1
|
Chernyak M, Hutchison D, Smith J. Papillary thyroid cancer and cutaneous melanoma: An often underrecognized pair. JAAD Case Rep 2024; 50:22-24. [PMID: 39036614 PMCID: PMC11259985 DOI: 10.1016/j.jdcr.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Affiliation(s)
- Michelle Chernyak
- Department of Dermatology, University of California, Irvine, Irvine, California
| | - Dana Hutchison
- Department of Dermatology, University of California, Irvine, Irvine, California
| | - Janellen Smith
- Department of Dermatology, University of California, Irvine, Irvine, California
| |
Collapse
|
2
|
Xu Q, Wang J, Mao Y, Xuan Z, Yang K, Tang X, Zhu X. Combined BRAF and PIM1 inhibitory therapy for papillary thyroid carcinoma based on BRAFV600E regulation of PIM1: Synergistic effect and metabolic mechanisms. Neoplasia 2024; 52:100996. [PMID: 38593698 PMCID: PMC11007432 DOI: 10.1016/j.neo.2024.100996] [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: 01/04/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
Papillary thyroid carcinoma (PTC) is the most common endocrine malignancy, and its incidence has increased rapidly in recent years. The BRAF inhibitor vemurafenib is effective against BRAFV600E-positive PTC; however, acquired resistance to single agent therapy frequently leads to tumor recurrence and metastasis, underscoring the need to develop tailored treatment strategies. We previously showed that the oncogenic kinase PIM1 was associated with the malignant phenotype and prognosis of PTC. In this study, we showed that sustained expression of the PIM1 protein in PTC was affected by the BRAFV600E mutation. Based on this regulatory mechanism, we tested the synergistic effects of inhibitors of BRAF (BRAFi) and PIM1 in BRAFV600E-positive PTC cell lines and xenograft tumors. LC-MS metabolomics analyses suggested that BRAFi/PIMi therapy acted by restricting the amounts of critical amino acids and nucleotides required by cancer cells as well as modulating DNA methylation. This study elucidates the role of BRAFV600E in the regulation of PIM1 in PTC and demonstrates the synergistic effect of a novel combination, BRAFi/PIMi, for the treatment of PTC. This discovery, along with the pathways that may be involved in the powerful efficacy of BRAFi/PIMi strategy from the perspective of cell metabolism, provides insight into the molecular basis of PTC progression and offers new perspectives for BRAF-resistant PTC treatment.
Collapse
Affiliation(s)
- Qianqian Xu
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, China
| | - Jiaqi Wang
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, China
| | - Yuting Mao
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, China
| | - Ziyang Xuan
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, China
| | - Ke Yang
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xi Tang
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xin Zhu
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, China.
| |
Collapse
|
3
|
Wang S, Cui Z, Zhu J, Zhou P, Cao X, Li X, Ma Y, He Q. Colchicine inhibits the proliferation and promotes the apoptosis of papillary thyroid carcinoma cells likely due to the inhibitory effect on HDAC1. Biochem Biophys Res Commun 2023; 679:129-138. [PMID: 37690423 DOI: 10.1016/j.bbrc.2023.09.006] [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: 06/20/2023] [Revised: 08/01/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Although the prognosis for papillary thyroid carcinoma (PTC) is generally good, a certain proportion of patients show recurrent or advanced disease, indicating the need for further development of targeted medications. The purpose of this study was to explore the interventional effects of colchicine on PTC and the potential mechanisms or targets. We obtained PTC-related targets from the database and colchicine targets by predicting them. We screened the common targets of colchicine and the PTC-related target histone deacetylase 1 (HDAC1) and verified through molecular docking that colchicine has a good affinity for HDAC1, i.e., colchicine may act on PTC by affecting HDAC1. We then used CCK-8, colony formation, mitochondrial membrane potential and apoptosis assays to confirm that colchicine could inhibit the proliferation and promote the apoptosis of PTC cells and verified by RT‒qPCR, Western blot, and cellular immunofluorescence assays that colchicine could inhibit the expression of HDAC1 in PTC cells. The cytotoxicity and inhibitory effect of colchicine on HDAC1 in PTC cells was stronger than that in normal thyroid cells. We then applied an HDAC1 inhibitor, pyroxamide, to verify that inhibition of HDAC1 inhibits proliferation and promotes apoptosis in PTC cells. Therefore, we conclude that colchicine can inhibit the proliferation and promote the apoptosis of PTC cells likely due to its inhibitory effect on HDAC1. This finding implies that colchicine may be helpful for therapeutic intervention in PTC and that HDAC1 may be a promising clinical therapeutic target.
Collapse
Affiliation(s)
- Shuai Wang
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Zhonghao Cui
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Jinan, 250000, China
| | - Jian Zhu
- Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China
| | - Peng Zhou
- Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China
| | - Xianjiao Cao
- Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China
| | - Xiaolei Li
- Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China
| | - Yunhan Ma
- Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China
| | - Qingqing He
- Department of Thyroid and Breast Surgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China.
| |
Collapse
|
4
|
Dang H, Sui M, He Q, Xie J, Liu Y, Hou P, Ji M. Pin1 inhibitor API-1 sensitizes BRAF-mutant thyroid cancers to BRAF inhibitors by attenuating HER3-mediated feedback activation of MAPK/ERK and PI3K/AKT pathways. Int J Biol Macromol 2023; 248:125867. [PMID: 37473892 DOI: 10.1016/j.ijbiomac.2023.125867] [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: 02/28/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/22/2023]
Abstract
BRAFV600E mutation is one of the most therapeutic targets in thyroid cancers. However, its specific inhibitors have shown little clinical benefit because they can reactivate the MAPK/ERK and PI3K/AKT pathways by feedback upregulating the transcription of HER3. Peptidyl-prolyl cis/trans isomerase Pin1 has been proven to be closely associated with tumor progression. Here, we aimed to determine antitumor activity of Pin1 inhibitor API-1 in thyroid cancer and its effect on cellular response to BRAF inhibitors. The results showed that API-1 exhibited strong antitumor activity against thyroid cancer. Meanwhile, it improved the response of BRAF-mutant thyroid cancer cells to BRAF inhibitor PLX4032 and there was a synergistic effect between them. Specially, a combination therapy of API-1 and PLX4032 significantly inhibited cell proliferation, colony formation, and the growth of xenograft tumors as well as induced cell apoptosis in BRAF-mutant thyroid cancer cells compared with API-1 or PLX4032 monotherapy. Similar results were also observed in transgenic mice with BrafV600E-driven thyroid cancer. Mechanistically, API-1 enhanced XPO5 ability to export pre-microRNA 20a (pre-miR-20a) from the nucleus to cytoplasm, thereby promoting the maturation of miR-20a-5p. Further studies showed that miR-20a-5p specifically targeted and down-regulated HER3, thereby blocking the reactivation of MAPK/ERK and PI3K/AKT signaling pathways caused by PLX4032. These results, taken together, demonstrate that Pin1 inhibitor API-1 significantly improves the sensitivity of BRAF-mutant thyroid cancer cells to PLX4032. Thus, this study not only determines the potential antitumor activity of Pin1 inhibitor API-1 in thyroid cancer but also offers an alternative therapeutic strategy for BRAF-mutant thyroid cancers by a combination of Pin1 inhibitor and BRAF kinase inhibitor.
Collapse
Affiliation(s)
- Hui Dang
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Mengjun Sui
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Qingyuan He
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Jingyi Xie
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Yan Liu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Peng Hou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Meiju Ji
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| |
Collapse
|
5
|
Zhang L, Li Z, Zhang M, Zou H, Bai Y, Liu Y, Lv J, Lv L, Liu P, Deng Z, Liu C. Advances in the molecular mechanism and targeted therapy of radioactive-iodine refractory differentiated thyroid cancer. Med Oncol 2023; 40:258. [PMID: 37524925 DOI: 10.1007/s12032-023-02098-3] [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: 04/12/2023] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
Most patients with differentiated thyroid cancer have a good prognosis after radioactive iodine-131 treatment, but there are still a small number of patients who are not sensitive to radioiodine treatment and may subsequently show disease progression. Therefore, radioactive-iodine refractory differentiated thyroid cancer treated with radioiodine usually shows reduced radioiodine uptake. Thus, when sodium iodine symporter expression, basolateral membrane localization and recycling degradation are abnormal, radioactive-iodine refractory differentiated thyroid cancer may occur. In recent years, with the deepening of research into the pathogenesis of this disease, an increasing number of molecules have become or are expected to become therapeutic targets. The application of corresponding inhibitors or combined treatment regimens for different molecular targets may be effective for patients with advanced radioactive-iodine refractory differentiated thyroid cancer. Currently, some targeted drugs that can improve the progression-free survival of patients with radioactive-iodine refractory differentiated thyroid cancer, such as sorafenib and lenvatinib, have been approved by the FDA for the treatment of radioactive-iodine refractory differentiated thyroid cancer. However, due to the adverse reactions and drug resistance caused by some targeted drugs, their application is limited. In response to targeted drug resistance and high rates of adverse reactions, research into new treatment combinations is being carried out; in addition to kinase inhibitor therapy, gene therapy and rutin-assisted iodine-131 therapy for radioactive-iodine refractory thyroid cancer have also made some progress. Thus, this article mainly focuses on sodium iodide symporter changes leading to the main molecular mechanisms in radioactive-iodine refractory differentiated thyroid cancer, some targeted drug resistance mechanisms and promising new treatments.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Zhi Li
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Meng Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Huangren Zou
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Yuke Bai
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Yanlin Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Juan Lv
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Ling Lv
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Pengjie Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Zhiyong Deng
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China.
| | - Chao Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| |
Collapse
|
6
|
Shi YB, Chen SY, Liu RB. The new insights into autophagy in thyroid cancer progression. J Transl Med 2023; 21:413. [PMID: 37355631 PMCID: PMC10290383 DOI: 10.1186/s12967-023-04265-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/09/2023] [Indexed: 06/26/2023] Open
Abstract
In recent decades, the incidence of thyroid cancer keeps growing at a shocking rate, which has aroused increasing concerns worldwide. Autophagy is a fundamental and ubiquitous biological event conserved in mammals including humans. Basically, autophagy is a catabolic process that cellular components including small molecules and damaged organelles are degraded for recycle to meet the energy needs, especially under the extreme conditions. The dysregulated autophagy has indicated to be involved in thyroid cancer progression. The enhancement of autophagy can lead to autophagic cell death during the degradation while the produced energies can be utilized by the rest of the cancerous tissue, thus this influence could be bidirectional, which plays either a tumor-suppressive or oncogenic role. Accordingly, autophagy can be suppressed by therapeutic agents and is thus regarded as a drug target for thyroid cancer treatments. In the present review, a brief description of autophagy and roles of autophagy in tumor context are given. We have addressed summary of the mechanisms and functions of autophagy in thyroid cancer. Some potential autophagy-targeted treatments are also summarized. The aim of the review is linking autophagy to thyroid cancer, so as to develop novel approaches to better control cancer progression.
Collapse
Affiliation(s)
- Yu-Bo Shi
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shu-Yuan Chen
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ren-Bin Liu
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
7
|
Jingtai Z, Linfei H, Yuyang Q, Ning K, Xinwei Y, Xin W, Xianhui R, Dongmei H, Weiwei Y, Xiangrui M, Tianze Z, Wei W, Xiangqian Z. Targeting Aurora-A inhibits tumor progression and sensitizes thyroid carcinoma to Sorafenib by decreasing PFKFB3-mediated glycolysis. Cell Death Dis 2023; 14:224. [PMID: 36990998 PMCID: PMC10060208 DOI: 10.1038/s41419-023-05709-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/30/2023]
Abstract
AbstractThyroid cancer (TC) is the most common endocrine tumor, amongst which anaplastic thyroid carcinoma (ATC) is the most deadly. Aurora-A usually functions as oncogenes, and its inhibitor Alisertib exerts a powerful antitumor effect in various tumors. However, the mechanism of Aurora-A in regulating TC cell energy supply remains unclear. In the present study, we demonstrated the antitumor effect of Alisertib and an association between high Aurora-A expression and shorter survival. Multi-omics data and in vitro validation data suggested that Aurora-A induced PFKFB3-mediated glycolysis to increase ATP supply, which significantly upregulated the phosphorylation of ERK and AKT. Furthermore, the combination of Alisertib and Sorafenib had a synergistic effect, further confirmed in xenograft models and in vitro. Collectively, our study provides compelling evidence of the prognostic value of Aurora-A expression and suggests that Aurora-A upregulates PFKFB3-mediated glycolysis to enhance ATP supply and promote TC progression. Combining Alisertib with Sorafenib has huge prospects for application in treating advanced thyroid carcinoma.
Collapse
|
8
|
Zeng Y, Ma W, Li L, Zhuang G, Luo G, Zhou H, Hao W, Liu Y, Guo F, Tian M, Ruan X, Gao M, Zheng X. Identification and validation of eight estrogen-related genes for predicting prognosis of papillary thyroid cancer. Aging (Albany NY) 2023; 15:1668-1684. [PMID: 36917092 PMCID: PMC10042678 DOI: 10.18632/aging.204582] [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: 10/17/2022] [Accepted: 02/06/2023] [Indexed: 03/16/2023]
Abstract
Papillary thyroid cancer (PTC) is one of the most common malignant tumors in female, and estrogen can affect its progression. However, the targets and mechanisms of estrogen action in PTC remain unclear. Therefore, this study focuses on the relationship between estrogen-related genes (ERGs) expression and prognosis in PTC, particularly neuropeptide U (NMU), and its important role in tumor progression. Based on The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, differentially expressed genes (DEGs) predominantly enriched in ERGs were identified between PTC and normal tissue. Then, we identified ERGs that contributed most to PTC prognosis, including Transducer of ERBB2 1 (TOB1), trefoil factor 1 (TFF1), phospholipase A and acyltransferase 3 (PLAAT3), NMU, kinesin family member 20A (KIF20A), DNA topoisomerase II alpha (TOP2A), tetraspanin 13 (TSPAN13), and carboxypeptidase E (CPE). In addition, we confirmed that NMU was highly expressed in PTC and explored the effect of NMU on PTC cells proliferation in vitro and in vivo. The results showed that the proliferative capacity of PTC cells was significantly reduced with NMU knockdown. Moreover, the phosphorylation levels of the Kirsten rat sarcoma virus (KRAS) signaling pathway were significantly lower with NMU knockdown. These results suggest that ERGs, especially NMU, may be novel prognostic indicators in PTC.
Collapse
Affiliation(s)
- Yu Zeng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Weike Ma
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lijuan Li
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Gaojian Zhuang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan 511500, China
| | - Guoqing Luo
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan 511500, China
| | - Hong Zhou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan 511500, China
| | - Weijing Hao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yu Liu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Fengli Guo
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Mengran Tian
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, Tianjin 300121, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xianhui Ruan
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ming Gao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, Tianjin 300121, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300121, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
9
|
Zhang Y, Sang R, Bao J, Jiang Z, Qian D, Zhou Y, Su W, Wei J, Zhao L, Wei Z, Zhao Y, Shi M, Chen G. Schwann cell-derived CXCL2 contributes to cancer pain by modulating macrophage infiltration in a mouse breast cancer model. Brain Behav Immun 2023; 109:308-320. [PMID: 36754246 DOI: 10.1016/j.bbi.2023.02.004] [Citation(s) in RCA: 3] [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] [Received: 09/25/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Pain is one of the most severe complications affecting the quality of life of cancer patients. Although substantial progress has been made in the diagnosis and treatment of cancer, the neurobiological mechanism of cancer pain is still unclear. In the present study, we identified the critical role of CXC chemokine 2 (CXCL2), released by Schwann cells after being activated by cancer cells, in maintaining cancer-induced macrophage infiltration and the resulting mechanical hypersensitivity and persistent spontaneous nociception. In vitro, Schwann cells cocultured with breast cancer cells exhibited a significant increase in CXCL2 expression; in addition, conditioned medium from Schwann cells activated by breast cancer cells had a similar effect to recombinant CXCL2 in terms of inducing macrophage migration. Targeting CXCL2 signaling by both CXC chemokine receptor 2 (CXCR2) antagonist pharmacological blockade and anti-CXCL2 mAb immunological blockade robustly prevented conditioned medium-induced macrophage migration. In vivo, both application of recombinant CXCL2 and perineural breast cancer cell implantation resulted in mechanical hypersensitivity and persistent spontaneous nociception in mice, along with increased macrophage infiltration into the sciatic nerves. Similar to the in vitro results, inhibition of CXCL2/CXCR2 signaling or conditional knockdown of CXCL2 in sciatic nerve Schwann cells effectively attenuated breast cancer cell-induced mechanical hypersensitivity, persistent spontaneous nociception, and macrophage recruitment in the sciatic nerve. Mechanistically, we found that redox effector factor-1 (Ref-1) secreted by breast cancer cells activated hypoxia inducible factor-1α (HIF-1α) expression and inhibited reactive oxygen species (ROS) production in Schwann cells, ultimately inducing CXCL2 expression in Schwann cells. In brief, the present study expands new insights into cancer pain mechanisms from promising animal models to provide new strategies for the control of cancer pain.
Collapse
Affiliation(s)
- Yonghui Zhang
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China; Department of Thoracic Surgery, Tumor Hospital Affiliated to Nantong University, Nantong 226001, Jiangsu Province, China
| | - Rui Sang
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Jingyin Bao
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Zhihao Jiang
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Danni Qian
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Yi Zhou
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wenfeng Su
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jinhuan Wei
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Long Zhao
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Zhongya Wei
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Yayu Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Minxin Shi
- Department of Thoracic Surgery, Tumor Hospital Affiliated to Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Gang Chen
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China; Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
10
|
Xie J, Liu J, Zhao M, Li X, Wang Y, Zhao Y, Cao H, Ji M, Chen M, Hou P. Disulfiram/Cu Kills and Sensitizes BRAF-Mutant Thyroid Cancer Cells to BRAF Kinase Inhibitor by ROS-Dependently Relieving Feedback Activation of MAPK/ERK and PI3K/AKT Pathways. Int J Mol Sci 2023; 24:ijms24043418. [PMID: 36834830 PMCID: PMC9968072 DOI: 10.3390/ijms24043418] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
BRAFV600E, the most common genetic alteration, has become a major therapeutic target in thyroid cancer. Vemurafenib (PLX4032), a specific inhibitor of BRAFV600E kinase, exhibits antitumor activity in patients with BRAFV600E-mutated thyroid cancer. However, the clinical benefit of PLX4032 is often limited by short-term response and acquired resistance via heterogeneous feedback mechanisms. Disulfiram (DSF), an alcohol-aversion drug, shows potent antitumor efficacy in a copper (Cu)-dependent way. However, its antitumor activity in thyroid cancer and its effect on cellular response to BRAF kinase inhibitors remain unclear. Antitumor effects of DSF/Cu on BRAFV600E-mutated thyroid cancer cells and its effect on the response of these cells to BRAF kinase inhibitor PLX4032 were systematically assessed by a series of in vitro and in vivo functional experiments. The molecular mechanism underlying the sensitizing effect of DSF/Cu on PLX4032 was explored by Western blot and flow cytometry assays. DSF/Cu exhibited stronger inhibitory effects on the proliferation and colony formation of BRAFV600E-mutated thyroid cancer cells than DSF treatment alone. Further studies revealed that DSF/Cu killed thyroid cancer cells by ROS-dependent suppression of MAPK/ERK and PI3K/AKT signaling pathways. Our data also showed that DSF/Cu strikingly increased the response of BRAFV600E-mutated thyroid cancer cells to PLX4032. Mechanistically, DSF/Cu sensitizes BRAF-mutant thyroid cancer cells to PLX4032 by inhibiting HER3 and AKT in an ROS-dependent way and subsequently relieving feedback activation of MAPK/ERK and PI3K/AKT pathways. This study not only implies potential clinical use of DSF/Cu in cancer therapy but also provides a new therapeutic strategy for BRAFV600E-mutated thyroid cancers.
Collapse
Affiliation(s)
- Jingyi Xie
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Juan Liu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Man Zhao
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Xinru Li
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yubo Wang
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yuelei Zhao
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Hongxin Cao
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Meiju Ji
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Mingwei Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Peng Hou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Correspondence:
| |
Collapse
|
11
|
Schubert L, Mariko ML, Clerc J, Huillard O, Groussin L. MAPK Pathway Inhibitors in Thyroid Cancer: Preclinical and Clinical Data. Cancers (Basel) 2023; 15:cancers15030710. [PMID: 36765665 PMCID: PMC9913385 DOI: 10.3390/cancers15030710] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Thyroid cancer is the most common endocrine cancer, with a good prognosis in most cases. However, some cancers of follicular origin are metastatic or recurrent and eventually become radioiodine refractory thyroid cancers (RAIR-TC). These more aggressive cancers are a clinical concern for which the therapeutic arsenal remains limited. Molecular biology of these tumors has highlighted a hyper-activation of the Mitogen-Activated Protein Kinases (MAPK) pathway (RAS-RAF-MEK-ERK), mostly secondary to the BRAFV600E hotspot mutation occurring in about 60% of papillary cancers and 45% of anaplastic cancers. Therapies targeting the different protagonists of this signaling pathway have been tested in preclinical and clinical models: first and second generation RAF inhibitors and MEK inhibitors. In clinical practice, dual therapies with a BRAF inhibitor and a MEK inhibitor are being recommended in anaplastic cancers with the BRAFV600E mutation. Concerning RAIR-TC, these inhibitors can be used as anti-proliferative drugs, but their efficacy is inconsistent due to primary or secondary resistance. A specific therapeutic approach in thyroid cancers consists of performing a short-term treatment with these MAPK pathway inhibitors to evaluate their capacity to redifferentiate a refractory tumor, with the aim of retreating the patients by radioactive iodine therapy in case of re-expression of the sodium-iodide symporter (NIS). In this work, we report data from recent preclinical and clinical studies on the efficacy of MAPK pathway inhibitors and their resistance mechanisms. We will also report the different preclinical and clinical studies that have investigated the redifferentiation with these therapies.
Collapse
Affiliation(s)
- Louis Schubert
- Department of Endocrinology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, 75014 Paris, France
| | - Mohamed Lamine Mariko
- Department of Endocrinology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, 75014 Paris, France
| | - Jérôme Clerc
- Department of Nuclear Medicine, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, 75014 Paris, France
| | - Olivier Huillard
- Institut du Cancer Paris CARPEM, Department of Medical Oncology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Lionel Groussin
- Department of Endocrinology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, 75014 Paris, France
- Correspondence:
| |
Collapse
|