51
|
Ng TSC, Gunda V, Li R, Prytyskach M, Iwamoto Y, Kohler RH, Parangi S, Weissleder R, Miller MA. Detecting Immune Response to Therapies Targeting PDL1 and BRAF by Using Ferumoxytol MRI and Macrin in Anaplastic Thyroid Cancer. Radiology 2020; 298:123-132. [PMID: 33107799 DOI: 10.1148/radiol.2020201791] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Anaplastic thyroid cancer (ATC) is aggressive with a poor prognosis, partly because of the immunosuppressive microenvironment created by tumor-associated macrophages (TAMs). Purpose To understand the relationship between TAM infiltration, tumor vascularization, and corresponding drug delivery by using ferumoxytol-enhanced MRI and macrin in an ATC mouse model. Materials and Methods ATC tumors were generated in 6-8-week-old female B6129SF1/J mice through intrathyroid injection to model orthotopic tumors, or intravenously to model hematogenous metastasis, and prospectively enrolled randomly into treatment cohorts (n = 94 total; August 1, 2018, to January 15, 2020). Mice were treated with vehicle or combined serine/threonine-protein kinase B-Raf (BRAF) kinase inhibitor (BRAFi) and anti-PDL1 antibody (aPDL1). A subset was cotreated with therapies, including an approximately 70-nm model drug delivery nanoparticle (DDNP) to target TAM, and an antibody-neutralizing colony stimulating factor 1 receptor (CSF1R). Imaging was performed at the macroscopic level with ferumoxytol-MRI and microscopically with macrin. Genetically engineered BrafV600E/WT p53-null allografts were used and complemented by a GFP-transgenic derivative and human xenografts. Tumor-bearing organs were processed by using tissue clearing and imaged with confocal microscopy and MRI. Two-tailed Wilcoxon tests were used for comparison (≥five per group). Results TAM levels were higher in orthotopic thyroid tumors compared with pulmonary metastatic lesions by 79% ± 23 (standard deviation; P < .001). These findings were concordant with ferumoxytol MRI, which showed 136% ± 88 higher uptake in thyroid lesions (P = .02) compared with lung lesions. BRAFi and aPDL1 combination therapy resulted in higher tumor DDNP delivery by 39% ± 14 in pulmonary lesions (P = .004). Compared with the untreated group, tumors following BRAFi, aPDL1, and CSF1R-blocking antibody combination therapy did not show greater levels of TAM or DDNP (P = .82). Conclusion In a mouse model of anaplastic thyroid cancer, ferumoxytol MRI showed 136% ± 88 greater uptake in orthotopic thyroid tumors compared with pulmonary lesions, which reflected high vascularization and greater tumor-associated macrophage (TAM) levels. Serine/threonine-protein kinase B-Raf inhibitor and anti-programmed death ligand 1 antibody elicited higher local TAM levels and 43% ± 20 greater therapeutic nanoparticle delivery but not higher vascularization in pulmonary tumors. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Luker in this issue.
Collapse
Affiliation(s)
- Thomas S C Ng
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Viswanath Gunda
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Ran Li
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Mark Prytyskach
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Yoshiko Iwamoto
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Rainer H Kohler
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Sareh Parangi
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Ralph Weissleder
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| | - Miles A Miller
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, 185 Cambridge St, Suite 5.210, Boston, MA 02114 (T.S.C.N., R.L., M.P., Y.I., R.H.K., R.W., M.A.M.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (T.S.C.N.); Departments of Surgery (V.G., S.P.) and Radiology (R.L., R.W., M.A.M.), Massachusetts General Hospital and Harvard Medical School, Boston, Mass; and Department of Systems Biology, Harvard Medical School, Boston, Mass (R.W.)
| |
Collapse
|
52
|
Targeting Tumor-Associated Macrophages in Anti-Cancer Therapies: Convincing the Traitors to Do the Right Thing. J Clin Med 2020; 9:jcm9103226. [PMID: 33050070 PMCID: PMC7600332 DOI: 10.3390/jcm9103226] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
In the last decade, it has been well-established that tumor-infiltrating myeloid cells fuel not only the process of carcinogenesis through cancer-related inflammation mechanisms, but also tumor progression, invasion, and metastasis. In particular, tumor-associated macrophages (TAMs) are the most abundant leucocyte subset in many cancers and play a major role in the creation of a protective niche for tumor cells. Their ability to generate an immune-suppressive environment is crucial to escape the immune system and to allow the tumor to proliferate and metastasize to distant sites. Conventional therapies, including chemotherapy and radiotherapy, are often not able to limit cancer growth due to the presence of pro-tumoral TAMs; these are also responsible for the failure of novel immunotherapies based on immune-checkpoint inhibition. Several novel therapeutic strategies have been implemented to deplete TAMs; however, more recent approaches aim to use TAMs themselves as weapons to fight cancer. Exploiting their functional plasticity, the reprogramming of TAMs aims to convert immunosuppressive and pro-tumoral macrophages into immunostimulatory and anti-tumor cytotoxic effector cells. This shift eventually leads to the reconstitution of a reactive immune landscape able to destroy the tumor. In this review, we summarize the current knowledge on strategies able to reprogram TAMs with single as well as combination therapies.
Collapse
|
53
|
Nasrollahzadeh E, Razi S, Keshavarz-Fathi M, Mazzone M, Rezaei N. Pro-tumorigenic functions of macrophages at the primary, invasive and metastatic tumor site. Cancer Immunol Immunother 2020; 69:1673-1697. [PMID: 32500231 DOI: 10.1007/s00262-020-02616-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) not only facilitates cancer progression from the early formation to distant metastasis, but also it differs itself from time to time alongside the tumor evolution. Tumor-associated macrophages (TAMs), whether as pre-existing tissue-resident macrophages or recruited monocytes, are an inseparable part of this microenvironment. As their parents are broadly classified into a dichotomic, simplistic M1 and M2 subtypes, TAMs also exert paradoxical and diverse phenotypes as they are settled in different regions of TME and receive different microenvironmental signals. Briefly, M1 macrophages induce an inflammatory precancerous niche and flame the early oncogenic mutations, whereas their M2 counterparts are reprogrammed to release various growth factors and providing an immunosuppressive state in TME as long as abetting hypoxic cancer cells to set up a new vasculature. Further, they mediate stromal micro-invasion and co-migrate with invasive cancer cells to invade the vascular wall and neural sheath, while another subtype of TAMs prepares suitable niches much earlier than metastatic cells arrive at the target tissues. Accordingly, at the neoplastic transformation, during the benign-to-malignant transition and through the metastatic cascade, macrophages are involved in shaping the primary, micro-invasive and pre-metastatic TMEs. Whether their behavioral plasticity is derived from distinct genotypes or is fueled by microenvironmental cues, it could define these cells as remarkably interesting therapeutic targets.
Collapse
Affiliation(s)
- Elaheh Nasrollahzadeh
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer Biology, VIB, KU Leuven, Louvain, B3000, Belgium
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
54
|
Lin B, Lu B, Hsieh IY, Liang Z, Sun Z, Yi Y, Lv W, Zhao W, Li J. Synergy of GSK-J4 With Doxorubicin in KRAS-Mutant Anaplastic Thyroid Cancer. Front Pharmacol 2020; 11:632. [PMID: 32477122 PMCID: PMC7239034 DOI: 10.3389/fphar.2020.00632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Anaplastic thyroid cancer is the most aggressive thyroid cancer and has a poor prognosis. At present, there is no effective treatment for it. Methods Here, we used different concentrations of GSK-J4 or a combination of GSK-J4 and doxorubicin to treat human Cal-62, 8505C, and 8305C anaplastic thyroid cancer (ATC) cell lines. The in vitro experiments were performed using cell viability assays, cell cycle assays, annexin-V/PI binding assays, Transwell migration assays, and wound-healing assays. Tumor xenograft models were used to observe effects in vivo. Results The half maximal inhibitory concentration (IC50) of GSK-J4 in Cal-62 cells was 1.502 μM, and as the dose of GSK-J4 increased, more ATC cells were blocked in the G2-M and S stage. The combination of GSK-J4 and doxorubicin significantly increased the inhibitory effect on proliferation, especially in KRAS-mutant ATC cells in vivo (inhibition rate 38.0%) and in vitro (suppresses rate Fa value 0.624, CI value 0.673). The invasion and migration abilities of the KRAS-mutant cell line were inhibited at a low concentration (p < 0.05). Conclusions The combination of GSK-J4 with doxorubicin in KRAS-mutant ATC achieved tumor-suppressive effects at a low dose. The synergy of the combination of GSK-J4 and doxorubicin may make it an effective chemotherapy regimen for KRAS-mutant ATC.
Collapse
Affiliation(s)
- Bo Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bing Lu
- Institute of Urology of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - I-Yun Hsieh
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Liang
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zicheng Sun
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Yi
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Weiming Lv
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
55
|
Ding YG, Ren YL, Xu YS, Wei CS, Zhang YB, Zhang SK, Guo CA. Identification of key candidate genes and pathways in anaplastic thyroid cancer by bioinformatics analysis. Am J Otolaryngol 2020; 41:102434. [PMID: 32093976 DOI: 10.1016/j.amjoto.2020.102434] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 02/10/2020] [Accepted: 02/16/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is a refractory and poor prognosis tumor Present study aimed to investigate the underlying biological functions and pathways involved in the development of ATC and to identify potential hub genes and candidate biomarkers of ATC. MATERIALS AND METHODS Bioinformatics analyses were performed to identify the differentially expressed genes (DEGs) between ATC tissue samples and adjacent normal tissue samples. Protein-protein interaction (PPI) networks of the DEGs were constructed using Search Tool for the Retrieval of Interacting Genes online tool and Cytoscape software and divided into sub-networks using the Molecular Complex Detection (MCODE) plug-in. DEGs in each module was analyzed by enrichment analysis of the KEGG Orthology Based Annotation System (KOBAS) web software version 3.0. Eventually, the hub genes from bioinformatics analysis were verified by qRT-PCR assay in different ATC cell lines. RESULTS Thirty hub genes were selected and three modules were built by the Cytoscape software from the PPI network. Seven genes (CDK1, CCNB2, BUB1B, CDC20, RRM2, CHEK1 and CDC45) were screened from thirty hub genes. Enrichment analysis showed that these hub genes were primarily accumulated in 'cell cycle', 'p53 signaling pathway', 'viral carcinogenesis', 'pyrimidine metabolism' and 'ubiquitin mediated proteolysis'. The results of qRT-PCR indicated that seven hub genes were unregulated in three ATC cell lines compared with normal thyroid gland cell. CONCLUSIONS These findings suggest that CDK1, CCNB2, BUB1B, CDC20, RRM2, CHEK1 and CDC45 may serve as novel diagnosis biomarkers and potential therapeutic target for ATC.
Collapse
Affiliation(s)
- Yong-Gang Ding
- Emergency Department, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, PR China
| | - Yu-Lin Ren
- Department of Urology Surgery, Affiliated Hospital of Northwest Minzu University, Second People's Hospital of Gansu Province, Lanzhou 730030, Gansu, PR China
| | - Yang-Shan Xu
- Department of Surgery, Liujiaxia Hospital of Fourth Engineering Bureau of China Water Resources and Hydropower, Linxia 731801, Gansu, PR China
| | - Chang-Sheng Wei
- Department of Thyroid Mammary Gland, Gansu Provincial Cancer Hospital, Lanzhou 730030, Gansu, PR China
| | - Yong-Bin Zhang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730030, Gansu, PR China
| | - Shou-Kai Zhang
- Department of Otolaryngology Head and Neck Surgery, Gansu Provincial Hospital, Lanzhou 730030, Gansu, PR China.
| | - Chang-An Guo
- Emergency Department, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, PR China.
| |
Collapse
|
56
|
Ma M, Lin B, Wang M, Liang X, Su L, Okose O, Lv W, Li J. Immunotherapy in anaplastic thyroid cancer. Am J Transl Res 2020; 12:974-988. [PMID: 32269728 PMCID: PMC7137046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 02/13/2020] [Indexed: 06/11/2023]
Abstract
Anaplastic thyroid cancer (ATC) is one of the worst human malignancies, with an associated median survival of only 5 months. It is resistant to conventional thyroid cancer therapies, including radioiodine and thyroid-stimulating hormone suppression. Cancer immunotherapy has emerged over the past few decades as a transformative approach to treating a wide variety of cancers. However, immunotherapy for ATC is still in the experimental stage. This review will cover several strategies of immunotherapy and discuss the possible application of these strategies in the treatment of ATC (such as targeted therapy for tumor-associated macrophages, cancer vaccines, adoptive immunotherapy, monoclonal antibodies and immune checkpoint blockade) with the hope of improving the prognosis of ATC in the future.
Collapse
Affiliation(s)
- Maoguang Ma
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Bo Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Mingdian Wang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterGuangzhou, China
| | - Xiaoli Liang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Lei Su
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Okenwa Okose
- Texas A & M College of MedicineCollege Station, TX 77843, USA
- Division of Thyroid and Parathyroid Surgery, Massachusetts Eye and Ear Infirmary, Harvard Medical SchoolBoston, MA, USA
| | - Weiming Lv
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
- Division of Thyroid and Parathyroid Surgery, Massachusetts Eye and Ear Infirmary, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
57
|
Ferrari SM, Elia G, Ragusa F, Ruffilli I, La Motta C, Paparo SR, Patrizio A, Vita R, Benvenga S, Materazzi G, Fallahi P, Antonelli A. Novel treatments for anaplastic thyroid carcinoma. Gland Surg 2020; 9:S28-S42. [PMID: 32055496 DOI: 10.21037/gs.2019.10.18] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Anaplastic thyroid cancer (ATC) is one of the deadliest human cancers and it is less than 2% of thyroid carcinomas (TCs). The standard treatment of ATC includes surgical debulking, accelerated hyperfractionated external beam radiation therapy (EBRT), and chemotherapy, in particular with cisplatin or doxorubicin, achieving about 10 months of median survival. Since ATC is a rare and aggressive tumor, it is still challenging to predict the patient clinical therapy responsiveness. Several genetic mutations have been described in ATC, involved in different molecular pathways linked to tumor progression, and novel therapies acting on these molecular pathways have been investigated, to improve the quality of life in these patients. Here we review the new targeted therapy of ATC. We report interesting results obtained with molecules targeting different pathways: angiogenesis (vandetanib, combretastatin, sorafenib, lenvatinib, sunitinib, CLM94, CLM3, etc.); EGFR (gefitinib, docetaxel); BRAF (dabrafenib/trametinib, vemurafenib); PPARγ agonists (rosiglitazone, pioglitazone, efatutazone); PD-1 and PD-L1 (pembrolizumab); TERT. To escape resistance to monotherapies, the evaluation of combination strategies with radiotherapy, chemotherapy, or targeted drugs is ongoing. The results of clinical trials with dabrafenib and trametinib led to the approval from FDA of this combination for patients with BRAF V600E mutated ATC with locally advanced, unresectable, or metastatic ATC. The anti-PD-L1 antibody immunotherapy, alone or combined with a BRAF inhibitor, has been shown also promising in the treatment of ATC. Furthermore, to increase the therapeutic success and not to use ineffective or even harmful treatments, a real tailored therapy should be pursued, and this can be achieved thanks to the new available genomic analysis methods and to the possibility to test in vitro novel treatments directly in primary cells from each ATC patient. Exploring new treatment strategies is mandatory to improve the survival of these patients, guaranteeing a good quality of life.
Collapse
Affiliation(s)
| | - Giusy Elia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ilaria Ruffilli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | | - Armando Patrizio
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Vita
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University of Messina, Messina, Italy
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University of Messina, Messina, Italy.,Master Program on Childhood, Adolescent and Women's Endocrine Health, University of Messina, Messina, Italy.,Interdepartmental Program on Molecular & Clinical Endocrinology, and Women's Endocrine Health, University hospital, A.O.U. Policlinico Gaetano Martino, Messina, Italy
| | - Gabriele Materazzi
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Poupak Fallahi
- Department of Translational Research of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
58
|
Meda Spaccamela V, Valencia RG, Pastukhov O, Duppenthaler A, Dettmer MS, Erb J, Steiner UC, Hillinger S, Speckmann C, Ehl S, Reichenbach J, Siler U. High Levels of IL-18 and IFN-γ in Chronically Inflamed Tissue in Chronic Granulomatous Disease. Front Immunol 2019; 10:2236. [PMID: 31681257 PMCID: PMC6813411 DOI: 10.3389/fimmu.2019.02236] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Chronic granulomatous disease (CGD) is caused by a malfunctioning nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex in phagocytes, leading to impaired bacterial and fungal killing and hyperinflammation. Objective: To characterize macrophage subsets and cytokine/chemokine signaling loops involved in CGD tissue hyperinflammation. Methods: Cytokine/chemokine production and surface marker expression were analyzed in inflamed tissue of four CGD patients and compared to cytokine/chemokine released by CGD macrophages upon priming to different macrophage subpopulations. Furthermore, the re-priming capacity of CGD pro-inflammatory M1 to M2a anti-inflammatory macrophages was evaluated. Results: In human CGD inflammatory tissue, IL-18 and IFN-γ were detected in significant quantity. Immunofluorescence analysis identified macrophages as one source of IL-18 in inflamed tissue. In vitro, CGD macrophages could be primed and re-primed into all inflammatory/anti-inflammatory macrophage subpopulations. IL-18 was also released by M1 CGD and control macrophages. Conclusion: CGD pro-inflammatory M1 macrophages remain M1 primed in vivo. As CGD M1 macrophages can be re-primed to anti-inflammatory M2a phenotype in vitro, macrophages are kept in M1 state in vivo by a persistent pro-inflammatory environment. Our results suggest a paracrine signaling loop between M1 macrophage derived IL-18 and non-macrophage derived IFN-γ maintaining macrophage pro-inflammatory activity in CGD tissue.
Collapse
Affiliation(s)
- Virginia Meda Spaccamela
- Division of Immunology, University Children's Hospital and Children's Research Center, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Rocio G Valencia
- Division of Immunology, University Children's Hospital and Children's Research Center, Zurich, Switzerland
| | - Oleksandr Pastukhov
- Division of Immunology, University Children's Hospital and Children's Research Center, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Andrea Duppenthaler
- Unit of Pediatric Infectious Diseases, University Children's Hospital Bern, Bern, Switzerland
| | | | - Juliane Erb
- Center for Dentistry, University of Zurich, Zurich, Switzerland
| | - Urs C Steiner
- Department of Clinical Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Sven Hillinger
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Carsten Speckmann
- Faculty of Medicine, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Institute for Immunodeficiency, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Stephan Ehl
- Faculty of Medicine, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Institute for Immunodeficiency, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Janine Reichenbach
- Division of Immunology, University Children's Hospital and Children's Research Center, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Unit of Pediatric Infectious Diseases, University Children's Hospital Bern, Bern, Switzerland.,Center for Applied Biotechnology and Molecular Medicine, University Zurich, Zurich, Switzerland
| | - Ulrich Siler
- Division of Immunology, University Children's Hospital and Children's Research Center, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
59
|
Boos LA, Schmitt A, Moch H, Komminoth P, Simillion C, Marinoni I, Nikiforov YE, Nikiforova MN, Perren A, Dettmer MS. MiRNAs Are Involved in Tall Cell Morphology in Papillary Thyroid Carcinoma. Cancers (Basel) 2019; 11:cancers11060885. [PMID: 31242620 PMCID: PMC6628239 DOI: 10.3390/cancers11060885] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 01/06/2023] Open
Abstract
Five percent of papillary thyroid carcinomas (PTC) show an adverse clinical outcome (ACO). The tall cell variant of papillary thyroid carcinomas (TCV) is a good predictor of an ACO, however, the identification of tall-cells is subjective. Micro RNAs are short non-coding ribonucleic acids (miRNA). Their expression in PTC could be a powerful, more objective predictor of prognosis. METHODS Forty-four PTC underwent miRNA profiling, twenty-four of them were TCV. The miRNA dataset was validated by analysis of expression of known target proteins (vascular endothelial growth factor (VEGF) and phosphatase and tensin homolog (PTEN)) in 125 patients including 48 TCV and 57 with an ACO. RESULTS One hundred and forty-nine miRNAs were significantly associated with an ACO, seventy-one of them with TC-morphology. Twenty-two miRNAs were identified as targets for VEGF and thirty-two as targets for PTEN. In univariate and multivariable analysis, reduced expression of PTEN and an increased expression of VEGF were associated with shorter relapse free survival. A classifier, including TC-morphology, pT-stage, VEGF, and PTEN, predicted relapse with an 80% accuracy. CONCLUSIONS Some miRNAs predict outcome in PTC and are involved in TC-morphology in PTC. These miRNAs may serve as more objective indicators of an ACO than tall cell morphology. PTEN and VEGF protein expression are prognostically relevant and are at least partially regulated by miRNAs.
Collapse
Affiliation(s)
- Laura A Boos
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH 3008 Bern, Switzerland.
| | - Anja Schmitt
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH 3008 Bern, Switzerland.
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| | - Paul Komminoth
- Institute of Surgical Pathology, Stadtspital Triemli, Birmensdorferstr. 497, 8063 Zürich, Switzerland.
| | - Cedric Simillion
- Department of BioMedical Research, University of Bern, Murtenstrasse 31, CH 3008 Bern, Switzerland.
| | - Ilaria Marinoni
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH 3008 Bern, Switzerland.
| | - Yuri E Nikiforov
- Department of Pathology and Laboratory Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| | - Marina N Nikiforova
- Department of Pathology and Laboratory Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| | - Aurel Perren
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH 3008 Bern, Switzerland.
| | - Matthias S Dettmer
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH 3008 Bern, Switzerland.
| |
Collapse
|