1
|
Li S, Han H, Yang K, Li X, Ma L, Yang Z, Zhao YX. Emerging role of metabolic reprogramming in the immune microenvironment and immunotherapy of thyroid cancer. Int Immunopharmacol 2025; 144:113702. [PMID: 39602959 DOI: 10.1016/j.intimp.2024.113702] [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: 09/23/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The metabolic reprogramming of cancer cells is a hallmark of many malignancies. To meet the energy acquisition needs of tumor cells for rapid proliferation, tumor cells reprogram their nutrient metabolism, which is caused by the abnormal expression of transcription factors and signaling molecules related to energy metabolic pathways as well as the upregulation and downregulation of abnormal metabolic enzymes, receptors, and mediators. Thyroid cancer (TC) is the most common endocrine tumor, and immunotherapy has become the mainstream choice for clinical benefit after the failure of surgical, endocrine, and radioiodine therapies. TC change the tumor microenvironment (TME) through nutrient competition and metabolites, causing metabolic reprogramming of immune cells, profoundly changing immune cell function, and promoting immune evasion of tumor cells. A deeper understanding of how metabolic reprogramming alters the TME and controls immune cell fate and function will help improve the effectiveness of TC immunotherapy and patient outcomes. This paper aims to elucidate the metabolic communication that occurs between immune cells around TC and discusses how metabolic reprogramming in TC affects the immune microenvironment and the effectiveness of anti-cancer immunotherapy. Finally, targeting key metabolic checkpoints during metabolic reprogramming, combined with immunotherapy, is a promising strategy.
Collapse
Affiliation(s)
- Shouhua Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Hengtong Han
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Kaili Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Xiaoxiao Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Libin Ma
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Ze Yang
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Yong-Xun Zhao
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Gu X, Chen B, Zhang S, Zhai X, Hu Y, Ye H. The expression of CCL17 and potential prognostic value on tumor immunity in thyroid carcinoma based on bioinformatics analysis. Sci Rep 2024; 14:31580. [PMID: 39738081 PMCID: PMC11686015 DOI: 10.1038/s41598-024-75750-1] [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: 03/17/2024] [Accepted: 10/08/2024] [Indexed: 01/01/2025] Open
Abstract
Although CCL17 has been reported to exert a vital role in many cancers, the related studies in the thyroid carcinoma have never reported. As a chemokine, CCL17 plays a positive role by promoting the infiltration of immune cells into the tumor microenviroment (TME) to influence tumor invasion and metastasis. Therefore, this study is aimed to investigate the association of CCL17 level with potential prognostic value on tumor immunity in the thyroid carcinoma (THCA) based on the bioinformatics analysis. GEPIA database was applied to analyze CCL17 mRNA expression in THCA data from TCGA database. Through the collection of the data, totally 500 tumor and 57 normal tissue samples were taken for the study. According to survival status and survival time in 500 tumor samples and CCL17 expression from RNA-seq data, all patients were categorized as high- expression (n = 64) and low-expression (n = 436) groups using X-tile program. Next, the association of CCL17 with survival in the thyroid carcinoma patients was examined by using the Kaplan-Meier plotter database. Then, weighted gene co-expression network (WGCNA) was employed to analyze the 1424 DEGs to classify 9 modules. Besides, STRING database was used to obtain the hub genes. GO and KEGG database were employed to explore blue module genes enrichment situations. In addition, TISIDB was used to analyze the relationship of CCL17 expression with tumor-infiltrating lymphocytes proportion, immunostimulators, and major histocompatibility complexes in THCA. The correlation of CCL17 with 22 TIIC subtypes was evaluated by ESTIMATE and CIBERSORT databases. The association of CCL17 level with gene marker of immune cells in THCA was analyzed by GEPIA and TIMER databases. Finally, immunohistochemistry was applied to validate CCL17 expression in 21 tumor and para-carcinoma tissue samples. CCL17 expression in tumors was significantly up-regulated relative to non-carcinoma samples. Patients from CCL17 high-expression group had significantly decreased overall survival compared with low-expression group, which has a significantly importantly potential prognostic value. Moreover, CCL17 and clinical characteristics were analyzed, suggesting that CCL17 expression significantly increased among patients of advanced stage, with advanced T classification, advanced N classification, and higher CCR4 expression. Based on WGCNA, expression of 1424 DEGs in blue module with 258 genes was negatively related to dismal survival and clinical lymph node metastasis in THCA patients. Moreover, CCR4 and CCL17 genes were identified as hub genes within blue module. CCL17 high-expression had greater ImmuneScore, StromalScore and ESTIMATEScore, while lower TumorPurity compared to the CCL17 low-expression. Then, GO and KEGG database were used to analyze blue module genes enrichment situations. The result showed that genes in blue module were associated with cytokine-cytokine receptor interaction, chemokine, and PI3K - Akt pathways. The results of tumor-infiltrating lymphocytes proportion, immunostimulators, and major histocompatibility complexes were significantly positive in CCL17 high-expression. Our findings showed that B cells naïve, T cells CD4 memory resting, T cells CD8, T cells regulatory (Tregs), and dendritic cells resting were the main immune components of THCA tumor microenvironment (TME). CCL17 high-expression in TC was significantly positively related to expression of immune cell gene markers. The result of immunohistochemistry demonstrated that CCL17 expression in tumor tissues significantly increased compared with para-carcinoma tissues. CCL17 high-expression was significantly positively associated with age and advanced N classification, suggesting that CCL17 could accelerate tumor progression by promoting the lymph node metastasis. CCL17 high-expression in THCA tumor microenvironment (TME) accelerates local infiltration of immune cells and enhances anticancer immunity, resulting in worse survival of patients and exerting potential prognostic value on tumor immunity in THCA.
Collapse
Affiliation(s)
- Xue Gu
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Bingfeng Chen
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Shengcan Zhang
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xinyu Zhai
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Hu
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Hui Ye
- Department of Thyroid Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
3
|
Huo MH, Adeerjiang Y, Abulitipu A, Khan U, Li XX, Zhang L, Tian Y, Jiang S, Xu CC, Chao XZ, Yang YF, Zhang JX, Du GL. Th17/Treg cell balance in patients with papillary thyroid carcinoma: a new potential biomarker and therapeutic target. Front Oncol 2024; 14:1325575. [PMID: 39534095 PMCID: PMC11554530 DOI: 10.3389/fonc.2024.1325575] [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: 10/21/2023] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common subtype of thyroid carcinoma. The most effective treatment for PTC is surgical resection, and patients who undergo surgery have good survival outcomes, but some patients have distant metastasis or even multiorgan metastases at the time of initial diagnosis. Distant metastasis is associated with poorer prognosis and a higher mortality rate. Helper T lymphocyte 17 (Th17) cells and regulatory T lymphocytes (Tregs) play different roles in PTC, and the Th17/Treg balance is closely related to the progression of PTC. Th17 cells play anticancer roles, whereas Tregs play cancer-promoting roles. A Th17/Treg imbalance promotes tumor progression and accelerates invasive behaviors such as tumor metastasis. Th17/Treg homeostasis can be regulated by the TGF-β/IL-2 and IL-6 cytokine axes. Immune checkpoint inhibitors contribute to Treg/Th17 cell homeostasis. For PTC, monoclonal antibodies against CTLA-4, PD-1 and PD-L1 inhibit the activation of Tregs, reversing the Th17/Treg cell imbalance and providing a new option for the prevention and treatment of PTC. This article reviews the role of Tregs and Th17 cells in PTC and their potential targets, aiming to provide better treatment options for PTC.
Collapse
Affiliation(s)
- Meng-Han Huo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Gastroenterology and Endocrinology, Tianjin Haihe Hospital, Tianjin, China
| | - Yilinuer Adeerjiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ayiguzhali Abulitipu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Umair Khan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xin-Xi Li
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lei Zhang
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye Tian
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Sheng Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Can-Can Xu
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xian-Zhen Chao
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye-Fan Yang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jin-Xia Zhang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guo-Li Du
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Endocrinology, Bayingolin Mongolian Autonomous Prefecture People's Hospital, Kuerle, China
| |
Collapse
|
4
|
Zheng X, Sun R, Wei T. Immune microenvironment in papillary thyroid carcinoma: roles of immune cells and checkpoints in disease progression and therapeutic implications. Front Immunol 2024; 15:1438235. [PMID: 39290709 PMCID: PMC11405226 DOI: 10.3389/fimmu.2024.1438235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Papillary thyroid cancer (PTC) is the most common type of primary thyroid cancer. Despite the low malignancy and relatively good prognosis, some PTC cases are highly aggressive and even develop refractory cancer in the thyroid. Growing evidence suggested that microenvironment in tumor affected PTC biological behavior due to different immune states. Different interconnected components in the immune system influence and participate in tumor invasion, and are closely related to PTC metastasis. Immune cells and molecules are widely distributed in PTC tissues. Their quantity and proportion vary with the host's immune status, which suggests that immunotherapy may be a very promising therapeutic modality for PTC. In this paper, we review the role of immune cells and immune checkpoints in PTC immune microenvironment based on the characteristics of the PTC tumor microenvironment.
Collapse
Affiliation(s)
- Xun Zheng
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ruonan Sun
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Wei
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Oh KY, Hong SD, Yoon HJ. Tumor immune microenvironment in odontogenic carcinomas: Evaluation of the therapeutic potential of immune checkpoint blockade. J Oral Pathol Med 2024; 53:217-225. [PMID: 38449350 DOI: 10.1111/jop.13525] [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: 11/07/2023] [Revised: 01/21/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Despite recent advances in the use of immune checkpoint blockade (ICB) across various cancer types, its efficacy in odontogenic carcinomas remains unexplored. This study aims to investigate PD-L1 expression and the tumor immune microenvironment (TIME) in odontogenic carcinomas to determine the therapeutic potential of ICB and the significance of immune markers. METHODS The expressions of PD-L1 and T cell markers (CD3, CD8, and FOXP3) were visualized by immunohistochemistry in 21 tissue samples of odontogenic carcinomas. Tumoral PD-L1 expression and the density and spatial distribution of T cell subsets were evaluated, from which TIME was determined. The associations of the variables with clinicopathological and prognostic factors were statistically analyzed. RESULTS PD-L1 was positively expressed in 52.4% (11/21) of the cases studied. Among tumor types, ameloblastic carcinoma showed significantly higher PD-L1 expression (p = 0.016). TIME based on the intratumoral and stromal T cell distribution was immune-inflamed in 61.9% (13/21) and immune-excluded in 38.1% (8/21), with no immune-desert cases. PD-L1 expression was associated with the densities of all intratumoral T cell subsets (p = 0.03 for CD3, p = 0.03 for CD8, and p = 0.008 for FOXP3) but not with those of stromal T cells. High PD-L1 expression was associated with larger tumor size (p = 0.021), while the intratumoral CD8/CD3 ratio was inversely correlated with tumor size (p = 0.048). CONCLUSION These findings indicate the involvement of adaptive immune resistance in a subset of odontogenic carcinomas and support the therapeutic potential of ICB in patients with these rare malignancies.
Collapse
Affiliation(s)
- Kyu-Young Oh
- Department of Oral Pathology, College of Dentistry, Dankook University, Cheonan, Republic of Korea
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Seong-Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hye-Jung Yoon
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Zhang L, Yu S, Hong S, Xiao X, Liao Z, Li Y, Xiao H. Comprehensive analysis of BTNL9 as a prognostic biomarker correlated with immune infiltrations in thyroid cancer. BMC Med Genomics 2023; 16:234. [PMID: 37798795 PMCID: PMC10552425 DOI: 10.1186/s12920-023-01676-8] [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: 04/22/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Thyroid cancer (THCA) is the most common type of endocrine cancers, and the disease recurrences were usually associated with the risks of metastasis and fatality. Butyrophilin-like protein 9 (BTNL9) is a member of the immunoglobulin families. This study investigated the prognostic role of BTNL9 in THCA. METHODS Gene enhancers of BTNL9 were identified by interrogating H3K27ac ChIP-seq and RNA-seq data of papillary thyroid cancer (PTC) and benign thyroid nodule (BTN) tissues. Meanwhile, BTNL9 expression level was verified by qRT-PCR in 30 pairs of primary THCA and adjacent normal tissues. Clinicopathological and RNA sequencing data were obtained from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) to analyze the relations between BTNL9 expression and immune cell infiltration, chemokines/cytokines, immune checkpoint genes, clinical parameters and prognosis values. Besides, survival analysis combining BTNL9 expression and immune cell infiltration scores was conducted. Functional enrichment analysis was performed to investigate the potential biological mechanisms. Cox regression analyses were used to explore independent clinical indicators, and a nomogram model incorporating BTNL9 expression with clinical parameters was established. RESULTS BTNL9 showed significantly stronger H3K27ac modifications in BTN than PTC tissues at the promoter region (chr5: 181,035,673-181,047,436) and gene body (chr5: 181,051,544-181,054,849). The expression levels of BTNL9 were significantly down-regulated in THCA samples compared to normal tissues, and were strongly associated with different tumor stages, immune cell infiltrations, chemokines/cytokines and immune checkpoint genes in THCA. Functional enrichment analyses indicated that BTNL9 was involved in immune-related and cancer-related pathways. The Kaplan-Meier analysis showed lower BTNL9 expression was associated with poorer progression-free interval (PFI). BTNL9 expression and pathologic stages were independent prognostic indicators of PFI in THCA. CONCLUSIONS The results implied an important role of BTNL9 in the tumor progression, with the possibility of serving as a novel prognostic biomarker and a potential therapeutic target for THCA.
Collapse
Affiliation(s)
- Luyao Zhang
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang Yu
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shubin Hong
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xi Xiao
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhihong Liao
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yanbing Li
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Haipeng Xiao
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Song M, Liu Q, Sun W, Zhang H. Crosstalk between Thyroid Carcinoma and Tumor-Correlated Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2023; 15:2863. [PMID: 37345200 DOI: 10.3390/cancers15102863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/07/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023] Open
Abstract
Thyroid cancer (TC) is the most common malignancy in the endocrine system. Although most TC can achieve a desirable prognosis, some refractory thyroid carcinomas, including radioiodine-refractory differentiated thyroid cancer, as well as anaplastic thyroid carcinoma, face a myriad of difficulties in clinical treatment. These types of tumors contribute to the majority of TC deaths due to limited initial therapy, recurrence, and metastasis of the tumor and tumor resistance to current clinically targeted drugs, which ultimately lead to treatment failure. At present, a growing number of studies have demonstrated crosstalk between TC and tumor-associated immune cells, which affects tumor deterioration and metastasis through distinct signal transduction or receptor activation. Current immunotherapy focuses primarily on cutting off the interaction between tumor cells and immune cells. Since the advent of immunotherapy, scholars have discovered targets for TC immunotherapy, which also provides new strategies for TC treatment. This review methodically and intensively summarizes the current understanding and mechanism of the crosstalk between distinct types of TC and immune cells, as well as potential immunotherapy strategies and clinical research results in the area of the tumor immune microenvironment. We aim to explore the current research advances to formulate better individualized treatment strategies for TC patients and to provide clues and references for the study of potential immune checkpoints and the development of immunotherapy technologies.
Collapse
Affiliation(s)
- Mingyuan Song
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Shenyang 110001, China
| | - Qi Liu
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Shenyang 110001, China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Shenyang 110001, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Shenyang 110001, China
| |
Collapse
|
8
|
Shin E, Koo JS. Cell Component and Function of Tumor Microenvironment in Thyroid Cancer. Int J Mol Sci 2022; 23:ijms232012578. [PMID: 36293435 PMCID: PMC9604510 DOI: 10.3390/ijms232012578] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Thyroid cancer is the most common cancer in the endocrine system. Most thyroid cancers have good prognosis, but some of them are resistant to treatment or show aggressive behavior. Like other tumors, thyroid cancers harbor tumor microenvironment (TME) composed of cancer associated fibroblasts (CAF) and immune cells. Autoimmune lymphocytic thyroiditis can occur in the thyroid, and it may be associated with cancer development. TME is involved in tumor progression through various mechanisms: (1) CAF is involved in tumor progression through cell proliferation and extracellular matrix (ECM) remodeling; and (2) immune cells are involved in tumor progression through cell proliferation, angiogenesis, epithelial mesenchymal transformation (EMT), and immune suppression. These events are activated by various cytokines, chemokines, and metabolites secreted from cells that comprise TME. This review is focused on how CAF and immune cells, two important cell components of thyroid cancer TME, are involved in tumor progression, and will explore their potential as therapeutic targets.
Collapse
Affiliation(s)
| | - Ja Seung Koo
- Correspondence: ; Tel.: +82-2-2228-1772; Fax: +82-2-362-0860
| |
Collapse
|
9
|
Gong Z, Jia H, Xue L, Li D, Zeng X, Wei M, Liu Z, Tong MCF, Chen GG. The emerging role of transcription factor FOXP3 in thyroid cancer. Rev Endocr Metab Disord 2022; 23:421-429. [PMID: 34463908 DOI: 10.1007/s11154-021-09684-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 12/19/2022]
Abstract
Transcription factor FOXP3 is a crucial regulator in the development and function of regulatory T cells (Treg) that are essential for immunological tolerance and homeostasis. Numerous studies have indicated the correlation of tumor infiltrating FOXP3+ Treg upregulation with poor prognostic parameters in thyroid cancer, including lymph node metastases, extrathyroidal extension, and multifocality. Most immune-checkpoint molecules are expressed in Treg. The blockage of such signals with checkpoint inhibitors has been approved for several solid tumors, but not yet for thyroid cancer. Thyroid abnormalities may be induced by checkpoint inhibitors. For example, hypothyroidism, thyrotoxicosis, painless thyroiditis, or even thyroid storm are more frequently associated with anti-PD-1 antibodies (pembrolizumab and nivolumab). Therefore, Targeting FOXP3+ Treg may have impacts on checkpoint molecules and the growth of thyroid cancer. Several factors may impact the role and stability of FOXP3, such as alternative RNA splicing, mutations, and post-translational modification. In addition, the role of FOXP3+ Treg in the tumor microenvironment is also affected by the complex regulatory network formed by FOXP3 and its transcriptional partners. Here we discussed how the expression and function of FOXP3 were regulated and how FOXP3 interacted with its targets in Treg, aiming to help the development of FOXP3 as a potential therapeutic target for thyroid cancer.
Collapse
Affiliation(s)
- Zhongqin Gong
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Hao Jia
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Lingbin Xue
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Dongcai Li
- Shenzhen Key Laboratory of ENT, Institute of ENT & Longgang, ENT Hospital, Shenzhen, China
| | - Xianhai Zeng
- Shenzhen Key Laboratory of ENT, Institute of ENT & Longgang, ENT Hospital, Shenzhen, China
| | - Minghui Wei
- Department of Head & Neck Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, Guangdong, China
| | - Zhimin Liu
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Michael C F Tong
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
| | - George G Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
| |
Collapse
|
10
|
Bao L, Xu T, Lu X, Huang P, Pan Z, Ge M. Metabolic Reprogramming of Thyroid Cancer Cells and Crosstalk in Their Microenvironment. Front Oncol 2021; 11:773028. [PMID: 34926283 PMCID: PMC8674491 DOI: 10.3389/fonc.2021.773028] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/05/2021] [Indexed: 12/18/2022] Open
Abstract
Metabolism differs significantly between tumor and normal cells. Metabolic reprogramming in cancer cells and metabolic interplay in the tumor microenvironment (TME) are important for tumor formation and progression. Tumor cells show changes in both catabolism and anabolism. Altered aerobic glycolysis, known as the Warburg effect, is a well-recognized characteristic of tumor cell energy metabolism. Compared with normal cells, tumor cells consume more glucose and glutamine. The enhanced anabolism in tumor cells includes de novo lipid synthesis as well as protein and nucleic acid synthesis. Although these forms of energy supply are uneconomical, they are required for the functioning of cancer cells, including those in thyroid cancer (TC). Increasing attention has recently focused on alterations of the TME. Understanding the metabolic changes governing the intricate relationship between TC cells and the TME may provide novel ideas for the treatment of TC.
Collapse
Affiliation(s)
- Lisha Bao
- Second Clinical College, Zhejiang Chinese Medical School, Hangzhou, China
- ENT-Head & Neck Surgery Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Tong Xu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xixuan Lu
- ENT-Head & Neck Surgery Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ping Huang
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Zongfu Pan
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Minghua Ge
- ENT-Head & Neck Surgery Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
11
|
Ala M. The footprint of kynurenine pathway in every cancer: a new target for chemotherapy. Eur J Pharmacol 2021; 896:173921. [PMID: 33529725 DOI: 10.1016/j.ejphar.2021.173921] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/08/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
Treatment of cancers has always been a challenge for physicians. Typically, several groups of anti-cancer medications are needed for effective management of an invasive and metastatic cancer. Recently, therapeutic potentiation of immune system markedly improved treatment of cancers. Kynurenine pathway has an interwoven correlation with immune system. Kynurenine promotes T Reg (regulatory) differentiation, which leads to increased production of anti-inflammatory cytokines and suppression of cytotoxic activity of T cells. Overactivation of kynurenine pathway in cancers provides an immunologically susceptible microenvironment for mutant cells to survive and invade surrounding tissues. Interestingly, kynurenine pathway vigorously interacts with other molecular pathways involved in tumorigenesis. For instance, kynurenine pathway interacts with phospoinosisitide-3 kinase (PI3K), extracellular signal-regulated kinase (ERK), Wnt/β-catenin, P53, bridging integrator 1 (BIN-1), cyclooxygenase 2 (COX-2), cyclin-dependent kinase (CDK) and collagen type XII α1 chain (COL12A1). Overactivation of kynurenine pathway, particularly overactivation of indoleamine 2,3-dioxygenase (IDO) predicts poor prognosis of several cancers such as gastrointestinal cancers, gynecological cancers, hematologic malignancies, breast cancer, lung cancer, glioma, melanoma, prostate cancer and pancreatic cancer. Furthermore, kynurenine increases the invasion, metastasis and chemoresistance of cancer cells. Recently, IDO inhibitors entered clinical trials and successfully passed their safety tests and showed promising therapeutic efficacy for cancers such as melanoma, brain cancer, renal cell carcinoma, prostate cancer and pancreatic cancer. However, a phase III trial of epacadostat, an IDO inhibitor, could not increase the efficacy of treatment with pembrolizumab for melanoma. In this review the expanding knowledge towards kynurenine pathway and its application in each cancer is discussed separately.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
12
|
Xie Z, Li X, He Y, Wu S, Wang S, Sun J, He Y, Lun Y, Zhang J. Immune Cell Confrontation in the Papillary Thyroid Carcinoma Microenvironment. Front Endocrinol (Lausanne) 2020; 11:570604. [PMID: 33193087 PMCID: PMC7642595 DOI: 10.3389/fendo.2020.570604] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022] Open
Abstract
Background Papillary thyroid cancer has been associated with chronic inflammation. A systematic understanding of immune cell infiltration in PTC is essential for subsequent immune research and new diagnostic and therapeutic strategies. Methods Three different algorithms, single-sample gene set enrichment analysis (ssGSEA), immune cell marker and CIBERSORT, were used to evaluate immune cell infiltration levels (abundance and proportion) in 10 data sets (The Cancer Genome Atlas [TCGA], GSE3467, GSE3678, GSE5364, GSE27155, GSE33630, GSE50901, GSE53157, GSE58545, and GSE60542; a total of 799 PTC and 194 normal thyroid samples). Consensus unsupervised clustering divided PTC patients into low-immunity and high-immunity groups. Weighted gene coexpression network analysis (WGCNA) and gene set enrichment analysis (GSEA) were used to analyze the potential mechanisms causing differences in the immune response. Results Compared with normal tissues, PTC tissues had a higher overall immune level and higher abundance levels and proportions of M2 macrophages, Tregs, monocytes, neutrophils, dendritic cells (DCs), mast cells (MCs), and M0 macrophages. Compared with early PTC, advanced PTC showed higher immune infiltration and higher abundance levels and proportions of M2 macrophages, Tregs, monocytes, neutrophils, DCs, MCs, and M0 macrophages. Compared to the low-immunity group, the high-immunity group exhibited more advanced stages, larger tumor sizes, greater lymph node metastases, higher tall-cell PTCs, lower follicular PTC proportions, more BRAF mutations, and fewer RAS mutations. Epstein-Barr virus (EBV) infection was the most significantly enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway for key module genes. Conclusions In human PTC, M2 macrophages, Tregs, monocytes, neutrophils, DCs, MCs, and M0 macrophages appear to play a tumor-promoting role, while M1 macrophages, CD8+ T cells, B cells, NK cells, and T follicular helper (TFH) cells (including eosinophils, γδ T cells, and Th17 cells with weak supporting evidence) appear to play an antitumor role. During the occurrence and development of PTC, the overall immune level was increased, and the abundance and proportion of tumor-promoting immune cells were significantly increased, indicating that immune escape had been aggravated. Finally, we speculate that EBV may play an important role in changing the immune microenvironment of PTC tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jian Zhang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| |
Collapse
|
13
|
Yin H, Tang Y, Guo Y, Wen S. Immune Microenvironment of Thyroid Cancer. J Cancer 2020; 11:4884-4896. [PMID: 32626535 PMCID: PMC7330689 DOI: 10.7150/jca.44506] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Thyroid cancer (TC) is a highly heterogeneous endocrine malignancy with an increased incidence in women than in men. Previous studies regarding the pathogenesis of TC focused on the pathological changes of the tumor cells while ignoring the importance of the mesenchymal cells in tumor microenvironment. However, more recently, the stable environment provided by the interaction of thyroid cancer cells with the peri-tumoral stroma has been widely studied. Studies have shown that components of an individual's immune system are closely related to the occurrence, invasion, and metastasis of TC, which may affect response to treatment and prognosis of the patients. This article presents a comprehensive review of the immune cells, secreted soluble mediators and immune checkpoints in the immune microenvironment, mechanisms that promoting TC cells immune evasion and existing immunotherapy strategies. Besides it provides new strategies for TC prognosis prediction and immunotherapy.
Collapse
Affiliation(s)
- Hongyu Yin
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Yemei Tang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Yujia Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Shuxin Wen
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,General Hospital, Shenzhen University, Shenzhen 518061, Guangdong, P.R. China
| |
Collapse
|
14
|
Menicali E, Guzzetti M, Morelli S, Moretti S, Puxeddu E. Immune Landscape of Thyroid Cancers: New Insights. Front Endocrinol (Lausanne) 2020; 11:637826. [PMID: 33986723 PMCID: PMC8112200 DOI: 10.3389/fendo.2020.637826] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/30/2020] [Indexed: 01/23/2023] Open
Abstract
Immune system plays a key role in cancer prevention as well as in its initiation and progression. During multistep development of tumors, cells must acquire the capability to evade immune destruction. Both in vitro and in vivo studies showed that thyroid tumor cells can avoid immune response by promoting an immunosuppressive microenvironment. The recruitment of immunosuppressive cells such as TAMs (tumor-associated macrophages), TAMCs (tumor-associated mast cells), MDSC (myeloid-derived suppressor cells), TANs (tumor-associated neutrophils) and Tregs (regulatory T cells) and/or the expression of negative immune checkpoints, like PD-L1 (programmed death-ligand 1), CTLA-4 (cytotoxic T-lymphocyte associated protein 4), and/or immunosuppressive enzymes, as IDO1 (indoleamine 2,3-dioxygenase 1), are just some of the mechanisms that thyroid cancer cells exploit to escape immune destruction. Some authors systematically characterized immune cell populations and soluble mediators (chemokines, cytokines, and angiogenic factors) that constitute thyroid cancer microenvironment. Their purpose was to verify immune system involvement in cancer growth and progression, highlighting the differences in immune infiltrate among tumor histotypes. More recently, some authors have provided a more comprehensive view of the relationships between tumor and immune system involved in thyroid carcinogenesis. The Cancer Genome Atlas (TCGA) delivered a large amount of data that allowed to combine information on the inflammatory microenvironment with gene expression data, genetic and clinical-pathological characteristics, and differentiation degree of papillary thyroid carcinoma (PTC). Moreover, using a new sensitive and highly multiplex analysis, the NanoString Technology, it was possible to divide thyroid tumors in two main clusters based on expression of immune-related genes. Starting from these results, the authors performed an immune phenotype analysis that allowed to classify thyroid cancers in hot, cold, or intermediate depending on immune infiltration patterns of the tumor microenvironment. The aim of this review is to provide a comprehensive and updated view of the knowledge on immune landscape of thyroid tumors. Understanding interactions between tumor and microenvironment is crucial to effectively direct immunotherapeutic approaches in the treatment of thyroid cancer, particularly for those not responsive to conventional therapies.
Collapse
|
15
|
Hsu YC, Lee JJ, Chien MN, Chen MJ, Leung CH, Cheng SP. Is papillary thyroid microcarcinoma a biologically different disease? A propensity score-matched analysis. J Surg Oncol 2019; 120:1023-1030. [PMID: 31407354 DOI: 10.1002/jso.25670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Papillary thyroid microcarcinoma exhibits an indolent clinical course and could be a candidate for active surveillance in the appropriate setting. It remains unknown whether papillary microcarcinoma is biologically different from larger papillary carcinoma >1 cm. METHODS We analyzed clinicopathological information and transcriptome data of papillary thyroid cancer samples from The Cancer Genome Atlas. Propensity-score matching was used to construct a matched cohort consisting of 29 microcarcinomas and 58 carcinomas. Principal component analysis and unsupervised hierarchical cluster analysis were carried out to investigate the similarity of gene expression profiles. RESULTS After adjustment for differences in baseline clinicopathological and genetic factors, transcriptome could be grouped mainly on the basis of tumor class (BRAF-like vs RAS-like) and tumor size (microcarcinoma vs carcinoma). The gene set enrichment analysis showed that extracellular matrix-associated pathways were enriched in the MSigDB database. CONCLUSION Papillary thyroid microcarcinomas display a distinct gene expression pattern different from the corresponding carcinomas. We hypothesize that tumor microenvironment may play a role in the microcarcinoma/carcinoma phenotypic divergence.
Collapse
Affiliation(s)
- Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Jie-Jen Lee
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Ming-Nan Chien
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Ming-Jen Chen
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Hsiang Leung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
16
|
Ferrari SM, Fallahi P, Galdiero MR, Ruffilli I, Elia G, Ragusa F, Paparo SR, Patrizio A, Mazzi V, Varricchi G, Marone G, Antonelli A. Immune and Inflammatory Cells in Thyroid Cancer Microenvironment. Int J Mol Sci 2019; 20:E4413. [PMID: 31500315 PMCID: PMC6769504 DOI: 10.3390/ijms20184413] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 12/22/2022] Open
Abstract
A hallmark of cancer is the ability of tumor cells to avoid immune destruction. Activated immune cells in tumor microenvironment (TME) secrete proinflammatory cytokines and chemokines which foster the proliferation of tumor cells. Specific antigens expressed by cancer cells are recognized by the main actors of immune response that are involved in their elimination (immunosurveillance). By the recruitment of immunosuppressive cells, decreasing the tumor immunogenicity, or through other immunosuppressive mechanisms, tumors can impair the host immune cells within the TME and escape their surveillance. Within the TME, cells of the innate (e.g., macrophages, mast cells, neutrophils) and the adaptive (e.g., lymphocytes) immune responses are interconnected with epithelial cancer cells, fibroblasts, and endothelial cells via cytokines, chemokines, and adipocytokines. The molecular pattern of cytokines and chemokines has a key role and could explain the involvement of the immune system in tumor initiation and progression. Thyroid cancer-related inflammation is an important target for diagnostic procedures and novel therapeutic strategies. Anticancer immunotherapy, especially immune checkpoint inhibitors, unleashes the immune system and activates cytotoxic lymphocytes to kill cancer cells. A better knowledge of the molecular and immunological characteristics of TME will allow novel and more effective immunotherapeutic strategies in advanced thyroid cancer.
Collapse
Affiliation(s)
- Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| | - Poupak Fallahi
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80138 Naples, Italy; (M.R.G.); (G.V.); (G.M.)
- WAO Center of Excellence, 80138 Naples, Italy
| | - Ilaria Ruffilli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| | - Giusy Elia
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| | - Sabrina Rosaria Paparo
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| | - Armando Patrizio
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| | - Valeria Mazzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80138 Naples, Italy; (M.R.G.); (G.V.); (G.M.)
- WAO Center of Excellence, 80138 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80138 Naples, Italy; (M.R.G.); (G.V.); (G.M.)
- WAO Center of Excellence, 80138 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), National Research Council (CNR), 80131 Naples, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (S.M.F.); (I.R.); (G.E.); (F.R.); (S.R.P.); (A.P.); (V.M.)
| |
Collapse
|
17
|
Varricchi G, Loffredo S, Marone G, Modestino L, Fallahi P, Ferrari SM, de Paulis A, Antonelli A, Galdiero MR. The Immune Landscape of Thyroid Cancer in the Context of Immune Checkpoint Inhibition. Int J Mol Sci 2019; 20:E3934. [PMID: 31412566 PMCID: PMC6720642 DOI: 10.3390/ijms20163934] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/01/2019] [Accepted: 08/08/2019] [Indexed: 12/26/2022] Open
Abstract
Immune cells play critical roles in tumor prevention as well as initiation and progression. However, immune-resistant cancer cells can evade the immune system and proceed to form tumors. The normal microenvironment (immune cells, fibroblasts, blood and lymphatic vessels, and interstitial extracellular matrix (ECM)) maintains tissue homeostasis and prevents tumor initiation. Inflammatory mediators, reactive oxygen species, cytokines, and chemokines from an altered microenvironment promote tumor growth. During the last decade, thyroid cancer, the most frequent cancer of the endocrine system, has emerged as the fifth most incident cancer in the United States (USA), and its incidence is steadily growing. Inflammation has long been associated with thyroid cancer, raising critical questions about the role of immune cells in its pathogenesis. A plethora of immune cells and their mediators are present in the thyroid cancer ecosystem. Monoclonal antibodies (mAbs) targeting immune checkpoints, such as mAbs anti-cytotoxic T lymphocyte antigen 4 (anti-CTLA-4) and anti-programmed cell death protein-1/programmed cell death ligand-1 (anti-PD-1/PD-L1), have revolutionized the treatment of many malignancies, but they induce thyroid dysfunction in up to 10% of patients, presumably by enhancing autoimmunity. Combination strategies involving immune checkpoint inhibitors (ICIs) with tyrosine kinase (TK) or serine/threonine protein kinase B-raf (BRAF) inhibitors are showing considerable promise in the treatment of advanced thyroid cancer. This review illustrates how different immune cells contribute to thyroid cancer development and the rationale for the antitumor effects of ICIs in combination with BRAF/TK inhibitors.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80131 Naples, Italy
- WAO Center of Excellence, 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80131 Naples, Italy
- WAO Center of Excellence, 80131 Naples, Italy
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Luca Modestino
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80131 Naples, Italy
| | - Poupak Fallahi
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, 56126 Pisa, Italy
| | - Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, 56126 Pisa, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80131 Naples, Italy
- WAO Center of Excellence, 80131 Naples, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, 56126 Pisa, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80131 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80131 Naples, Italy.
- WAO Center of Excellence, 80131 Naples, Italy.
| |
Collapse
|
18
|
Liotti F, Prevete N, Vecchio G, Melillo RM. Recent advances in understanding immune phenotypes of thyroid carcinomas: prognostication and emerging therapies. F1000Res 2019; 8:F1000 Faculty Rev-227. [PMID: 30854191 PMCID: PMC6396838 DOI: 10.12688/f1000research.16677.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2019] [Indexed: 12/29/2022] Open
Abstract
Tumors modulate the host immune cells within their microenvironment to avoid recognition and elimination by our immune system, a phenotype called cancer immune escape. Different mechanisms responsible for cancer immune escape that result either in decreased tumor immunogenicity or in increased tumor immunosuppressive activity have been identified. Recently, various immunotherapeutic approaches have been developed with the aim to revert tumor immune escape. The aims of this review are to explore the immunological aspects of thyroid cancer and to assess whether these features can be exploited in the prognosis and treatment of advanced forms of this disease. Therefore, we will describe the immune landscape and phenotypes of thyroid cancer, summarize studies investigating the expression of immunomodulatory molecules, and finally describe the preclinical and clinical trials investigating the utility of immunotherapies in the management of thyroid cancer. The aim of this review is to explore the immunological aspects of thyroid cancer and to assess whether these features can be exploited in the prognosis and treatment of advanced forms of this disease. Therefore, we will describe the immune-landscape and phenotypes of thyroid cancer, we will summarize studies investigating the expression of immunomodulatory molecules, and we will finally describe the preclinical and clinical trials investigating the utility of immunotherapies in the management of thyroid cancer.
Collapse
Affiliation(s)
- Federica Liotti
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy, Italy
| | - Nella Prevete
- Dipartimento di Scienze Mediche Traslazionali, Università ¨Federico II¨ di Napoli, Naples, Italy, Italy
- Istituto di Endocrinologia e Oncologia Sperimentale “G. Salvatore”, CNR, Naples, Italy, Italy
| | - Giancarlo Vecchio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy, Italy
- Istituto Superiore di Oncologia, Naples, Italy
- Istituto Superiore di Oncologia, Genoa, Italy
| | - Rosa Marina Melillo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy, Italy
- Istituto di Endocrinologia e Oncologia Sperimentale “G. Salvatore”, CNR, Naples, Italy, Italy
| |
Collapse
|
19
|
Wei YF, Chu CY, Chang CC, Lin SH, Su WC, Tseng YL, Lin CC, Yen YT. Different pattern of PD-L1, IDO, and FOXP3 Tregs expression with survival in thymoma and thymic carcinoma. Lung Cancer 2018; 125:35-42. [PMID: 30429036 DOI: 10.1016/j.lungcan.2018.09.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVES The expression of immune checkpoint ligand PD-L1 has been reported in various tumors. The expression of IDO and FOXP3 Tregs are considered to be associated with tumor-induced tolerance and poor outcome. Their prognostic role in surgically treated thymoma and thymic carcinoma, however, has not been investigated. MATERIALS AND METHODS Tissue microarray (TMA) blocks comprised of 100 surgically treated thymomas and 69 surgically treated thymic carcinomas were conducted. Tissue sections were incubated with primary antibodies against PD-L1 (clone E1L3N, 1:100), IDO (clone 10.1, 1:50), and FOXP3 (clone 236 A/E7, 1:50). Comparisons for categorical variables were performed using χ2 test and Fisher's exact test. Survival analysis was established using Kaplan-Meier method and log-rank test. Univariate and multivariate analyses were performed using Cox regression model. RESULTS AND CONCLUSIONS High expression of PD-L1, IDO, and FOXP3 Tregs were identified in 36 (36%), 13 (13%), and 16 (16%) thymoma patients, respectively. High expression of PD-L1, IDO, and FOXP3 Tregs was associated with higher grade of tumor histology (P < 0.001, P = 0.007, and 0.014, respectively). High expression of PD-L1 was also associated with advanced Masaoka staging (P < 0.001). In patients with thymic carcinoma, high expression of PD-L1, IDO, and FOXP3 Tregs were identified in 25 (36%), 10 (14%), and 20 (29%) patients, respectively. Complete resection, low expression of IDO, and high expression of FOXP3 Tregs were associated with better overall survival (P = 0.001, 0.004, and 0.032, respectively), and progression-free survival (P < 0.001, P = 0.026, and 0.047, respectively) in multivariate analysis. In surgically treated thymoma, high PD-L1 expression was associated with advanced Masaoka staging. High PD-L1, IDO, and FOXP3 Tregs expression was associated with high grade histology. In surgically treated thymic carcinoma, significant survival benefit was noted in patients with complete resection, low IDO expression, and high FOXP3 Tregs expression.
Collapse
Affiliation(s)
- Yu-Feng Wei
- Division of Chest Medicine, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Chang-Yao Chu
- Division of Surgical Pathology, Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Chun Chang
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Hsiang Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Chou Su
- Division of Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Lin Tseng
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chung Lin
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ting Yen
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Trauma and Acute Care Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
20
|
Teoh ST, Lunt SY. Metabolism in cancer metastasis: bioenergetics, biosynthesis, and beyond. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2017; 10. [DOI: 10.1002/wsbm.1406] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/10/2017] [Accepted: 08/28/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Shao Thing Teoh
- Department of Biochemistry and Molecular Biology; Department of Chemical Engineering and Materials Science, Michigan State University; East Lansing MI USA
| | - Sophia Y. Lunt
- Department of Biochemistry and Molecular Biology; Department of Chemical Engineering and Materials Science, Michigan State University; East Lansing MI USA
| |
Collapse
|
21
|
Sharma R, Di Dalmazi G, Caturegli P. Exacerbation of Autoimmune Thyroiditis by CTLA-4 Blockade: A Role for IFNγ-Induced Indoleamine 2, 3-Dioxygenase. Thyroid 2016; 26:1117-24. [PMID: 27296629 PMCID: PMC4976247 DOI: 10.1089/thy.2016.0092] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cytotoxic T-lymphocyte associated protein 4 (CTLA-4) is a negative regulator of immune responses that suppresses the activity of effector T cells and contributes to the maintenance of self tolerance. When blocked therapeutically, CTLA-4 leads to an overall activation of T cells that has been exploited for cancer control, a control associated however with a variety of immune-related side effects such as autoimmune thyroiditis. To investigate the mechanism(s) underlying this form of thyroiditis, we used the NOD-H2(h4) mouse, a model that develops thyroiditis at very high incidence after addition of iodine to the drinking water. METHODS NOD-H2(h4) mice were started on drinking water supplemented with 0.05% sodium iodide when 8 weeks old and then injected with a hamster monoclonal antibody against mouse CTLA-4, polyclonal hamster immunoglobulins, or phosphate buffered saline when 11 weeks old. One month later (15 weeks of age), mice were sacrificed to assess thyroiditis, general immune responses in blood and spleen, and expression of indoleamine 2, 3-dioxygenase (IDO) in the thyroid and in isolated antigen-presenting cells after stimulation with interferon gamma. The study also analyzed IDO expression in four autopsy cases of metastatic melanoma who had received treatment with a CTLA-4 blocking antibody, and six surgical pathology Hashimoto thyroiditis controls. RESULTS CTLA-4 blockade worsened autoimmune thyroiditis, as assessed by a greater incidence, a more aggressive mononuclear cell infiltration in thyroids, and higher thyroglobulin antibody levels when compared to the control groups. CTLA-4 blockade also expanded the proportion of splenic CD4+ effector T cells, as well as the production of interleukin (IL)-2, interferon gamma, IL-10, and IL-13 cytokines. Interestingly, CTLA-4 blockade induced a strong expression of IDO in mouse and human thyroid glands, an expression that could represent a counter-regulatory mechanism to protect against the inflammatory environment. CONCLUSIONS This study shows that CTLA-4 blockade exacerbates the iodine-accelerated form of thyroiditis typical of the NOD-H2(h4) mouse. The study could also have implications for cancer patients who develop thyroiditis as an immune-related adverse event after CTLA-4 blockade.
Collapse
Affiliation(s)
- Rajni Sharma
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Giulia Di Dalmazi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Medicine, G. d'Annunzio University of Chieti, Cheti, Italy
| | - Patrizio Caturegli
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
22
|
Galdiero MR, Varricchi G, Marone G. The immune network in thyroid cancer. Oncoimmunology 2016; 5:e1168556. [PMID: 27471646 PMCID: PMC4938375 DOI: 10.1080/2162402x.2016.1168556] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/14/2016] [Accepted: 03/14/2016] [Indexed: 01/17/2023] Open
Abstract
The immune system plays critical roles in tumor prevention, but also in its initiation and progression. Tumors are subjected to immunosurveillance, but cancer cells generate an immunosuppressive microenvironment that favors their escape from immune-mediated elimination. During chronic inflammation, immune cells can contribute to the formation and progression of tumors by producing mitogenic, prosurvival, proangiogenic and lymphangiogenic factors. Thyroid cancer is the most frequent type of endocrine neoplasia and is the most rapidly increasing cancer in the US. In this review, we discuss recent findings on how different immune cells and mediators can contribute to thyroid cancer development and progression.
Collapse
Affiliation(s)
- Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DiSMeT), School of Medicine, University of Naples Federico II, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT), School of Medicine, University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT), School of Medicine, University of Naples Federico II, Naples, Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
23
|
Liu JZ, Zhou LY. Elevated amino acids in gastric juice as potential biomarkers for gastric cancer. Shijie Huaren Xiaohua Zazhi 2016; 24:1984-1992. [DOI: 10.11569/wcjd.v24.i13.1984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Early diagnosis plays a vital role in the improvement of the curative rate of gastric cancer. However, the poor specificity and sensitivity of traditional gastric cancer biomarkers, such as carcino-embryonic antigen and carbohydrate antigen 199, make it difficult to screen early gastric cancer. Gastric juice amino acid profile may be an alternative tumor marker, since abnormal elevation of amino acids in gastric juice has been reported in precancerous patients. Normal concentration of amino acids is extremely low in gastric juice, and special techniques are required to detect amino acids in gastric juice. The change of amino acids in gastric juice is more significant than in blood and tumor tissue. The amino acid profile in gastric juice may work as efficient biomarkers for gastric cancer with the development of amino acid detection technique.
Collapse
|
24
|
Porporato PE, Payen VL, Baselet B, Sonveaux P. Metabolic changes associated with tumor metastasis, part 2: Mitochondria, lipid and amino acid metabolism. Cell Mol Life Sci 2016; 73:1349-63. [PMID: 26646069 PMCID: PMC11108268 DOI: 10.1007/s00018-015-2100-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/16/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022]
Abstract
Metabolic alterations are a hallmark of cancer controlling tumor progression and metastasis. Among the various metabolic phenotypes encountered in tumors, this review focuses on the contributions of mitochondria, lipid and amino acid metabolism to the metastatic process. Tumor cells require functional mitochondria to grow, proliferate and metastasize, but shifts in mitochondrial activities confer pro-metastatic traits encompassing increased production of mitochondrial reactive oxygen species (mtROS), enhanced resistance to apoptosis and the increased or de novo production of metabolic intermediates of the TCA cycle behaving as oncometabolites, including succinate, fumarate, and D-2-hydroxyglutarate that control energy production, biosynthesis and the redox state. Lipid metabolism and the metabolism of amino acids, such as glutamine, glutamate and proline are also currently emerging as focal control points of cancer metastasis.
Collapse
Affiliation(s)
- Paolo E Porporato
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium
| | - Valéry L Payen
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium
| | - Bjorn Baselet
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, 2400 Mol, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium.
| |
Collapse
|