signaling pathway is a potential therapeutic treatment for anticancer activity and to overcome drug resistance to BRAFi in thyroid cancer.
Collapse
Affiliation(s)
- Jessica Limberg
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Caitlin E. Egan
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Mandeep Singh
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Yanping Yang
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Hala Al Asadi
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Parima Safe
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Steve El Eshaky
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Heng Liang
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Weibin Wang
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Wei Li
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Tuo Zhang
- Genomics Resource Core Facility, Weill Cornell Medicine, New York, NY 10065
| | - Jenny Xiang
- Genomics Resource Core Facility, Weill Cornell Medicine, New York, NY 10065
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065
| | | | - Moonsoo M. Jin
- Department of Radiology, Weill Cornell Medicine, New York, NY 10065
| | - Rasa Zarnegar
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Thomas J. Fahey
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Irene M. Min
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| |
Collapse
|
27
|
Buczyńska A, Sidorkiewicz I, Kościuszko M, Adamska A, Siewko K, Dzięcioł J, Szumowski P, Myśliwiec J, Popławska-Kita A, Krętowski AJ. The Relationship between Oxidative Status and Radioiodine Treatment Qualification among Papillary Thyroid Cancer Patients. Cancers (Basel) 2023; 15:cancers15092436. [PMID: 37173902 PMCID: PMC10177082 DOI: 10.3390/cancers15092436] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Total oxidative status (TOS), total antioxidant capacity (TAC), tumor protein 53 (p53), nuclear factor kappa B (NF-κB), forkhead box protein O1 (FOXO), and sirtuin 1 (SIRT1) play crucial roles in oxidative homeostasis and the progression of papillary thyroid cancer (PTC), as previously demonstrated in the literature. Therefore, profiling these markers among PTC patients may be useful in determining their eligibility for radioiodine (RAI) treatment. Since treatment indications are based on multiple and dynamic recommendations, additional criteria for adjuvant RAI therapy are still needed. In our study, we evaluated the TOS, TAC, and serum concentrations of p53, NF-κB, FOXO, and SIRT1 to analyze the relationship between oxidative status and qualification for RAI treatment. For the purpose of this study, we enrolled 60 patients with PTC allocated for RAI treatment as the study group and 25 very low-risk PTC patients not allocated for RAI treatment as a reference group. The serum TOS and SIRT1 concentrations were significantly higher in the study group compared to the reference group (both p < 0.001), whereas the TAC and p53, NK-κB, and FOXO concentrations were significantly lower (all p < 0.05). We also demonstrated the diagnostic utility of TAC (AUC = 0.987), FOXO (AUC = 0.648), TOS (AUC = 0.664), SIRT1 (AUC = 0.709), p53 (AUC = 0.664), and NF-κB (AUC = 0.651) measurements as indications for RAI treatment based on American Thyroid Association recommendations. Our study revealed that oxidative status-related markers may become additional criteria for RAI treatment in PTC patients.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Maria Kościuszko
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Agnieszka Adamska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Katarzyna Siewko
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Janusz Dzięcioł
- Department of Human Anatomy, Medical University of Bialystok, ul. Mickiewicza 2A, 15-230 Bialystok, Poland
| | - Piotr Szumowski
- Nuclear Medicine, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Janusz Myśliwiec
- Nuclear Medicine, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Anna Popławska-Kita
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Skłodowskiej 24a, 15-276 Bialystok, Poland
| |
Collapse
|
28
|
Gao X, Hong C, Xie Y, Zeng X. Immunotherapy or targeted therapy: What will be the future treatment for anaplastic thyroid carcinoma? Front Oncol 2023; 13:1103147. [PMID: 37007127 PMCID: PMC10063970 DOI: 10.3389/fonc.2023.1103147] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare and aggressive form of thyroid carcinoma (TC). Currently, there are no effective treatments for this condition. In the past few years, targeted therapy and immunotherapy have made significant progress in ATC treatment. Several common genetic mutations have been found in ATC cells, involving different molecular pathways related to tumor progression, and new therapies that act on these molecular pathways have been studied to improve the quality of life of these patients. In 2018, the FDA approved dabrafenib combined with trametinib to treat BRAF-positive ATC, confirming its therapeutic potential. At the same time, the recent emergence of immunotherapy has also attracted wide attention from researchers. While immunotherapy for ATC is still in the experimental stage, numerous studies have shown that immunotherapy is a potential therapy for ATC. In addition, it has also been found that the combination of immunotherapy and targeted therapy may enhance the anti-tumor effect of targeted therapy. In recent years, there has been some progress in the study of targeted therapy or immunotherapy combined with radiotherapy or chemotherapy, showing the prospect of combined therapy in ATC. In this review, we analyze the response mechanism and potential effects of targeted therapy, immunotherapy, and combination therapy in ATC treatment and explore the future of treatment for ATC.
Collapse
Affiliation(s)
- Xiaoni Gao
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chengcheng Hong
- Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yang Xie
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiangtai Zeng
- Department of Thyroid and Hernia Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Thyroid Diseases, Gannan Medical University, Ganzhou, Jiangxi, China
- *Correspondence: Xiangtai Zeng,
| |
Collapse
|
29
|
Veschi V, Turdo A, Modica C, Verona F, Di Franco S, Gaggianesi M, Tirrò E, Di Bella S, Iacono ML, Pantina VD, Porcelli G, Mangiapane LR, Bianca P, Rizzo A, Sciacca E, Pillitteri I, Vella V, Belfiore A, Bongiorno MR, Pistone G, Memeo L, Colarossi L, Giuffrida D, Colarossi C, Vigneri P, Todaro M, Stassi G. Recapitulating thyroid cancer histotypes through engineering embryonic stem cells. Nat Commun 2023; 14:1351. [PMID: 36906579 PMCID: PMC10008571 DOI: 10.1038/s41467-023-36922-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
Thyroid carcinoma (TC) is the most common malignancy of endocrine organs. The cell subpopulation in the lineage hierarchy that serves as cell of origin for the different TC histotypes is unknown. Human embryonic stem cells (hESCs) with appropriate in vitro stimulation undergo sequential differentiation into thyroid progenitor cells (TPCs-day 22), which maturate into thyrocytes (day 30). Here, we create follicular cell-derived TCs of all the different histotypes based on specific genomic alterations delivered by CRISPR-Cas9 in hESC-derived TPCs. Specifically, TPCs harboring BRAFV600E or NRASQ61R mutations generate papillary or follicular TC, respectively, whereas addition of TP53R248Q generate undifferentiated TCs. Of note, TCs arise by engineering TPCs, whereas mature thyrocytes have a very limited tumorigenic capacity. The same mutations result in teratocarcinomas when delivered in early differentiating hESCs. Tissue Inhibitor of Metalloproteinase 1 (TIMP1)/Matrix metallopeptidase 9 (MMP9)/Cluster of differentiation 44 (CD44) ternary complex, in cooperation with Kisspeptin receptor (KISS1R), is involved in TC initiation and progression. Increasing radioiodine uptake, KISS1R and TIMP1 targeting may represent a therapeutic adjuvant option for undifferentiated TCs.
Collapse
Affiliation(s)
- Veronica Veschi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Alice Turdo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Chiara Modica
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Francesco Verona
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Elena Tirrò
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy.,Department of Clinical and Experimental Medicine, A.O.U. Policlinico-Vittorio Emanuele, Center of Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - Sebastiano Di Bella
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Melania Lo Iacono
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Vincenzo Davide Pantina
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Laura Rosa Mangiapane
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Paola Bianca
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | | | - Elisabetta Sciacca
- Queen Mary University, Experimental Medicine & Rheumatology, London, United Kingdom
| | - Irene Pillitteri
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Veronica Vella
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Antonino Belfiore
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Maria Rita Bongiorno
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giuseppe Pistone
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Lorenzo Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Dario Giuffrida
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Cristina Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, A.O.U. Policlinico-Vittorio Emanuele, Center of Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.,A.O.U.P. "Paolo Giaccone", University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy.
| |
Collapse
|
30
|
Nikiforovich PA, Polyakov AP, Sleptsov I V, Boyko NS, Gronskaya YA, Timofeeva N I, Chernikov RA. Targeted therapy of anaplastic thyroid cancer. HEAD AND NECK TUMORS (HNT) 2023. [DOI: 10.17650/2222-1468-2022-12-4-33-38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Introduction. Anaplastic thyroid cancer (ATC) is a very rare malignant tumor of the thyroid comprising 1–2 % of all thyroid cancers. In this pathology, response rate for standard systemic therapy is less than 15 %, and long-term results remain unsatisfactory. Additionally, there are no data conclusively showing that cytotoxic chemotherapy improves survival or quality of life in patients with ATC.Aim. To improve the results of treatment of patients with ATC through evaluation of the effectiveness of targeted therapy in cases of BRAFV600E mutation.Materials and methods. The multicenter prospective study included 29 patients with ATC IVB–C, T4a–bN1a–bM0–1. The patients were divided into 2 groups. The Group 1 (control) included 15 patients with resectable / nonresectable, metastatic / nonmetastatic ATC (without BRAFV600E mutation), stages IVB–C who received standard types of treatment (surgical intervention, radiation, and chemotherapy). The Group 2 consisted of 14 patients with nonresectable or metastatic ATC, stages IVB–C, who received combination therapy (surgical intervention, radiation, and chemotherapy) with inclusion of inhibitors of BRAF dabrafenib and trametinib in neoadjuvant and adjuvant regimens.Results. The study showed the effectiveness of targeted therapy with inhibitors of BRAF mutations in treatment of locally advanced non-operable metastatic ATC with BRAFV600E mutation. Overall response (complete response + partial response) in the Group 1 was 0 %, in the Group 2 it was 64 %. Therefore, treatment scheme dabrafenib + trametinib is a prmising approach to combination targeted therapy in patients with ATC and BRAFV600E mutation. Conclusion. Dabrafenib + trametinib is a promising combination targeted therapy option for patients with ATC with a BRAFV600 mutation demonstrates a high overall response rate, a prolonged duration of response, and an increase in survival rates with controlled toxicity.
Collapse
Affiliation(s)
- P. A. Nikiforovich
- National Medical Research Center of Endocrinology, Ministry of Health of Russia; National Medical Research Center of Radiology, Ministry of Health of Russia
| | - A. P. Polyakov
- National Medical Research Center of Radiology, Ministry of Health of Russia
| | - I. V. Sleptsov
- N.I. Pirogov Clinic of High Medical Technologies of the St. Petersburg State University
| | - N. S. Boyko
- N.I. Pirogov Clinic of High Medical Technologies of the St. Petersburg State University
| | - Yu. A. Gronskaya
- N.I. Pirogov Clinic of High Medical Technologies of the St. Petersburg State University
| | - N. I. Timofeeva
- N.I. Pirogov Clinic of High Medical Technologies of the St. Petersburg State University
| | - R. A. Chernikov
- N.I. Pirogov Clinic of High Medical Technologies of the St. Petersburg State University
| |
Collapse
|
31
|
Lang M, Longerich T, Anamaterou C. Targeted therapy with vemurafenib in BRAF(V600E)-mutated anaplastic thyroid cancer. Thyroid Res 2023; 16:5. [PMID: 36855200 PMCID: PMC9976495 DOI: 10.1186/s13044-023-00147-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/08/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC) is one of the most aggressive malignancies, representing less than 5% of all thyroid carcinomas. Τhe median survival is limited to months due to the resistance of ATC to surgery, radioiodine therapy, radiotherapy and chemotherapy. This review will cover novel agents involving several cellular signaling pathways including the BRAF pathway. The BRAF inhibitor vemurafenib improves survival among patients with metastatic melanoma, hairy-cell leukemia and intracranial neoplasms with BRAF gene mutations. The frequency of a BRAF (V600E) mutation in ATC is about 25%. CASE PRESENTATION We report the first case of a marked partial response to adjuvant first line monotherapy with vemurafenib in BRAF V600E-mutated ATC. The 78-year-old man showed a sustained response for 7 months, thereafter scans revealed progressive disease and the patient died 10 months after first diagnosis. This case report is accompanied by a comprehensive review of current strategies and tools for ATC treatment. CONCLUSIONS This case and the review of current data confirm the benefit of BRAF inhibition in BRAF-mutated ATC, limited by acquired resistance to targeted therapy.
Collapse
Affiliation(s)
- Matthias Lang
- Department of General-, Visceral- and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany.
| | - Thomas Longerich
- grid.5253.10000 0001 0328 4908Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Chrysanthi Anamaterou
- grid.7700.00000 0001 2190 4373Department of Nuclear Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
32
|
Song Y, Bai Y, Wang J, Xu G, Wang T, Zhang B. Pathologic complete response after neoadjuvant v-Raf murine sarcoma viral oncogene homolog B (BRAF) inhibition combined with immunotherapy therapy for anaplastic thyroid carcinoma: a case report and literature review. Endocr J 2023; 70:223-228. [PMID: 36351596 DOI: 10.1507/endocrj.ej22-0366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a highly malignant tumor with invasive nature. Most patients present with locally advanced and/or distant metastatic diseases that are difficult to treat. We report a case of a previously inoperable patient with v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutated ATC. After a trial of neoadjuvant Dabrafenib/Trametinib with immunotherapy, the tumor became operable, and surgical pathology indicated a pathologic complete response (pCR). We also reviewed cases from the literature that utilized neoadjuvant BRAF-directed therapy in ATCs. These cases emphasize that BRAF-and immune-directed therapy is a feasible option in patients with inoperable ATC and may lead to improved outcomes.
Collapse
Affiliation(s)
- Yuntao Song
- Department of Head and Neck Surgery, Peking University Cancer Hospital and Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing, China
| | - Yanhua Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing, China
- Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiaxin Wang
- Department of Head and Neck Surgery, Peking University Cancer Hospital and Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing, China
| | - Guohui Xu
- Department of Head and Neck Surgery, Peking University Cancer Hospital and Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing, China
| | - Tianxiao Wang
- Department of Head and Neck Surgery, Peking University Cancer Hospital and Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing, China
| | - Bin Zhang
- Department of Head and Neck Surgery, Peking University Cancer Hospital and Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing, China
| |
Collapse
|
33
|
Transcriptomic Analysis Reveals Dysregulation of the Mycobiome and Archaeome and Distinct Oncogenic Characteristics according to Subtype and Gender in Papillary Thyroid Carcinoma. Int J Mol Sci 2023; 24:ijms24043148. [PMID: 36834564 PMCID: PMC9967748 DOI: 10.3390/ijms24043148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Papillary Thyroid Carcinoma (PTC) is characterized by unique tumor morphology, treatment response, and patient outcomes according to subtype and gender. While previous studies have implicated the intratumor bacterial microbiome in the incidence and progression of PTC, few studies have investigated the potential role of fungal and archaeal species in oncogenesis. In this study, we aimed to characterize the intratumor mycobiome and archaeometry in PTC with respect to its three primary subtypes: Classical (CPTC), Follicular Variant (FVPTC), and Tall Cell (TCPTC), and also with respect to gender. RNA-sequencing data were downloaded from The Cancer Genome Atlas (TCGA), including 453 primary tumor tissue samples and 54 adjacent solid tissue normal samples. The PathoScope 2.0 framework was used to extract fungal and archaeal microbial read counts from raw RNA-sequencing data. Overall, we found that the intratumor mycobiome and archaeometry share significant similarities in CPTC, FVPTC, and TCPTC, although most dysregulated species in CPTC are underabundant compared to normal. Furthermore, differences between the mycobiome and archaeometry were more significant between males and females, with a disproportionate number of fungal species overabundant in female tumor samples. Additionally, the expression of oncogenic PTC pathways was distinct across CPTC, FVPTC, and TCPTC, indicating that these microbes may uniquely contribute to PTC pathogenesis in each subtype. Furthermore, differences in the expression of these pathways were observed between males and females. Finally, we found a specific panel of fungi to be dysregulated in BRAF V600E-positive tumors. This study demonstrates the potential importance of microbial species to PTC incidence and oncogenesis.
Collapse
|
34
|
Zhang L, Xu M, Zhang W, Zhu C, Cui Z, Fu H, Ma Y, Huang S, Cui J, Liang S, Huang L, Wang H. Three-dimensional genome landscape comprehensively reveals patterns of spatial gene regulation in papillary and anaplastic thyroid cancers: a study using representative cell lines for each cancer type. Cell Mol Biol Lett 2023; 28:1. [PMID: 36609218 PMCID: PMC9825046 DOI: 10.1186/s11658-022-00409-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/21/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Spatial chromatin structure is intricately linked with somatic aberrations, and somatic mutations of various cancer-related genes, termed co-mutations (CoMuts), occur in certain patterns during cancer initiation and progression. The functional mechanisms underlying these genetic events remain largely unclear in thyroid cancer (TC). With discrepant differentiation, papillary thyroid cancer (PTC) and anaplastic thyroid cancer (ATC) differ greatly in characteristics and prognosis. We aimed to reveal the spatial gene alterations and regulations between the two TC subtypes. METHODS We systematically investigated and compared the spatial co-mutations between ATC (8305C), PTC (BCPAP and TPC-1), and normal thyroid cells (Nthy-ori-3-1). We constructed a framework integrating whole-genome sequencing (WGS), high-throughput chromosome conformation capture (Hi-C), and transcriptome sequencing, to systematically detect the associations between the somatic co-mutations of cancer-related genes, structural variations (SVs), copy number variations (CNVs), and high-order chromatin conformation. RESULTS Spatial co-mutation hotspots were enriched around topologically associating domains (TADs) in TC. A common set of 227 boundaries were identified in both ATC and PTC, with significant overlaps between them. The spatial proximities of the co-mutated gene pairs in the two TC types were significantly greater than in the gene-level and overall backgrounds, and ATC cells had higher TAD contact frequency with CoMuts > 10 compared with PTC cells. Compared with normal thyroid cells, in ATC the number of the created novel three-dimensional chromatin structural domains increased by 10%, and the number of shifted TADs decreased by 7%. We found five TAD blocks with CoMut genes/events specific to ATC with certain mutations in genes including MAST-NSUN4, AM129B/TRUB2, COL5A1/PPP1R26, PPP1R26/GPSM1/CCDC183, and PRAC2/DLX4. For the majority of ATC and PTC cells, the HOXA10 and HIF2α signals close to the transcription start sites of CoMut genes within TADs were significantly stronger than those at the background. CNV breakpoints significantly overlapped with TAD boundaries in both TC subtypes. ATCs had more CNV losses overlapping with TAD boundaries, and noncoding SVs involved in intrachromosomal SVs, amplified inversions, and tandem duplication differed between ATC and PTC. TADs with short range were more abundant in ATC than PTC. More switches of A/B compartment types existed in ATC cells compared with PTC. Gene expression was significantly synchronized, and orchestrated by complex epigenetics and regulatory elements. CONCLUSION Chromatin interactions and gene alterations and regulations are largely heterogeneous in TC. CNVs and complex SVs may function in the TC genome by interplaying with TADs, and are largely different between ATC and PTC. Complexity of TC genomes, which are highly organized by 3D genome-wide interactions mediating mutational and structural variations and gene activation, may have been largely underappreciated. Our comprehensive analysis may provide key evidence and targets for more customized diagnosis and treatment of TC.
Collapse
Affiliation(s)
- Linlin Zhang
- grid.412987.10000 0004 0630 1330Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Miaomiao Xu
- grid.412987.10000 0004 0630 1330Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Wanchun Zhang
- grid.470966.aDepartment of Nuclear Medicine, Shanxi Bethune Hospital (Shanxi Academy of Medical Sciences), Taiyuan, 03003 China
| | - Chuanying Zhu
- grid.16821.3c0000 0004 0368 8293Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092 China
| | - Zhilei Cui
- grid.412987.10000 0004 0630 1330Department of Respiratory Medicine, XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Hongliang Fu
- grid.412987.10000 0004 0630 1330Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Yufei Ma
- grid.412987.10000 0004 0630 1330Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Shuo Huang
- grid.412987.10000 0004 0630 1330Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Jian Cui
- BioGenius Bioinformatics Institute, Shanghai, 200050 People’s Republic of China
| | - Sheng Liang
- grid.412987.10000 0004 0630 1330Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Lei Huang
- grid.16821.3c0000 0004 0368 8293Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hui Wang
- grid.412987.10000 0004 0630 1330Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| |
Collapse
|
35
|
Jeon MJ, Haugen BR. Preclinical Models of Follicular Cell-Derived Thyroid Cancer: An Overview from Cancer Cell Lines to Mouse Models. Endocrinol Metab (Seoul) 2022; 37:830-838. [PMID: 36604954 PMCID: PMC9816502 DOI: 10.3803/enm.2022.1636] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/11/2022] [Indexed: 12/28/2022] Open
Abstract
The overall prognosis of thyroid cancer is excellent, but some patients have grossly invasive disease and distant metastases with limited responses to systemic therapies. Thus, relevant preclinical models are needed to investigate thyroid cancer biology and novel treatments. Different preclinical models have recently emerged with advances in thyroid cancer genetics, mouse modeling and new cell lines. Choosing the appropriate model according to the research question is crucial to studying thyroid cancer. This review will discuss the current preclinical models frequently used in thyroid cancer research, from cell lines to mouse models, and future perspectives on patient-derived and humanized preclinical models in this field.
Collapse
Affiliation(s)
- Min Ji Jeon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bryan R. Haugen
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Corresponding author: Bryan R. Haugen. Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, 12801 East 17th Avenue, Aurora, CO 80045, USA Tel: +1-303-724-3921, Fax: +1-303-724-3920, E-mail:
| |
Collapse
|
36
|
Preclinical Models of Neuroendocrine Neoplasia. Cancers (Basel) 2022; 14:cancers14225646. [PMID: 36428741 PMCID: PMC9688518 DOI: 10.3390/cancers14225646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Neuroendocrine neoplasia (NENs) are a complex and heterogeneous group of cancers that can arise from neuroendocrine tissues throughout the body and differentiate them from other tumors. Their low incidence and high diversity make many of them orphan conditions characterized by a low incidence and few dedicated clinical trials. Study of the molecular and genetic nature of these diseases is limited in comparison to more common cancers and more dependent on preclinical models, including both in vitro models (such as cell lines and 3D models) and in vivo models (such as patient derived xenografts (PDXs) and genetically-engineered mouse models (GEMMs)). While preclinical models do not fully recapitulate the nature of these cancers in patients, they are useful tools in investigation of the basic biology and early-stage investigation for evaluation of treatments for these cancers. We review available preclinical models for each type of NEN and discuss their history as well as their current use and translation.
Collapse
|
37
|
Busaidy NL, Konda B, Wei L, Wirth LJ, Devine C, Daniels GA, DeSouza JA, Poi M, Seligson ND, Cabanillas ME, Sipos JA, Ringel MD, Eisfeld AK, Timmers C, Shah MH. Dabrafenib Versus Dabrafenib + Trametinib in BRAF-Mutated Radioactive Iodine Refractory Differentiated Thyroid Cancer: Results of a Randomized, Phase 2, Open-Label Multicenter Trial. Thyroid 2022; 32:1184-1192. [PMID: 35658604 PMCID: PMC9595631 DOI: 10.1089/thy.2022.0115] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background: Oncogenic BRAF mutations are commonly found in advanced differentiated thyroid cancer (DTC), and reports have shown efficacy of BRAF inhibitors in these tumors. We investigated the difference in response between dabrafenib monotherapy and dabrafenib + trametinib therapy in patients with BRAF-mutated radioactive iodine refractory DTC. Methods: In this open-label randomized phase 2 multicenter trial, patients aged ≥18 years with BRAF-mutated radioactive iodine refractory DTC with progressive disease by Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 within 13 months before enrollment were eligible. Patients were randomly assigned to receive dabrafenib alone or dabrafenib + trametinib. The primary endpoint was objective response rate by modified RECIST (minor response of -20% to -29%, partial and complete response) within the first 24 weeks of therapy. Trial Registration Number: NCT01723202. Results: A total of 53 patients were enrolled. The objective response rate (modified RECIST) was 42% (11/26 [95% confidence interval {CI} 23-63%]) with dabrafenib versus 48% (13/27 [CI 29-68%]) with dabrafenib + trametinib (p = 0.67). Objective response rate (RECIST 1.1) was 35% (9/26 [CI 17-56%]) with dabrafenib and 30% (8/27 [CI 14-51%]) with dabrafenib + trametinib. Most common treatment-related adverse events included skin and subcutaneous tissue disorders (17/26, 65%), fever (13/26, 50%), hyperglycemia (12/26, 46%) with dabrafenib alone and fever (16/27, 59%), nausea, chills, fatigue (14/27, 52% each) with dabrafenib + trametinib. There were no treatment-related deaths. Conclusions: Combination dabrafenib + trametinib was not superior in efficacy compared to dabrafenib monotherapy in patients with BRAF-mutated radioiodine refractory progressive DTC.
Collapse
Affiliation(s)
- Naifa L. Busaidy
- Division of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bhavana Konda
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Lai Wei
- Department of Biomedical Informatics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Lori J. Wirth
- Division of Hematology and Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Catherine Devine
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gregory A. Daniels
- Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, California, USA
| | - Jonas A. DeSouza
- Division of Medical Oncology, The University of Chicago, Chicago, Illinois, USA
| | - Ming Poi
- Department of Pharmacology, The Ohio State University, Columbus, Ohio, USA
| | - Nathan D. Seligson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Maria E. Cabanillas
- Division of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer A. Sipos
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University and The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Matthew D. Ringel
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University and The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Ann-Kathrin Eisfeld
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Cynthia Timmers
- Translational Sciences Discovery Lab, Incyte Corporation, Wilmington, Delaware, USA
| | - Manisha H. Shah
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
38
|
Pani F, Caria P, Yasuda Y, Makoto M, Mariotti S, Leenhardt L, Roshanmehr S, Caturegli P, Buffet C. The Immune Landscape of Papillary Thyroid Cancer in the Context of Autoimmune Thyroiditis. Cancers (Basel) 2022; 14:cancers14174287. [PMID: 36077831 PMCID: PMC9454449 DOI: 10.3390/cancers14174287] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The association between papillary thyroid cancer and Hashimoto’s thyroiditis went through a long-standing human debate recently elucidated by the establishment of a novel mouse model. Papillary thyroid carcinoma is an excellent model for studying the tumor immune microenvironment because it is naturally accompanied by immune cells, making it a good candidate for the treatment with immune checkpoint inhibitors. Abstract Papillary thyroid cancer (PTC) often co-occurs with Hashimoto’s thyroiditis, an association that has long been reported in clinical studies, remaining controversial. Experimental evidence has recently shown that pre-existing thyroiditis has a beneficial effect on PTC growth and progression by a distinctive expansion of effector memory CD8 T cells. Although the link between inflammation and PTC might involve different components of the immune system, a deep characterization of them which includes T cells, B cells and tertiary lymphoid structures, Mye-loid cells, Neutrophils, NK cells and dendritic cells will be desirable. The present review article considers the role of the adaptive and innate immune response surrounding PTC in the context of Hashimoto’s thyroiditis. This review will focus on the current knowledge by in vivo and in vitro studies specifically performed on animals’ models; thyroid cancer cells and human samples including (i) the dual role of tumor-infiltrating lymphocytes; (ii) the emerging role of B cells and tertiary lymphoid structures; (iii) the role of myeloid cells, dendritic cells, and natural killer cells; (iv) the current knowledge of the molecular biomarkers implicated in the complex link between thyroiditis and PTC and the potential implication of cancer immunotherapy in PTC patients in the context of thyroiditis.
Collapse
Affiliation(s)
- Fabiana Pani
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
- Correspondence: or
| | - Paola Caria
- Department of Biomedical Sciences, Biochemistry, Biology and Genetics Unit, University of Cagliari, Cittadella Universitaria di Monserrato, SP 8, Km 0.700, Monserrato, 09042 Cagliari, Italy
| | - Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Miyara Makoto
- Inserm, Centre d’Immunologie et des Maladies Infectieuses-Paris (CIMI-PARIS), AP-HP Hôpital Pitié-Salpêtrière, Sorbonne Université, 75013 Paris, France
| | - Stefano Mariotti
- Department of Medical Sciences and Public Health, Endocrinology Unit, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Laurence Leenhardt
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
| | - Solmaz Roshanmehr
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Patrizio Caturegli
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Camille Buffet
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
| |
Collapse
|
39
|
Wang JR, Montierth M, Xu L, Goswami M, Zhao X, Cote G, Wang W, Iyer P, Dadu R, Busaidy NL, Lai SY, Gross ND, Ferrarotto R, Lu C, Gunn GB, Williams MD, Routbort M, Zafereo ME, Cabanillas ME. Impact of Somatic Mutations on Survival Outcomes in Patients With Anaplastic Thyroid Carcinoma. JCO Precis Oncol 2022; 6:e2100504. [PMID: 35977347 PMCID: PMC10530586 DOI: 10.1200/po.21.00504] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/02/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Anaplastic thyroid carcinoma (ATC) uniformly present with aggressive disease, but the mutational landscape of tumors varies. We aimed to determine whether tumor mutations affect survival outcomes in ATC. MATERIALS AND METHODS Patients who underwent mutation sequencing using targeted gene panels between 2005 and 2019 at a tertiary referral center were included. Associations between mutation status and survival outcomes were assessed using Cox proportional hazards models. RESULTS A total of 202 patients were included, where 122 died of ATC (60%). The median follow-up was 31 months (interquartile range, 18-45 months). The most common mutations were in TP53 (59%), BRAF (41%), TERT promoter (37%), and the RAS gene family (22%). Clinicopathologic characteristics and overall survival (OS) significantly correlated with mutations in BRAFV600E and RAS, which were mutually exclusive. The BRAFV600E mutation was associated with the presence of a papillary thyroid carcinoma precursor and significantly better OS (median OS: 24 months). RAS-mutated patients more commonly presented without cervical lymph node involvement but had the worst OS (median OS: 6 months). Tumors that were wild-type for both BRAF and RAS were enriched for NF1 mutations and harbored intermediate prognosis (median OS: 15 months). In multivariate analyses, RAS mutations were associated with a more than 2.5-fold higher risk of death (adjusted hazard ratio, 2.64; 95% CI, 1.66 to 4.20) compared with BRAFV600E. In patients treated with BRAF-directed therapy (n = 60), disease progression occurred in 48% of patients (n = 29). The median progression-free survival was 14 months. The presence of a TP53 mutation was independently associated with reduced progression-free survival in BRAFV600E-mutated patients treated with BRAF-directed therapy (adjusted hazard ratio, 2.89; 95% CI, 1.35 to 6.21). CONCLUSION Mutation analysis provides prognostic information in ATC and should be incorporated into routine clinical care.
Collapse
Affiliation(s)
- Jennifer Rui Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Matthew Montierth
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Li Xu
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Maitrayee Goswami
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Xiao Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Gilbert Cote
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Priyanka Iyer
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Ramona Dadu
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Neil D Gross
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Renata Ferrarotto
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Charles Lu
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Gary Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Michelle D Williams
- Department of Pathology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Mark Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Mark E Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| |
Collapse
|
40
|
Ohno K, Shibata T, Ito KI. Epidermal growth factor receptor activation confers resistance to lenvatinib in thyroid cancer cells. Cancer Sci 2022; 113:3193-3210. [PMID: 35723021 PMCID: PMC9459297 DOI: 10.1111/cas.15465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
Thyroid cancer is the most common endocrine malignancy. A multitargeted tyrosine kinase inhibitor, lenvatinib, has been used for the treatment of advanced thyroid cancer. To elucidate the mechanism of resistance to lenvatinib in thyroid cancer cells, we established lenvatinib‐resistant sublines and analyzed the molecular mechanisms of resistance. Two thyroid cancer cell lines (TPC‐1 and FRO) were used, and resistant sublines for lenvatinib (TPC‐1/LR, FRO/LR) were established. In TPC‐1/LR, the phosphorylation of epidermal growth factor receptor (EGFR), extracellular signal‐regulated kinase (ERK), and Akt was enhanced whereas in FRO/LR, the phosphorylation of EGFR and downstream signal transduction molecules was not enhanced. The addition of epidermal growth factor decreased sensitivity to lenvatinib in TPC‐1 and FRO. The combination of EGFR inhibitors lapatinib and lenvatinib significantly inhibited the growth of TPC‐1/LR in both in vitro and mouse xenograft models. Short‐term exposure to lenvatinib enhanced the phosphorylation of EGFR in six thyroid cancer cell lines regardless of their histological origin or driver gene mutations; however, phosphorylation of ERK was enhanced in all cells except TPC‐1. A synergistic growth‐inhibitory effect was observed in three thyroid cancer cell lines, including intrinsically lenvatinib‐resistant cells. The results indicate that signal transduction via the EGFR pathway may be involved in the development of lenvatinib resistance in thyroid cancer cells. The inhibition of the EGFR pathway simultaneously by an EGFR inhibitor may have therapeutic potential for overcoming lenvatinib resistance in thyroid cancer.
Collapse
Affiliation(s)
- Koichi Ohno
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Tomohiro Shibata
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Ken-Ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| |
Collapse
|
41
|
Bertol BC, Bales ES, Calhoun JD, Mayberry A, Ledezma ML, Sams SB, Orlicky DJ, Donadi EA, Haugen BR, French JD. Lenvatinib Plus Anti-PD-1 Combination Therapy for Advanced Cancers: Defining Mechanisms of Resistance in an Inducible Transgenic Model of Thyroid Cancer. Thyroid 2022; 32:153-163. [PMID: 34641722 PMCID: PMC8861922 DOI: 10.1089/thy.2021.0371] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background: Combination therapy with lenvatinib plus programmed death-1 (PD-1) immune checkpoint blockades (ICBs) is under investigation in many solid tumors, including thyroid cancer. Lenvatinib is known to reduce angiogenesis and may overturn the immunosuppressive effects of vascular endothelial growth factor in the tumor microenvironment. Previous studies investigating the effects of VEGF receptor inhibition on the immune response were performed in rapidly growing tumor models where immune equilibrium is not established before treatment. We hypothesize that physiologically relevant preclinical models are necessary to define mechanisms of resistance to immune-targeted combination therapies. Methods: We utilized the TPO-CreER/BrafV600E/wt/Trp53Δex2-10/Δex2-10 inducible transgenic model of advanced thyroid cancer to investigate lenvatinib treatment in the context of an anti-PD-1 ICB. Following tumor establishment, 3.5 months postinduction, mice were treated with high- (10 mg/kg) or low-dose (2 mg/kg) lenvatinib, anti-PD-1, or combination of lenvatinib with anti-PD-1. Tumor volume and lung metastases were assessed in each group. Immune infiltrate was characterized by flow cytometry and immunohistochemistry, and TCRß sequencing was performed to further investigate the T cell response. Results: Both low- and high-dose lenvatinib reduced tumor volume, while anti-PD-1 had no effect, alone or in combination. Although both low- and high-dose lenvatinib reduced vascular density, low-dose lenvatinib was superior in controlling tumor size. Lung metastases and survival were not improved with therapy despite the effects of lenvatinib on primary tumor size. Low-dose lenvatinib treatment led to a subtle reduction in the dominant Ly6G+CD11b+ myeloid cell population and was associated with increased CD4+ T cell infiltrate and enrichment in 4-1BB+ and granzyme B+ CD4+ T cells and FoxP3+ regulatory T cells. Polyclonal T cell expansion was evident in the majority of mice, suggesting that a tumor-specific T cell response was generated. Conclusions: The effects of lenvatinib on the immune response were most pronounced in mice treated with low-dose lenvatinib, suggesting that dose should be considered in clinical application. While the immune-modulating potential of lenvatinib is encouraging, alterations in the immune milieu and T cell activation status were insufficient to sustain durable tumor regression, even with added anti-PD-1. Additional studies are necessary to develop more effective combination approaches in low-mutation burden tumors, such as thyroid cancer.
Collapse
Affiliation(s)
- Bruna C. Bertol
- Postgraduate Program of Basic and Applied Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Aurora, Colorado, USA
| | - Elise S. Bales
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Aurora, Colorado, USA
| | - Jacob D. Calhoun
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Aurora, Colorado, USA
| | - Alanna Mayberry
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Aurora, Colorado, USA
| | - Melissa L. Ledezma
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Aurora, Colorado, USA
| | - Sharon B. Sams
- Department of Pathology; University of Colorado, Aurora, Colorado, USA
| | - David J. Orlicky
- Department of Pathology; University of Colorado, Aurora, Colorado, USA
| | - Eduardo A. Donadi
- Postgraduate Program of Basic and Applied Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Division of Clinical Immunology, Department of Medicine; Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bryan R. Haugen
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Aurora, Colorado, USA
| | - Jena D. French
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Aurora, Colorado, USA
- Address correspondence to: Jena D. French, PhD, Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, 12801 E. 17th Avenue, RC1 South, 7401D, Campus Box 8106, Aurora, CO 80045, USA
| |
Collapse
|
42
|
Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, Wen PY, Zielinski CC, Cabanillas ME, Boran A, Ilankumaran P, Burgess P, Romero Salas T, Keam B. Dabrafenib plus trametinib in patients with BRAF V600E–mutant anaplastic thyroid cancer: updated analysis from the phase II ROAR basket study. Ann Oncol 2022; 33:406-415. [PMID: 35026411 PMCID: PMC9338780 DOI: 10.1016/j.annonc.2021.12.014] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Combined therapy with dabrafenib plus trametinib was approved in several countries for treatment of BRAF V600E-mutant anaplastic thyroid cancer (ATC) based on an earlier interim analysis of 23 response-assessable patients in the ATC cohort of the phase II Rare Oncology Agnostic Research (ROAR) basket study. We report an updated analysis describing the efficacy and safety of dabrafenib plus trametinib in the full ROAR ATC cohort of 36 patients with ~4 years of additional study follow-up. Patients and methods: ROAR (NCT02034110) is an open-label, nonrandomized, phase II basket study evaluating dabrafenib plus trametinib in BRAF V600E-mutant rare cancers. The ATC cohort comprised 36 patients with unresectable or metastatic ATC who received dabrafenib 150 mg twice daily plus trametinib 2 mg once daily orally until disease progression, unacceptable toxicity, or death. The primary endpoint was investigator-assessed overall response rate (ORR) per Response Evaluation Criteria in Solid Tumors version 1.1. Secondary endpoints were duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. Results: At data cutoff (14 September 2020), median follow-up was 11.1 months (range, 0.9–76.6 months). The investigator-assessed ORR was 56% (95% confidence interval, 38.1% to 72.1%), including three complete responses; the 12-month DOR rate was 50%. Median PFS and OS were 6.7 and 14.5 months, respectively. The respective 12-month PFS and OS rates were 43.2% and 51.7%, and the 24-month OS rate was 31.5%. No new safety signals were identified with additional follow-up, and adverse events were consistent with the established tolerability of dabrafenib plus trametinib. Conclusions: These updated results confirm the substantial clinical benefit and manageable toxicity of dabrafenib plus trametinib in BRAF V600E-mutant ATC. Dabrafenib plus trametinib notably improved long-term survival and represents a meaningful treatment option for this rare, aggressive cancer.
Collapse
Affiliation(s)
- V Subbiah
- The University of Texas MD Anderson Cancer Center, Houston
| | | | | | - J Y Cho
- Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| | | | - J C Soria
- Institut Gustave Roussy, University of Paris-Sud, and University of Paris-Saclay, Villejuif, France
| | - P Y Wen
- Dana-Farber Cancer Institute, Boston, USA
| | | | - M E Cabanillas
- The University of Texas MD Anderson Cancer Center, Houston
| | - A Boran
- Novartis Pharmaceuticals Corporation, East Hanover, USA
| | - P Ilankumaran
- Novartis Pharmaceuticals Corporation, East Hanover, USA
| | - P Burgess
- Novartis Pharma AG, Basel, Switzerland
| | | | - B Keam
- Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Lee M, Untch BR, Xu B, Ghossein R, Han C, Kuo F, Valero C, Nadeem Z, Patel N, Makarov V, Dogan S, Wong RJ, Sherman EJ, Ho AL, Chan TA, Fagin JA, Morris LGT. Genomic and Transcriptomic Correlates of Thyroid Carcinoma Evolution after BRAF Inhibitor Therapy. Mol Cancer Res 2022; 20:45-55. [PMID: 34635506 PMCID: PMC8738128 DOI: 10.1158/1541-7786.mcr-21-0442] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/12/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Targeted inhibition of BRAF V600E achieves tumor control in a subset of advanced thyroid tumors. Nearly all tumors develop resistance, and some have been observed to subsequently undergo dedifferentiation. The molecular alterations associated with thyroid cancer dedifferentiation in the setting of BRAF inhibition are unknown. We analyzed targeted next-generation sequencing data from 639 advanced, recurrent and/or metastatic thyroid carcinomas, including 15 tumors that were treated with BRAF inhibitor drugs and had tissue sampled during or posttreatment, 8 of which had matched pretherapy samples. Pre- and posttherapy tissues from one additional patient were profiled with whole-exome sequencing and RNA expression profiling. Mutations in genes comprising the SWI/SNF chromatin remodeling complex and the PI3K-AKT-mTOR, MAPK, and JAK-STAT pathways all increased in prevalence across more dedifferentiated thyroid cancer histologies. Of 7 thyroid cancers that dedifferentiated after BRAF inhibition, 6 had mutations in these pathways. These mutations were mostly absent from matched pretreatment samples and were rarely detected in tumors that did not dedifferentiate. Additional analyses in one of the vemurafenib-treated tumors before and after anaplastic transformation revealed the emergence of an oncogenic PIK3CA mutation, activation of ERK signaling, dedifferentiation, and development of an immunosuppressive tumor microenvironment. These findings validate earlier preclinical data implicating these genetic pathways in resistance to BRAF inhibitors, and suggest that genetic alterations mediating acquired drug resistance may also promote thyroid tumor dedifferentiation. IMPLICATIONS: The possibility that thyroid cancer dedifferentiation may be attributed to selective pressure applied by BRAF inhibitor-targeted therapy should be investigated further.
Collapse
Affiliation(s)
- Mark Lee
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian R Untch
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Catherine Han
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fengshen Kuo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina Valero
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zaineb Nadeem
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neal Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - James A Fagin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luc G T Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
44
|
Sex Bias in Differentiated Thyroid Cancer. Int J Mol Sci 2021; 22:ijms222312992. [PMID: 34884794 PMCID: PMC8657786 DOI: 10.3390/ijms222312992] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 01/03/2023] Open
Abstract
Differentiated thyroid cancers are more frequent in women than in men. These different frequencies may depend on differences in patient's behavior and in thyroid investigations. However, an impact on sexual hormones is likely, although this has been insufficiently elucidated. Estrogens may increase the production of mutagenic molecules in the thyroid cell and favor the proliferation and invasion of tumoral cells by regulating both the thyrocyte enzymatic machinery and the inflammatory process associated with tumor growth. On the other hand, the worse prognosis of thyroid cancer associated with the male gender is poorly explained.
Collapse
|
45
|
Iesato A, Li S, Roti G, Hacker MR, Fischer AH, Nucera C. Lenvatinib Targets PDGFR-β Pericytes and Inhibits Synergy With Thyroid Carcinoma Cells: Novel Translational Insights. J Clin Endocrinol Metab 2021; 106:3569-3590. [PMID: 34302727 PMCID: PMC8864753 DOI: 10.1210/clinem/dgab552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 12/22/2022]
Abstract
CONTEXT Pericyte populations abundantly express tyrosine kinases (eg, platelet-derived growth factor receptor-β [PDGFR-β]) and impact therapeutic response. Lenvatinib is a clinically available tyrosine kinase inhibitor that also targets PDGFR-β. Duration of therapeutic response was shorter in patients with greater disease burden and metastasis. Patients may develop drug resistance and tumor progression. OBJECTIVES Develop a gene signature of pericyte abundance to assess with tumor aggressiveness and determine both the response of thyroid-derived pericytes to lenvatinib and their synergies with thyroid carcinoma-derived cells. DESIGN Using a new gene signature, we estimated the relative abundance of pericytes in papillary thyroid carcinoma (PTC) and normal thyroid (NT) TCGA samples. We also cocultured CD90+;PAX8- thyroid-derived pericytes and BRAFWT/V600E-PTC-derived cells to determine effects of coculture on paracrine communications and lenvatinib response. RESULTS Pericyte abundance is significantly higher in BRAFV600E-PTC with hTERT mutations and copy number alterations compared with NT or BRAFWT-PTC samples, even when data are corrected for clinical-pathologic confounders. We have identified upregulated pathways important for tumor survival, immunomodulation, RNA transcription, cell-cycle regulation, and cholesterol metabolism. Pericyte growth is significantly increased by platelet-derived growth factor-BB, which activates phospho(p)-PDGFR-β, pERK1/2, and pAKT. Lenvatinib strongly inhibits pericyte viability by down-regulating MAPK, pAKT, and p-p70S6-kinase downstream PDGFR-β. Critically, lenvatinib significantly induces higher BRAFWT/V600E-PTC cell death when cocultured with pericytes, as a result of pericyte targeting via PDGFR-β. CONCLUSIONS This is the first thyroid-specific model of lenvatinib therapeutic efficacy against pericyte viability, which disadvantages BRAFWT/V600E-PTC growth. Assessing pericyte abundance in patients with PTC could be essential to selection rationales for appropriate targeted therapy with lenvatinib.
Collapse
Affiliation(s)
- Asumi Iesato
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, MA, USA
- Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, MA, USA
| | - Stephanie Li
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, MA, USA
- Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, MA, USA
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| | - Michele R Hacker
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, MA, USA
| | - Andrew H Fischer
- Department of Pathology, UMass Memorial Medical Center, Worcester, 01605, MA, USA
| | - Carmelo Nucera
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, MA, USA
- Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, 02142, MA, USA
| |
Collapse
|
46
|
Kang J, Park JH, Kong JS, Kim MJ, Lee SS, Park S, Myung JK. PINX1 promotes malignant transformation of thyroid cancer through the activation of the AKT/MAPK/β-catenin signaling pathway. Am J Cancer Res 2021; 11:5485-5495. [PMID: 34873474 PMCID: PMC8640828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 10/03/2021] [Indexed: 06/13/2023] Open
Abstract
Although thyroid cancer is the most prevalent endocrine malignancy, overall patients with thyroid cancer have a good long-term survival. However, a small percentage of patients with progressive thyroid cancer have poor outcomes, and the genetic drivers playing a key role thyroid cancer progression are mostly unknown. Here, we investigated the role of the PINX1 in thyroid cancer progression. Interestingly, PINX1 expression was significantly higher in ATC than in PTC in both patients and cell lines. When PINX1 was knockdown in ATC cells, cell proliferation rates, colony formation capacity, and cell cycle progression were significantly reduced. Furthermore, cell motility and the expression of EMT drivers were reduced by PINX1 downregulation. In contrast, the overexpression of PINX1 in PTC cells significantly increased those phenotypes of tumor progression, which demonstrates that PINX1 could promote tumor proliferation and malignant transformation in both PTC and ATC cells. To further understand whether PINX1 is also involved in the progression of PTC to ATC, we examined PI3K/AKT, MAPK, and β-catenin signaling activation after PINX1 modulation. Decreased PINX1 expression reduced the levels of p-AKT, p-ERK, p-p38, and β-catenin in ATC cells, but the increase of PINX1 expression upregulated the phosphorylation of AKT, ERK, and p38 and the levels of β-catenin in PTC cells. These results were all confirmed in xenograft mouse tumors. Our findings suggest that PINX1 regulates thyroid cancer progression by promoting cell proliferation, EMT, and signaling activation, and support the hypothesis that PINX1 could be a prognostic marker and a therapeutic target of thyroid cancer.
Collapse
Affiliation(s)
- JiHoon Kang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical SciencesSeoul, Republic of Korea
| | - Ji-Hye Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical SciencesSeoul, Republic of Korea
| | - Jun Suk Kong
- Department of Pathology, Korea Cancer Center HospitalSeoul, Republic of Korea
| | - Min Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical SciencesSeoul, Republic of Korea
| | - Seung-Sook Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical SciencesSeoul, Republic of Korea
- Department of Pathology, Korea Cancer Center HospitalSeoul, Republic of Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical SciencesSeoul, Republic of Korea
- Department of Pathology, Korea Cancer Center HospitalSeoul, Republic of Korea
| | - Jae Kyung Myung
- Department of Pathology, Korea Cancer Center HospitalSeoul, Republic of Korea
- Department of Pathology, College of Medicine, Hanyang UniversitySeoul, Republic of Korea
| |
Collapse
|
47
|
Lee WK, Cheng SY. Targeting transcriptional regulators for treatment of anaplastic thyroid cancer. JOURNAL OF CANCER METASTASIS AND TREATMENT 2021; 7. [PMID: 34761120 PMCID: PMC8577520 DOI: 10.20517/2394-4722.2021.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dysregulation of genes perpetuates cancer progression. During carcinogenesis, cancer cells acquire dependency of aberrant transcriptional programs (known as “transcription addiction”) to meet the high demands for uncontrolled proliferation. The needs for particular transcription programs for cancer growth could be cancer-type-selective. The dependencies of certain transcription regulators could be exploited for therapeutic benefits. Anaplastic thyroid cancer (ATC) is an extremely aggressive human cancer for which new treatment modalities are urgently needed. Its resistance to conventional treatments and the lack of therapeutic options for improving survival might have been attributed to extensive genetic heterogeneity due to subsequent evolving genetic alterations and clonal selections during carcinogenesis. Despite this genetic complexity, mounting evidence has revealed a characteristic transcriptional addiction of ATC cells resulting in evolving diverse oncogenic signaling for cancer cell survival. The transcriptional addiction has presented a huge challenge for effective targeting as shown by the failure of previous targeted therapies. However, an emerging notion is that many different oncogenic signaling pathways activated by multiple upstream driver mutations might ultimately converge on the transcriptional responses, which would provide an opportunity to target transcriptional regulators for treatment of ATC. Here, we review the current understanding of how genetic alterations in cancer distorted the transcription program, leading to acquisition of transcriptional addiction. We also highlight recent findings from studies aiming to exploit the opportunity for targeting transcription regulators as potential therapeutics for ATC.
Collapse
Affiliation(s)
- Woo Kyung Lee
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
48
|
Pani F, Yasuda Y, Di Dalmazi G, Chalan P, Gabrielson K, Adamo L, Sabini E, Mariotti S, Caturegli P. Pre-existing Thyroiditis Ameliorates Papillary Thyroid Cancer: Insights From a New Mouse Model. Endocrinology 2021; 162:6332851. [PMID: 34331442 PMCID: PMC8389179 DOI: 10.1210/endocr/bqab144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 01/27/2023]
Abstract
Papillary thyroid cancer (PTC) often co-occurs with Hashimoto's thyroiditis, an association that has long been reported in clinical studies yet remains controversial. Some studies, in fact, have suggested a protective effect of thyroiditis while others have not. We generated a mouse model where PTC and thyroiditis develop in a predictable manner, combining the oncogenic drive of the BRAFv600E mutation (inducible by tamoxifen) to the thyroiditis susceptibility of the NOD.H2h4 strain (inducible by iodine). A total of 113 NOD.H2h4_TPO-CRE-ER_BRAFV600E mice (50 followed throughout lifetime and 63 sacrificed at 16 weeks post tamoxifen) were used to determine whether the PTC phenotype differs when thyroiditis precedes or coincides with the onset of PTC. Mice with pre-existing thyroiditis lived longer (median survival of 28.2 weeks post tamoxifen) than those with concomitant (25.6 weeks) or no (24.5 weeks) thyroiditis (P < 0.01 by Laplace regression). PTC developed less frequently (33%) in the pre-existing thyroiditis group than the concomitant (100%) or no (100%) thyroiditis groups (P < 0.001 by chi-squared) and showed less aggressive histopathological features. The intratumoral mononuclear cell infiltration was more prominent in mice with pre-existing thyroiditis (P = 0.002 vs the other groups) and sustained by a significant expansion of effector memory CD8 + T cells and CD19 + B cells. These findings shed light on the controversial PTC-thyroiditis association and emphasize the contribution of intratumoral T and B lymphocytes to the evolution of PTC.
Collapse
Affiliation(s)
- Fabiana Pani
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Yoshinori Yasuda
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Giulia Di Dalmazi
- Division of Endocrinology, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Paulina Chalan
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, Pathology and Oncology and Environmental Health Engineering Johns Hopkins School of Medicine and Bloomberg School of Public Health, Baltimore, MD, USA
| | - Luigi Adamo
- Division of Cardiology, Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Elena Sabini
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stefano Mariotti
- Retired from Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Patrizio Caturegli
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Correspondence: Patrizio Caturegli, MD, MPH, Johns Hopkins Pathology, Ross Building, Room 656, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| |
Collapse
|
49
|
Lin A, Feng J, Chen X, Wang D, Wong M, Zhang G, Na J, Zhang T, Chen Z, Chen YT, Nancy Du YC. High levels of truncated RHAMM cooperate with dysfunctional p53 to accelerate the progression of pancreatic cancer. Cancer Lett 2021; 514:79-89. [PMID: 34044069 PMCID: PMC8235875 DOI: 10.1016/j.canlet.2021.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/19/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer has the lowest survival rate out of all types of cancer. Pancreatic cancer patients are often diagnosed at advanced stages, hence an urgent need for a better therapeutic development of this devastating disease. Receptor for hyaluronan-mediated motility (RHAMM), not expressed in adult normal pancreas, has been suggested as a prognostic factor and a potential therapeutic target for pancreatic ductal adenocarcinoma (PDAC) and pancreatic neuroendocrine tumor (PNET). In this study, we initially sought to determine whether genetic deletion of RHAMM would slow down pancreatic cancer progression using Rhamm-/- mice. However, we found that Rhamm-/- mice expressed a truncated HMMRΔexon8-16 protein at higher abundance levels than wild-type RHAMM. While HMMRΔexon8-16 did not enable malignant progression of pancreatic intraepithelial neoplasia in p48-Cre; LSL-KRASG12D mice, it accelerated the formation of invasive PDAC and shortened the survival of p48-Cre; LSL-KRASG12D mice with heterozygous p53 knockout. KrasG12D PDAC mice with homozygous p53 knockout mice died around 10 weeks, and the effect of HMMRΔexon8-16 was not apparent in these short lifespan mice. In addition, HMMRΔexon8-16 shortened the survival of PNET-bearing RIP-Tag mice, which had inactivated p53. In our analysis of TCGA dataset, pancreatic cancer patients with mutant TP53 or loss of one copy of TP53 had higher RHAMM expression, which, combined, predicted worse outcomes. Taken together, by collaborating with dysfunctional p53, high levels of HMMRΔexon8-16 , which lacks the centrosome targeting domain and degrons for interaction with the Anaphase-Promoting Complex (APC), accelerated pancreatic cancer progression.
Collapse
Affiliation(s)
- Anthony Lin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jennifer Feng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Xiang Chen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Dunrui Wang
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Megan Wong
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - George Zhang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Joseph Na
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Tiantian Zhang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhengming Chen
- Division of Biostatistics and Epidemiology, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Yao-Tseng Chen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Yi-Chieh Nancy Du
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
50
|
Luo H, Xia X, Kim GD, Liu Y, Xue Z, Zhang L, Shu Y, Yang T, Chen Y, Zhang S, Chen H, Zhang W, Li R, Tang H, Dong B, Fu X, Cheng W, Zhang W, Yang L, Peng Y, Dai L, Hu H, Jiang Y, Gong C, Hu Y, Zhu J, Li Z, Caulin C, Wei T, Park J, Xu H. Characterizing dedifferentiation of thyroid cancer by integrated analysis. SCIENCE ADVANCES 2021; 7:7/31/eabf3657. [PMID: 34321197 PMCID: PMC8318367 DOI: 10.1126/sciadv.abf3657] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/10/2021] [Indexed: 02/05/2023]
Abstract
Understanding of dedifferentiation, an indicator of poo prognosis for patients with thyroid cancer, has been hampered by imprecise and incomplete characterization of its heterogeneity and its attributes. Using single-cell RNA sequencing, we explored the landscape of thyroid cancer at single-cell resolution with 46,205 cells and delineated its dedifferentiation process and suppressive immune microenvironment. The developmental trajectory indicated that anaplastic thyroid cancer (ATC) cells were derived from a small subset of papillary thyroid cancer (PTC) cells. Moreover, a potential functional role of CREB3L1 on ATC development was revealed by integrated analyses of copy number alteration and transcriptional regulatory network. Multiple genes in differentiation-related pathways (e.g., EMT) were involved as the downstream targets of CREB3L1, increased expression of which can thus predict higher relapse risk of PTC. Collectively, our study provided insights into the heterogeneity and molecular evolution of thyroid cancer and highlighted the potential driver role of CREB3L1 in its dedifferentiation process.
Collapse
Affiliation(s)
- Han Luo
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuyang Xia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Gyeong Dae Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yang Liu
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhinan Xue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Lingyun Zhang
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China.,Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Yang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Shouyue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Haining Chen
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weihan Zhang
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruicen Li
- Health Promotion Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huairong Tang
- Health Promotion Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Birong Dong
- National Clinical Research Center for Geriatrics, Chengdu, Sichuan, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Wei Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Hongbo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yong Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yiguo Hu
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Jingqiang Zhu
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhihui Li
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Carlos Caulin
- Department of Otolaryngology-Head & Neck Surgery and University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Tao Wei
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.
| | - Heng Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China. .,Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|