1
|
Lv P, Wang Z, Si X, Su J, Yu Z, Yu H, Ji G, Song W. Biopolymer immune implants co-loaded with TMZ, R848 and IOX1 for perioperative therapy of glioblastoma. Acta Biomater 2024:S1742-7061(24)00581-6. [PMID: 39368721 DOI: 10.1016/j.actbio.2024.09.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma (GBM), a prevalent and aggressive brain tumor, poses significant treatment challenges due to its rapid progression and the difficulty in achieving complete surgical resection. The current treatment regime, primarily surgery followed by radiotherapy and chemotherapy, offers limited success, with a five-year survival rate of less than 10 %. For addressing the challenges faced in the treatment of GBM, an approach using a biopolymer implant constructed with dynamic reversible covalent bonds, was designed to achieve controlled and constant-rate release of chemotherapy drug (Temozolomide, TMZ), immune adjuvant (Resiquimod, R848) and checkpoint inhibitor (5-carboxy-8-hydroxyquinoline, IOX1). The safety evaluation demonstrated the biocompatibility of the implants, with no significant inflammatory response or adverse effects on various systemic organs. In vivo antitumor study showed that the local delivery of drug combination via this implant significantly inhibited tumor recurrence of orthotopic GBM. Immune analysis revealed that the combination of the three drugs effectively activated systemic antitumor immune responses and induced memory effects. The synergistic mechanism of the drug combination was further validated by RNA whole sequencing. The innovative approach of combining chemotherapy and immunotherapy in biopolymer immune implants for GBM treatment showed promising and opens new avenues for treating GBM, particularly in addressing postoperative recurrence. STATEMENT OF SIGNIFICANCE: Our research introduces a pioneering approach in treating orthotopic brain glioblastoma (GBM), characterized by inevitable tumor recurrence, poor immune infiltration and the restrictive nature of the blood-brain barrier. To break the impasse of ineffective treatment for GBM, the innovative use of dynamically reversible covalent bonds in polymer matrix ensures the controlled, stable and sustained release of drug combinations of the chemotherapeutic agent temozolomide, immune adjuvants and checkpoint inhibitors, which maintains the optimal concentration in the tumor, overcoming problems associated with conventional chemotherapy such as systemic toxicity and low tumor targeting. Empirical evidence from in vivo experiments on the rat GBM model demonstrates significant outcomes: 90 % tumor size reduction and prolonged survival with over 70 % tumor cure rate.
Collapse
Affiliation(s)
- Pinxin Lv
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhanfeng Wang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China
| | - Zhifei Yu
- The Second Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Hongquan Yu
- Department of Oncological Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Guofeng Ji
- Xuanwu Hospital, Capital Medical University, Beijing 100010, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| |
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
|
Lee M, Morris LGT. Genetic alterations in thyroid cancer mediating both resistance to BRAF inhibition and anaplastic transformation. Oncotarget 2024; 15:36-48. [PMID: 38275291 PMCID: PMC10812235 DOI: 10.18632/oncotarget.28544] [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: 10/31/2023] [Accepted: 12/08/2023] [Indexed: 01/27/2024] Open
Abstract
A subset of thyroid cancers present at advanced stage or with dedifferentiated histology and have limited response to standard therapy. Tumors harboring the BRAF V600E mutation may be treated with BRAF inhibitors; however, tumor response is often short lived due to multiple compensatory resistance mechanisms. One mode of resistance is the transition to an alternative cell state, which on rare occasions can correspond to tumor dedifferentiation. DNA sequencing and RNA expression profiling show that thyroid tumors that dedifferentiate after BRAF inhibition are enriched in known genetic alterations that mediate resistance to BRAF blockade, and may also drive tumor dedifferentiation, including mutations in the PI3K/AKT/MTOR (PIK3CA, MTOR), MAP/ERK (MET, NF2, NRAS, RASA1), SWI/SNF chromatin remodeling complex (ARID2, PBRM1), and JAK/STAT pathways (JAK1). Given these findings, recent investigations have evaluated the efficacy of dual-target therapies; however, continued lack of long-term tumor control illustrates the complex and multifactorial nature of these compensatory mechanisms. Transition to an immune-suppressed state is another correlate of BRAF inhibitor resistance and tumor dedifferentiation, suggesting a possible role for concurrent targeted therapy with immunotherapy. Investigations into combined targeted and immunotherapy are ongoing, but early results with checkpoint inhibitors, viral therapies, and CAR T-cells suggest enhanced anti-tumor immune activity with these combinations.
Collapse
Affiliation(s)
- Mark Lee
- Department of Otolaryngology-Head and Neck Surgery, New York Presbyterian Hospital, New York, NY 10032, USA
| | - Luc GT Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
4
|
Ying X, Ma X, Yang Z, Zhou B. Th9 Cytokines Inhibit Proliferation, Promote Apoptosis, and Immune Escape in Thyroid Carcinoma Cells. Appl Biochem Biotechnol 2024:10.1007/s12010-023-04821-2. [PMID: 38224392 DOI: 10.1007/s12010-023-04821-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/16/2024]
Abstract
To investigate the regulatory effects of T helper 9 (Th9) cytokines on the proliferation, apoptosis and immune escape of thyroid cancer cells. The survival rate of human thyroid cancer cell line TPC-1 after treatment with 0, 1, 2.5, 5, 10, 20 ng/ml IL-9 (or IL-21) was determined by CCK-8 method and suitable concentrations of IL-9 and IL-21 were screened out. The TPC-1 cells cultured in vitro were randomly grouped into control group, IL-9 group, IL-21 group and IL-9+IL-21 group. After treatment with IL-9 and IL-21 factors, the proliferation and apoptosis of TPC-1 cells in each group were detected by CCK-8 method and flow cytometry, respectively. The flow cytometry was applied to detect the proportion of Th9 and activated CD8+ T cells in human peripheral blood lymphocytes co-cultured with TPC-1 in each group. The expression of TPC-1 and IL-9R and IL-21R protein in each group and human peripheral blood lymphocytes. Compared with the control group, the cell viability PCNA and Bcl-2 protein expression in TPC-1 cells were lower in the IL-9 group, IL-21 group and IL-9+IL-21 group (P<0.05). The apoptosis rate, proportions of Th9 and activated CD8+ T cells, killing rate of human peripheral blood lymphocytes, the expression of Bax and caspase-3 proteins in TPC-1 cells, the expression of TPC-1 and human peripheral blood lymphocytes IL-9R and IL-21R proteins were all higher (P<0.05) in IL-9+IL-21 group compared with the IL-9 group and the IL-21 group. The cell viability, PCNA and Bcl-2 protein expression in TPC-1 cells in the IL-9+IL-21 group were all lower (P<0.05). Th9 cytokines can promote the differentiation of Th9 cells and CD8+ T cells, enhance their lethality, reduce the immune escape of thyroid cancer cells, and then inhibit their proliferation and promote their apoptosis.
Collapse
Affiliation(s)
- Xinyu Ying
- Department of Clinical Laboratory, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China
| | - Xinyi Ma
- Medical School, Ningbo University, Ningbo, China
| | - Ziru Yang
- Department of Radiotherapy, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China
| | - Bo Zhou
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, 315000, China.
| |
Collapse
|
5
|
Komatsuda H, Kono M, Wakisaka R, Sato R, Inoue T, Kumai T, Takahara M. Harnessing Immunity to Treat Advanced Thyroid Cancer. Vaccines (Basel) 2023; 12:45. [PMID: 38250858 PMCID: PMC10820966 DOI: 10.3390/vaccines12010045] [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: 11/20/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
The incidence of thyroid cancer (TC) has increased over the past 30 years. Although differentiated thyroid cancer (DTC) has a good prognosis in most patients undergoing total thyroidectomy followed by radioiodine therapy (RAI), 5-10% of patients develop metastasis. Anaplastic thyroid cancer (ATC) has a low survival rate and few effective treatments have been available to date. Recently, tyrosine kinase inhibitors (TKIs) have been successfully applied to RAI-resistant or non-responsive TC to suppress the disease. However, TC eventually develops resistance to TKIs. Immunotherapy is a promising treatment for TC, the majority of which is considered an immune-hot malignancy. Immune suppression by TC cells and immune-suppressing cells, including tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, is complex and dynamic. Negative immune checkpoints, cytokines, vascular endothelial growth factors (VEGF), and indoleamine 2,3-dioxygenase 1 (IDO1) suppress antitumor T cells. Basic and translational advances in immune checkpoint inhibitors (ICIs), molecule-targeted therapy, tumor-specific immunotherapy, and their combinations have enabled us to overcome immune suppression and activate antitumor immune cells. This review summarizes current findings regarding the immune microenvironment, immunosuppression, immunological targets, and immunotherapy for TC and highlights the potential efficacy of immunotherapy.
Collapse
Affiliation(s)
- Hiroki Komatsuda
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Michihisa Kono
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Risa Wakisaka
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Ryosuke Sato
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Takahiro Inoue
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
- Department of Innovative Head & Neck Cancer Research and Treatment, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Miki Takahara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
- Department of Innovative Head & Neck Cancer Research and Treatment, Asahikawa Medical University, Asahikawa 078-8510, Japan
| |
Collapse
|
6
|
Galligan A, Wallace R, Krishnamurthy B, Kay TWH, Sachithanandan N, Chiang C, Sandhu S, Hicks RJ, Iravani A. Increased Thyroidal Activity on Routine FDG-PET/CT after Combination Immune Checkpoint Inhibition: Temporal Associations with Clinical and Biochemical Thyroiditis. Cancers (Basel) 2023; 15:5803. [PMID: 38136348 PMCID: PMC10741830 DOI: 10.3390/cancers15245803] [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: 10/24/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND FDG-PET/CT used for immune checkpoint inhibitor (ICI) response assessment can incidentally identify immune-related adverse events (irAEs), including thyroiditis. This study aimed to correlate the time course of FDG-PET/CT evidence of thyroiditis with clinical and biochemical evolution of thyroid dysfunction. METHODS A retrospective review was performed by two independent blinded nuclear medicine physicians (NMPs) of thyroidal FDG uptake in 127 patients who underwent PET/CT between January 2016 and January 2019 at baseline and during treatment monitoring of combination ICI therapy for advanced melanoma. Interobserver agreement was assessed and FDG-PET/CT performance defined by a receiver-operating characteristic (ROC) curve using thyroid function tests (TFTs) as the standard of truth. Thyroid maximum standardized uptake value (SUVmax) and its temporal changes with respect to the longitudinal biochemistry were serially recorded. RESULTS At a median of 3 weeks after commencing ICI, 43/127 (34%) had a diagnosis of thyroiditis established by abnormal TFTs. FDG-PET/CT was performed at baseline and at a median of 11 weeks (range 3-32) following the start of therapy. ROC analysis showed an area under the curve of 0.87 (95% CI 0.80, 0.94) for FDG-PET/CT for detection of thyroiditis with a positive predictive value of 93%. Among patients with biochemical evidence of thyroiditis, those with a positive FDG-PET/CT were more likely to develop overt hypothyroidism (77% versus 35%, p < 0.01). In the evaluation of the index test, there was an almost perfect interobserver agreement between NMPs of 93.7% (95% CI 89.4-98.0), kappa 0.83. CONCLUSION Increased metabolic activity of the thyroid on routine FDG-PET/CT performed for tumoral response of patients undergoing ICI therapy is generally detected well after routine biochemical diagnosis. Elevation of FDG uptake in the thyroid is predictive of overt clinical hypothyroidism and suggests that an ongoing robust inflammatory response beyond the initial thyrotoxic phase may be indicative of thyroid destruction.
Collapse
Affiliation(s)
- Anna Galligan
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Roslyn Wallace
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (R.W.); (S.S.)
| | - Balasubramanian Krishnamurthy
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Thomas W. H. Kay
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Nirupa Sachithanandan
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Department of Internal Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
| | - Cherie Chiang
- Department of Internal Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Department of Medicine, Royal Melbourne Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Shahneen Sandhu
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (R.W.); (S.S.)
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Rodney J. Hicks
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Amir Iravani
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
7
|
Tao Y, Li P, Feng C, Cao Y. New Insights into Immune Cells and Immunotherapy for Thyroid Cancer. Immunol Invest 2023; 52:1039-1064. [PMID: 37846977 DOI: 10.1080/08820139.2023.2268656] [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] [Indexed: 10/18/2023]
Abstract
Thyroid cancer (TC) is the most common endocrine malignancy worldwide, and the incidence of TC has gradually increased in recent decades. Differentiated thyroid cancer (DTC) is the most common subtype and has a good prognosis. However, advanced DTC patients with recurrence, metastasis and iodine refractoriness, as well as more aggressive subtypes such as poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC), still pose a great challenge for clinical management. Therefore, it is necessary to continue to explore the inherent molecular heterogeneity of different TC subtypes and the global landscape of the tumor immune microenvironment (TIME) to find new potential therapeutic targets. Immunotherapy is a promising therapeutic strategy that can be used alone or in combination with drugs targeting tumor-driven genes. This article focuses on the genomic characteristics, tumor-associated immune cell infiltration and immune checkpoint expression of different subtypes of TC patients to provide guidance for immunotherapy.
Collapse
Affiliation(s)
- Yujia Tao
- School of Medical Laboratory, Weifang Medical University, Weifang, Shandong, China
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| | - Peng Li
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| | - Chao Feng
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| | - Yuan Cao
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| |
Collapse
|
8
|
Fagin JA, Krishnamoorthy GP, Landa I. Pathogenesis of cancers derived from thyroid follicular cells. Nat Rev Cancer 2023; 23:631-650. [PMID: 37438605 PMCID: PMC10763075 DOI: 10.1038/s41568-023-00598-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
The genomic simplicity of differentiated cancers derived from thyroid follicular cells offers unique insights into how oncogenic drivers impact tumour phenotype. Essentially, the main oncoproteins in thyroid cancer activate nodes in the receptor tyrosine kinase-RAS-BRAF pathway, which constitutively induces MAPK signalling to varying degrees consistent with their specific biochemical mechanisms of action. The magnitude of the flux through the MAPK signalling pathway determines key elements of thyroid cancer biology, including differentiation state, invasive properties and the cellular composition of the tumour microenvironment. Progression of disease results from genomic lesions that drive immortalization, disrupt chromatin accessibility and cause cell cycle checkpoint dysfunction, in conjunction with a tumour microenvironment characterized by progressive immunosuppression. This Review charts the genomic trajectories of these common endocrine tumours, while connecting them to the biological states that they confer.
Collapse
Affiliation(s)
- James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Gnana P Krishnamoorthy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iñigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
9
|
Luo Z, Xu J, Xu D, Xu J, Zhou R, Deng K, Chen Z, Zou F, Yao L, Hu Y. Mechanism of immune escape mediated by receptor tyrosine kinase KIT in thyroid cancer. Immun Inflamm Dis 2023; 11:e851. [PMID: 37506147 PMCID: PMC10336654 DOI: 10.1002/iid3.851] [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: 12/08/2021] [Accepted: 04/16/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVE Thyroid cancer (TC) is one of the fastest-growing malignant tumors. This study sought to explore the mechanism of immune escape mediated by receptor tyrosine kinase (KIT) in TC. METHODS The expression microarray of TC was acquired through the GEO database, and the difference analysis and Kyoto encyclopedia of genes and genomes pathway enrichment analysis were carried out. KIT levels in TC cell lines (K1/SW579/BCPAP) and human normal thyroid cells were detected using reverse transcription quantitative polymerase chain reaction and western blot analysis. TC cells were transfected with overexpressed (oe)-KIT and CD8+ T cells were cocultured with SW579 cells. Subsequently, cell proliferation, migration, and invasion abilities, CD8+ T cell proliferation, cytokine levels (interferon-γ [IFN-γ]/tumor necrosis factor-α [TNF-α]) were determined using colony formation assay, Transwell assays, flow cytometry, and enzyme-linked immunosorbent assay. The phosphorylation of MAPK pathway-related protein (ERK) was measured by western blot analysis. After transfection with oe-KIT, cells were treated with anisomycin (an activator of the MAPK pathway), and the protein levels of p-ERK/ERK and programmed death-ligand 1 (PD-L1) were detected. RESULTS Differentially expressed genes (N = 2472) were obtained from the GEO database. KIT was reduced in TC samples and lower in tumor cells than those in normal cells. Overexpression of KIT inhibited immune escape of TC cells. Specifically, the proliferation, migration, and invasion abilities of TC cells were lowered, the proliferation level of CD8+ T cells was elevated, and IFN-γ and TNF-α levels were increased. KIT inhibited the activation of the MAPK pathway in TC cells and downregulated PD-L1. CONCLUSION KIT suppressed immune escape of TC by blocking the activation of the MAPK pathway and downregulating PD-L1.
Collapse
Affiliation(s)
- Zhen Luo
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Jin Xu
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Dayong Xu
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Jiaojiao Xu
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Rongjun Zhou
- Department of SurgeryChangsha Hospital for Maternal and Child Health CareChangshaHunanChina
| | - Keping Deng
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Zheng Chen
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Fang Zou
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Libo Yao
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| | - Yuqin Hu
- Department of General SurgeryMinimally Invasive Surgery Center, The First Hospital of ChangshaChangshaHunanChina
| |
Collapse
|
10
|
Cai X, Wu Y, Wan Q, Zhang X. Minimal change disease associated with thyroid cancer: a case report. Front Med (Lausanne) 2023; 10:1132259. [PMID: 37234240 PMCID: PMC10205983 DOI: 10.3389/fmed.2023.1132259] [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: 12/27/2022] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
A patient complaining of edema of the face and lower extremities was admitted to the nephrology department for nephrotic syndrome. Renal biopsy revealed findings of minimal change disease (MCD). Thyroid ultrasound showed a hypoechoic 16 × 13 mm nodule in the right lobe, suspicious of malignancy. Later, total thyroidectomy confirmed the diagnosis of papillary thyroid carcinoma (PTC). After surgery, MCD remitted rapidly and completely, strongly suggesting the diagnosis of MCD secondary to PTC. We report here the first adult case of the paraneoplastic finding of MCD secondary to PTC. Additionally, we discuss the possible role of the BRAF gene in the pathophysiology of PTC-associated MCD in this case and highlight the importance of tumor screening.
Collapse
Affiliation(s)
- Xiaoyi Cai
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Medical College, Shantou University, Shantou, China
| | - Yuenv Wu
- École Doctorale Interdisciplinaire Sciences-Santé, Université Claude Bernard Lyon 1, Lyon, France
| | - Qijun Wan
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Xiuli Zhang
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
11
|
Hargadon KM. Genetic dysregulation of immunologic and oncogenic signaling pathways associated with tumor-intrinsic immune resistance: a molecular basis for combination targeted therapy-immunotherapy for cancer. Cell Mol Life Sci 2023; 80:40. [PMID: 36629955 PMCID: PMC11072992 DOI: 10.1007/s00018-023-04689-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023]
Abstract
Since the turn of the century, advances in targeted therapy and immunotherapy have revolutionized the treatment of cancer. Although these approaches have far outperformed traditional therapies in various clinical settings, both remain plagued by mechanisms of innate and acquired resistance that limit therapeutic efficacy in many patients. With a focus on tumor-intrinsic resistance to immunotherapy, this review highlights our current understanding of the immunologic and oncogenic pathways whose genetic dysregulation in cancer cells enables immune escape. Emphasis is placed on genomic, epigenomic, transcriptomic, and proteomic aberrations that influence the activity of these pathways in the context of immune resistance. Specifically, the role of pathways that govern interferon signaling, antigen processing and presentation, and immunologic cell death as determinants of tumor immune susceptibility are discussed. Likewise, mechanisms of tumor immune resistance mediated by dysregulated RAS-MAPK, WNT, PI3K-AKT-mTOR, and cell cycle pathways are described. Finally, this review highlights the ways in which recent insight into genetic dysregulation of these immunologic and oncogenic signaling pathways is informing the design of combination targeted therapy-immunotherapy regimens that aim to restore immune susceptibility of cancer cells by overcoming resistance mechanisms that often limit the success of monotherapies.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA.
| |
Collapse
|
12
|
Jiang C, He L, Xiao S, Wu W, Zhao Q, Liu F. E3 Ubiquitin Ligase RNF125 Suppresses Immune Escape in Head and Neck Squamous Cell Carcinoma by Regulating PD-L1 Expression. Mol Biotechnol 2022; 65:891-903. [DOI: 10.1007/s12033-022-00587-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022]
|
13
|
Zhang P, Guan H, Yuan S, Cheng H, Zheng J, Zhang Z, Liu Y, Yu Y, Meng Z, Zheng X, Zhao L. Targeting myeloid derived suppressor cells reverts immune suppression and sensitizes BRAF-mutant papillary thyroid cancer to MAPK inhibitors. Nat Commun 2022; 13:1588. [PMID: 35332119 PMCID: PMC8948260 DOI: 10.1038/s41467-022-29000-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 02/22/2022] [Indexed: 02/07/2023] Open
Abstract
MAPK signaling inhibitor (MAPKi) therapies show limited efficacy for advanced thyroid cancers despite constitutive activation of the signaling correlates with disease recurrence and persistence. Understanding how BRAF pathway stimulates tumorigenesis could lead to new therapeutic targets. Here, through genetic and pathological approaches, we demonstrate that BRAFV600E promotes thyroid cancer development by increasing myeloid-derived suppressor cells (MDSCs) penetrance. This BRAFV600E-induced immune suppression involves re-activation of the developmental factor TBX3, which in turn up-regulates CXCR2 ligands in a TLR2-NFκB dependent manner, leading to MDSCs recruitment into the tumor microenvironment. CXCR2 inhibition or MDSCs repression improves MAPKi therapy effect. Clinically, high TBX3 expression correlates with BRAFV600E mutation and increased CXCR2 ligands, along with abundant MDSCs infiltration. Thus, our study uncovers a BRAFV600E-TBX3-CXCLs-MDSCs axis that guides patient stratification and could be targeted to improve the efficacy of MAPKi therapy in advanced thyroid cancer patients. BRAF-V600E mutation is common in patients with papillary thyroid carcinoma (PTC) and has been associated with an aggressive phenotype. Here the authors show that the mutation supports cancer progression by reactivating the developmental factor TBX3 and promoting the recruitment of myeloid derived suppressive cells.
Collapse
Affiliation(s)
- Peitao Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Haixia Guan
- Department of Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong Province, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shukai Yuan
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Huili Cheng
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jian Zheng
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhenlei Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yifan Liu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yang Yu
- Department of Thyroid and Neck Oncology, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Oncology, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Li Zhao
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
14
|
Jin A, Zhou J, Yu P, Zhou S, Chang C. High Expression of THBS1 Leads to a Poor Prognosis in Papillary Thyroid Cancer and Suppresses the Anti-Tumor Immune Microenvironment. Technol Cancer Res Treat 2022; 21:15330338221085360. [PMID: 35315710 PMCID: PMC8943644 DOI: 10.1177/15330338221085360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objectives: To study the role of thrombospondin-1 (THBS1) in
papillary thyroid cancer (PTC) prognosis and the immune microenvironment.
Methods: A retrospective cohort study was designed, and data
from The Cancer Genome Atlas database and PTC tissues from Fudan University
Shanghai Cancer Center were used. Weighted gene co-expression network analysis
was performed to build a THBS1-immune-related gene prognostic
index (T-I index). Results: High THBS1 expression
was correlated with advanced TNM stage, higher recurrence risk, and shorter
progression-free interval. High THBS1 expression correlated
with MAPK and PD1 pathways indicating a tumor promoting and immunity-inhibiting
tendency. The T-I index showed a powerful capacity to predict progression-free
survival and immunotherapy benefit. Conclusion: High expression of
THBS1 leads to a poor prognosis in PTCs and suppresses the
anti-tumor immune microenvironment.
Collapse
Affiliation(s)
- Anqi Jin
- 89667Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jin Zhou
- 89667Fudan University Shanghai Cancer Center, Shanghai, China
| | - Pengcheng Yu
- 89667Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shichong Zhou
- 89667Fudan University Shanghai Cancer Center, Shanghai, China
| | - Cai Chang
- 89667Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
15
|
Huang Q, Lin Y, Chen C, Lou J, Ren T, Huang Y, Zhang H, Yu Y, Guo Y, Wang W, Wang B, Niu J, Xu J, Guo L, Guo W. Immune-Related LncRNAs Affect the Prognosis of Osteosarcoma, Which Are Related to the Tumor Immune Microenvironment. Front Cell Dev Biol 2021; 9:731311. [PMID: 34692688 PMCID: PMC8529014 DOI: 10.3389/fcell.2021.731311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/20/2021] [Indexed: 01/21/2023] Open
Abstract
Background: Abnormal expression of lncRNA is closely related to the occurrence and metastasis of osteosarcoma. The tumor immune microenvironment (TIM) is considered to be an important factor affecting the prognosis and treatment of osteosarcoma. This study aims to explore the effect of immune-related lncRNAs (IRLs) on the prognosis of osteosarcoma and its relationship with the TIM. Methods: Ninety-five osteosarcoma samples from the TARGET database were included. Iterative LASSO regression and multivariate Cox regression analysis were used to screen the IRLs signature with the optimal AUC. The predict function was used to calculate the risk score and divide osteosarcoma into a high-risk group and low-risk group based on the optimal cut-off value of the risk score. The lncRNAs in IRLs signature that affect metastasis were screened for in vitro validation. Single sample gene set enrichment analysis (ssGSEA) and ESTIMATE algorithms were used to evaluate the role of TIM in the influence of IRLs on osteosarcoma prognosis. Results: Ten IRLs constituted the IRLs signature, with an AUC of 0.96. The recurrence and metastasis rates of osteosarcoma in the high-risk group were higher than those in the low-risk group. In vitro experiments showed that knockdown of lncRNA (AC006033.2) could increase the proliferation, migration, and invasion of osteosarcoma. ssGSEA and ESTIMATE results showed that the immune cell content and immune score in the low-risk group were generally higher than those in the high-risk group. In addition, the expression levels of immune escape-related genes were higher in the high-risk group. Conclusion: The IRLs signature is a reliable biomarker for the prognosis of osteosarcoma, and they alter the prognosis of osteosarcoma. In addition, IRLs signature and patient prognosis may be related to TIM in osteosarcoma. The higher the content of immune cells in the TIM of osteosarcoma, the lower the risk score of patients and the better the prognosis. The higher the expression of immune escape-related genes, the lower the risk score of patients and the better the prognosis.
Collapse
Affiliation(s)
- Qingshan Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yilin Lin
- Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China
| | - Chenglong Chen
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Jingbing Lou
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Hongliang Zhang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yiyang Yu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yu Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Wei Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Boyang Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Jianfang Niu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Jiuhui Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Lei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| |
Collapse
|