1
|
Bandini A, Banchi M, Orlandi P, Vaglini F, Alì G, Fontanini G, Ottani A, Giuliani D, Vandini E, Francia G, Carli M, Scarselli M, Bocci G. Melanocortin-4 Receptor Antagonism Inhibits Colorectal and Anaplastic Thyroid Cancer In Vitro and In Vivo. J Clin Med 2025; 14:1165. [PMID: 40004697 PMCID: PMC11856147 DOI: 10.3390/jcm14041165] [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: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives MC4R expression and its role in colorectal and anaplastic thyroid cancers, where resistance to therapy and lack of standard treatments remain significant challenges, are poorly understood. This study aimed to investigate MC4R as a potential therapeutic target in these cancers using the selective antagonist ML00253764 (ML), alone and in combination with vinorelbine (VNR) and irinotecan (or its active metabolite SN-38). Methods: Human colorectal adenocarcinoma HT-29, Caco-2, and anaplastic thyroid carcinoma 8305C cell lines were used. MC4R expression was assessed by Real-Time PCR with validated primers (Assay ID Hs00271877_s1), immunofluorescence, and Western blotting. Proliferation and apoptosis assays were conducted with ML, and synergy with VNR and SN-38 was evaluated by Combination Index and Loewe methods. ERK1/2 phosphorylation was measured using an ELISA assay. In vivo studies were conducted by injecting tumor cells into Athymic Nude-Foxn1nu mice, treated with ML, VNR, irinotecan, or their combinations. Results: MC4R expression was confirmed in all cell lines. ML treatment inhibited MC4R, producing antiproliferative and pro-apoptotic effects, with IC50 values of 7667 ± 2144.6 nM (8305C), 806.4 ± 321.8 nM (HT-29), and 2993 ± 1135.2 nM (Caco-2). In combination with VNR and SN-38, ML exhibited significant synergy in vitro and reduced tumor volume in vivo without causing weight loss or adverse effects in mice. Conclusions This study identifies ML as a promising therapeutic agent that, when combined with chemotherapy, may offer a novel strategy for treating colorectal and anaplastic thyroid cancers.
Collapse
Affiliation(s)
- Arianna Bandini
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, 56126 Pisa, Italy; (M.B.); (F.V.); (M.C.); (M.S.)
| | - Marta Banchi
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, 56126 Pisa, Italy; (M.B.); (F.V.); (M.C.); (M.S.)
| | - Paola Orlandi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, 56126 Pisa, Italy;
| | - Francesca Vaglini
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, 56126 Pisa, Italy; (M.B.); (F.V.); (M.C.); (M.S.)
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università di Pisa, 56126 Pisa, Italy; (G.A.); (G.F.)
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università di Pisa, 56126 Pisa, Italy; (G.A.); (G.F.)
| | - Alessandra Ottani
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Sezione di Farmacologia e Medicina Molecolare, Università di Modena e Reggio Emilia, 41121 Modena, Italy; (A.O.); (D.G.); (E.V.)
| | - Daniela Giuliani
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Sezione di Farmacologia e Medicina Molecolare, Università di Modena e Reggio Emilia, 41121 Modena, Italy; (A.O.); (D.G.); (E.V.)
| | - Eleonora Vandini
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Sezione di Farmacologia e Medicina Molecolare, Università di Modena e Reggio Emilia, 41121 Modena, Italy; (A.O.); (D.G.); (E.V.)
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso (UTEP), El Paso, TX 79968, USA;
| | - Marco Carli
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, 56126 Pisa, Italy; (M.B.); (F.V.); (M.C.); (M.S.)
| | - Marco Scarselli
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, 56126 Pisa, Italy; (M.B.); (F.V.); (M.C.); (M.S.)
| | - Guido Bocci
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, 56126 Pisa, Italy; (M.B.); (F.V.); (M.C.); (M.S.)
| |
Collapse
|
2
|
Zheng X, Zhang X, Yu S. Organoids derived from metastatic cancers: Present and future. Heliyon 2024; 10:e30457. [PMID: 38720734 PMCID: PMC11077038 DOI: 10.1016/j.heliyon.2024.e30457] [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] [Received: 01/03/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
Organoids are three-dimensional structures derived from primary tissue or tumors that closely mimic the architecture, histology, and function of the parental tissue. In recent years, patient-derived organoids (PDOs) have emerged as powerful tools for modeling tumor heterogeneity, drug screening, and personalized medicine. Although most cancer organoids are derived from primary tumors, the ability of organoids from metastatic cancer to serve as a model for studying tumor biology and predicting the therapeutic response is an area of active investigation. Recent studies have shown that organoids derived from metastatic sites can provide valuable insights into tumor biology and may be used to validate predictive models of the drug response. In this comprehensive review, we discuss the feasibility of culturing organoids from multiple metastatic cancers and evaluate their potential for advancing basic cancer research, drug development, and personalized therapy. We also explore the limitations and challenges associated with using metastasis organoids for cancer research. Overall, this review provides a comprehensive overview of the current state and future prospects of metastatic cancer-derived organoids.
Collapse
Affiliation(s)
- Xuejing Zheng
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinxin Zhang
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengji Yu
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Stribbling SM, Beach C, Ryan AJ. Orthotopic and metastatic tumour models in preclinical cancer research. Pharmacol Ther 2024; 257:108631. [PMID: 38467308 PMCID: PMC11781865 DOI: 10.1016/j.pharmthera.2024.108631] [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: 08/17/2023] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Mouse models of disease play a pivotal role at all stages of cancer drug development. Cell-line derived subcutaneous tumour models are predominant in early drug discovery, but there is growing recognition of the importance of the more complex orthotopic and metastatic tumour models for understanding both target biology in the correct tissue context, and the impact of the tumour microenvironment and the immune system in responses to treatment. The aim of this review is to highlight the value that orthotopic and metastatic models bring to the study of tumour biology and drug development while pointing out those models that are most likely to be encountered in the literature. Important developments in orthotopic models, such as the increasing use of early passage patient material (PDXs, organoids) and humanised mouse models are discussed, as these approaches have the potential to increase the predictive value of preclinical studies, and ultimately improve the success rate of anticancer drugs in clinical trials.
Collapse
Affiliation(s)
- Stephen M Stribbling
- Department of Chemistry, University College London, Gower Street, London WC1E 6BT, UK.
| | - Callum Beach
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Anderson J Ryan
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK; Fast Biopharma, Aston Rowant, Oxfordshire, OX49 5SW, UK.
| |
Collapse
|
4
|
Shen H, Zhu R, Liu Y, Hong Y, Ge J, Xuan J, Niu W, Yu X, Qin JJ, Li Q. Radioiodine-refractory differentiated thyroid cancer: Molecular mechanisms and therapeutic strategies for radioiodine resistance. Drug Resist Updat 2024; 72:101013. [PMID: 38041877 DOI: 10.1016/j.drup.2023.101013] [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: 08/13/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 12/04/2023]
Abstract
Radioiodine-refractory differentiated thyroid cancer (RAIR-DTC) is difficult to treat with radioactive iodine because of the absence of the sodium iodide transporter in the basement membrane of thyroid follicular cells for iodine uptake. This is usually due to the mutation or rearrangement of genes and the aberrant activation of signal pathways, which result in abnormal expression of thyroid-specific genes, leading to resistance of differentiated thyroid cancer cells to radioiodine therapy. Therefore, inhibiting the proliferation and growth of RAIR-DTC with multikinase inhibitors and other drugs or restoring its differentiation and then carrying out radioiodine therapy have become the first-line treatment strategies and main research directions. The drugs that regulate these kinases or signaling pathways have been studied in clinical and preclinical settings. In this review, we summarized the major gene mutations, gene rearrangements and abnormal activation of signaling pathways that led to radioiodine resistance of RAIR-DTC, as well as the medicine that have been tested in clinical and preclinical trials.
Collapse
Affiliation(s)
- Huize Shen
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Rui Zhu
- Department of stomatology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Yanyang Liu
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yangjian Hong
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiaming Ge
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jie Xuan
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenyuan Niu
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xuefei Yu
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Jiang-Jiang Qin
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Qinglin Li
- Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Choi HR, Kim K. Mouse Models to Examine Differentiated Thyroid Cancer Pathogenesis: Recent Updates. Int J Mol Sci 2023; 24:11138. [PMID: 37446316 DOI: 10.3390/ijms241311138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Although the overall prognosis of differentiated thyroid cancer (DTC), the most common endocrine malignancy, is favorable, a subset of patients exhibits aggressive features. Therefore, preclinical models that can be utilized to investigate DTC pathogenesis and novel treatments are necessary. Various mouse models have been developed based on advances in thyroid cancer genetics. This review focuses on recent progress in mouse models that have been developed to elucidate the molecular pathogenesis of DTC.
Collapse
Affiliation(s)
- Hye Ryeon Choi
- Department of Surgery, Eulji Medical Center, Eulji University School of Medicine, Seoul 01830, Republic of Korea
| | - Kwangsoon Kim
- Department of Surgery, College of Medicine, Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
6
|
Jeon MJ, Haugen BR. Preclinical Models of Follicular Cell-Derived Thyroid Cancer: An Overview from Cancer Cell Lines to Mouse Models. Endocrinol Metab (Seoul) 2022; 37:830-838. [PMID: 36604954 PMCID: PMC9816502 DOI: 10.3803/enm.2022.1636] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/11/2022] [Indexed: 12/28/2022] Open
Abstract
The overall prognosis of thyroid cancer is excellent, but some patients have grossly invasive disease and distant metastases with limited responses to systemic therapies. Thus, relevant preclinical models are needed to investigate thyroid cancer biology and novel treatments. Different preclinical models have recently emerged with advances in thyroid cancer genetics, mouse modeling and new cell lines. Choosing the appropriate model according to the research question is crucial to studying thyroid cancer. This review will discuss the current preclinical models frequently used in thyroid cancer research, from cell lines to mouse models, and future perspectives on patient-derived and humanized preclinical models in this field.
Collapse
Affiliation(s)
- Min Ji Jeon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bryan R. Haugen
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Corresponding author: Bryan R. Haugen. Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, 12801 East 17th Avenue, Aurora, CO 80045, USA Tel: +1-303-724-3921, Fax: +1-303-724-3920, E-mail:
| |
Collapse
|
7
|
Zerfaoui M, Tsumagari K, Toraih E, Errami Y, Ruiz E, Elaasar MSM, Krzysztof M, Sholl AB, Magdeldin S, Soudy M, Abd Elmageed ZY, Boulares AH, Kandil E. Nuclear interaction of Arp2/3 complex and BRAF V600E promotes aggressive behavior and vemurafenib resistance of thyroid cancer. Am J Cancer Res 2022; 12:3014-3033. [PMID: 35968344 PMCID: PMC9360225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/12/2022] [Indexed: 04/22/2023] Open
Abstract
The presence of mutant BRAF V600E correlates with the risk of recurrence in papillary thyroid cancer (PTC) patients. However, not all PTC patients with BRAF V600E are associated with poor prognosis. Thus, understanding the mechanisms by which certain PTC patients with nuclear BRAF V600E become aggressive and develop resistance to a selective BRAF inhibitor, PLX-4032, is urgently needed. The effect of nuclear localization of BRAFV600E using in vitro studies, xenograft mouse-model and human tissues was evaluated. PTC cells harboring a nuclear localization signal (NLS) of BRAFV600E were established and examined in nude mice implanted with TPC1-NLS-BRAFV600E cells followed by PLX-4032 treatment. Immunohistochemical (IHC) analysis was performed on 100 PTC specimens previously confirmed that they have BRAFV600E mutations. Our results demonstrate that 21 of 100 (21%) PTC tissues stained with specific BRAFV600E antibody had nuclear staining with more aggressive features compared to their cytosolic counterparts. In vitro studies show that BRAFV600E is transported between the nucleus and the cytosol through CRM1 and importin (α/β) system. Sequestration of BRAFV600E in the cytosol sensitized resistant cells to PLX-4032, whereas nuclear BRAFV600E was associated with aggressive phenotypes and developed drug resistance. Proteomic analysis revealed Arp2/3 complex members, actin-related protein 2 (ACTR2 aliases ARP2) and actin-related protein 3 (ACTR3 aliases ARP3), as the most enriched nuclear BRAFV600E partners. ACTR3 was highly correlated to lymph node stage and extrathyroidal extension and was validated with different functional assays. Our findings provide new insights into the clinical utility of the nuclear BRAFV600E as a prognostic marker for PTC aggressiveness and determine the efficacy of selective BRAFV600E inhibitor treatment which opens new avenues for future treatment options.
Collapse
Affiliation(s)
- Mourad Zerfaoui
- Department of Surgery, Tulane University School of MedicineUSA
| | - Koji Tsumagari
- Department of Surgery, Tulane University School of MedicineUSA
| | - Eman Toraih
- Department of Surgery, Tulane University School of MedicineUSA
| | - Youssef Errami
- Department of Surgery, Tulane University School of MedicineUSA
| | - Emmanuelle Ruiz
- Department of Surgery, Tulane University School of MedicineUSA
| | | | - Moroz Krzysztof
- Department of Pathology, Tulane University School of MedicineUSA
| | - Andrew B Sholl
- Department of Otolaryngology, Tulane University School of MedicineUSA
| | - Sameh Magdeldin
- Proteomics Research Program Unit, Basic Research Department, Children Cancer Hospital CairoEgypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal UniversityIsmailia 41522, Egypt
| | - Mohamed Soudy
- Proteomics Research Program Unit, Basic Research Department, Children Cancer Hospital CairoEgypt
| | - Zakaria Y Abd Elmageed
- Department of Surgery, Tulane University School of MedicineUSA
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of LouisianaMonroe, USA
| | - A Hamid Boulares
- Department of Pharmacology, LSU Health Sciences CenterNew Orleans, LA, USA
| | - Emad Kandil
- Department of Surgery, Tulane University School of MedicineUSA
| |
Collapse
|
8
|
Banchi M, Orlandi P, Gentile D, Alì G, Fini E, Fontanini G, Francia G, Bocci G. Synergistic activity of linifanib and irinotecan increases the survival of mice bearing orthotopically implanted human anaplastic thyroid cancer. Am J Cancer Res 2020; 10:2120-2127. [PMID: 32775005 PMCID: PMC7407355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 06/27/2020] [Indexed: 06/11/2023] Open
Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive form of thyroid cancer, and novel combined therapies are urgently needed to prolong patient survival. No data are currently available on the preclinical activity of the combination of linifanib, a CSF-1R inhibitor, and irinotecan in ATC. The aim of the study was to evaluate the in vitro and in vivo activity of linifanib plus irinotecan. Proliferation and apoptosis assays were performed on 8305C and 8505C human ATC cell lines exposed to SN-38, the active metabolite of irinotecan, linifanib alone, and their concomitant combination. Synergism was evaluated by the combination index method. Quantification of pospho-CSF-1R levels was performed by ELISA. In vivo ATC orthotopic xenografts were treated with the single drugs, or their combination, to evaluate their impact on survival. Histology and immunohistochemistry were performed on ATC tissue samples. Both SN-38 and linifanib inhibited in vitro the proliferation of 8305C and 8505C cells in a concentration-dependent manner, whereas their concomitant treatment revealed a strong synergism in the ATC cells. A significant pro-apoptotic activity was found in both ATC cell lines treated with linifanib alone and in combination with SN-38. Moreover, linifanib significantly decreased the levels of phospho-CSF-1R after 24 h and 72 h in both 8505C and 8305C cells, and this was also observed with the concomitant administration of SN-38. In vivo, the combination of linifanib and irinotecan produced a greater survival result than either monotherapy, and resulted in a significant higher median survival. In some of the mice the combination produced a complete response with a macroscopic disappearance of the disease, as confirmed by histology. In conclusion, the synergistic ATC antitumor activity of linifanib/irinotecan combination significantly increased the survival of ATC affected mice and induced some complete responses, suggesting a potential role of this schedule in ATC patient's treatment.
Collapse
Affiliation(s)
- Marta Banchi
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Paola Orlandi
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Daniela Gentile
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell’Area Critica, Università di PisaPisa, Italy
| | - Elisabetta Fini
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell’Area Critica, Università di PisaPisa, Italy
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El PasoEl Paso, TX, USA
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| |
Collapse
|
9
|
Hegedűs L, Rittler D, Garay T, Stockhammer P, Kovács I, Döme B, Theurer S, Hager T, Herold T, Kalbourtzis S, Bankfalvi A, Schmid KW, Führer D, Aigner C, Hegedűs B. HDAC Inhibition Induces PD-L1 Expression in a Novel Anaplastic Thyroid Cancer Cell Line. Pathol Oncol Res 2020; 26:2523-2535. [PMID: 32591993 PMCID: PMC7471186 DOI: 10.1007/s12253-020-00834-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Abstract
While papillary thyroid cancer (PTC) has largely favorable prognosis, anaplastic thyroid cancer (ATC) is a rare but extremely aggressive malignancy with grim clinical outcome. Even though new therapeutic options are emerging for ATC, additional preclinical models and novel combinations are needed for specific subsets of patients. We established a novel cell line (PF49) from the malignant pleural effusion of a 68-year-old male patient with ATC that rapidly transformed from a BRAF and TERT promoter mutant PTC. PF49 cells demonstrated a robust migratory activity in vitro and strong invasive capacity in vivo in a pleural carcinosis model. Combined BRAF and MEK inhibition decreased the proliferation and migration of PF49 cells, however could not induce cell death. Importantly, HDAC inhibitor treatment with SAHA or valproic acid induced cell cycle arrest and strongly increased PD-L1 expression of the tumor cells. Induction of PD-L1 expression was also present when paclitaxel-cisplatin chemotherapeutic treatment was combined with HDAC inhibitor treatment. Increased PD-L1 expression after HDAC inhibition was recapitulated in an international ATC cell model. Our data suggest that HDAC inhibition alone or in combination with standard chemotherapy may potentiate anaplastic thyroid cancer cells for immunotherapy.
Collapse
Affiliation(s)
- Luca Hegedűs
- Department of Thoracic Surgery, University Medicine Essen - Ruhrlandklinik, University Duisburg-Essen, Essen, Germany
| | - Dominika Rittler
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Tamás Garay
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Paul Stockhammer
- Department of Thoracic Surgery, University Medicine Essen - Ruhrlandklinik, University Duisburg-Essen, Essen, Germany.,Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Ildikó Kovács
- National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Balázs Döme
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,National Korányi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, Semmelweis University-National Institute of Oncology, Budapest, Hungary
| | - Sarah Theurer
- Institute of Pathology, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Hager
- Institute of Pathology, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Herold
- Institute of Pathology, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Stavros Kalbourtzis
- Institute of Pathology, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Agnes Bankfalvi
- Institute of Pathology, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Kurt W Schmid
- Institute of Pathology, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Dagmar Führer
- Department of Endocrinology, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Clemens Aigner
- Department of Thoracic Surgery, University Medicine Essen - Ruhrlandklinik, University Duisburg-Essen, Essen, Germany
| | - Balázs Hegedűs
- Department of Thoracic Surgery, University Medicine Essen - Ruhrlandklinik, University Duisburg-Essen, Essen, Germany. .,2nd Department of Pathology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
10
|
Sheen MR, Fields JL, Northan B, Lacoste J, Ang LH, Fiering S. Replication Study: Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis. eLife 2019; 8:45120. [PMID: 31845647 PMCID: PMC6917490 DOI: 10.7554/elife.45120] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 11/06/2019] [Indexed: 01/06/2023] Open
Abstract
As part of the Reproducibility Project: Cancer Biology we published a Registered Report (Fiering et al., 2015) that described how we intended to replicate selected experiments from the paper 'Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis' (Goetz et al., 2011). Here we report the results. Primary mouse embryonic fibroblasts (pMEFs) expressing caveolin 1 (Cav1WT) demonstrated increased extracellular matrix remodeling in vitro compared to Cav1 deficient (Cav1KO) pMEFs, similar to the original study (Goetz et al., 2011). In vivo, we found higher levels of intratumoral stroma remodeling, determined by fibronectin fiber orientation, in tumors from cancer cells co-injected with Cav1WT pMEFs compared to cancer cells only or cancer cells plus Cav1KO pMEFs, which were in the same direction as the original study (Supplemental Figure S7C; Goetz et al., 2011), but not statistically significant. Primary tumor growth was similar between conditions, like the original study (Supplemental Figure S7Ca; Goetz et al., 2011). We found metastatic burden was similar between Cav1WT and Cav1KO pMEFs, while the original study found increased metastases with Cav1WT (Figure 7C; Goetz et al., 2011); however, the duration of our in vivo experiments (45 days) were much shorter than in the study by Goetz et al. (2011) (75 days). This makes it difficult to interpret the difference between the studies as it is possible that the cells required more time to manifest the difference between treatments observed by Goetz et al. We also found a statistically significant negative correlation of intratumoral remodeling with metastatic burden, while the original study found a statistically significant positive correlation (Figure 7Cd; Goetz et al., 2011), but again there were differences between the studies in terms of the duration of the metastasis studies and the imaging approaches that could have impacted the outcomes. Finally, we report meta-analyses for each result.
Collapse
Affiliation(s)
- Mee Rie Sheen
- Geisel School of Medicine at Dartmouth, Department of Microbiology and Immunology, Lebanon, United States
| | - Jennifer L Fields
- Geisel School of Medicine at Dartmouth, Department of Microbiology and Immunology, Lebanon, United States
| | | | | | - Lay-Hong Ang
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
| | - Steven Fiering
- Geisel School of Medicine at Dartmouth, Department of Microbiology and Immunology, Lebanon, United States
| | | |
Collapse
|
11
|
Fiordelisi MF, Cavaliere C, Auletta L, Basso L, Salvatore M. Magnetic Resonance Imaging for Translational Research in Oncology. J Clin Med 2019; 8:jcm8111883. [PMID: 31698697 PMCID: PMC6912299 DOI: 10.3390/jcm8111883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The translation of results from the preclinical to the clinical setting is often anything other than straightforward. Indeed, ideas and even very intriguing results obtained at all levels of preclinical research, i.e., in vitro, on animal models, or even in clinical trials, often require much effort to validate, and sometimes, even useful data are lost or are demonstrated to be inapplicable in the clinic. In vivo, small-animal, preclinical imaging uses almost the same technologies in terms of hardware and software settings as for human patients, and hence, might result in a more rapid translation. In this perspective, magnetic resonance imaging might be the most translatable technique, since only in rare cases does it require the use of contrast agents, and when not, sequences developed in the lab can be readily applied to patients, thanks to their non-invasiveness. The wide range of sequences can give much useful information on the anatomy and pathophysiology of oncologic lesions in different body districts. This review aims to underline the versatility of this imaging technique and its various approaches, reporting the latest preclinical studies on thyroid, breast, and prostate cancers, both on small laboratory animals and on human patients, according to our previous and ongoing research lines.
Collapse
|
12
|
Saiselet M, Pita JM, Augenlicht A, Dom G, Tarabichi M, Fimereli D, Dumont JE, Detours V, Maenhaut C. miRNA expression and function in thyroid carcinomas: a comparative and critical analysis and a model for other cancers. Oncotarget 2018; 7:52475-52492. [PMID: 27248468 PMCID: PMC5239568 DOI: 10.18632/oncotarget.9655] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/16/2016] [Indexed: 12/15/2022] Open
Abstract
As in many cancer types, miRNA expression profiles and functions have become an important field of research on non-medullary thyroid carcinomas, the most common endocrine cancers. This could lead to the establishment of new diagnostic tests and new cancer therapies. However, different studies showed important variations in their research strategies and results. In addition, the action of miRNAs is poorly considered as a whole because of the use of underlying dogmatic truncated concepts. These lead to discrepancies and limits rarely considered. Recently, this field has been enlarged by new miRNA functional and expression studies. Moreover, studies using next generation sequencing give a new view of general miRNA differential expression profiles of papillary thyroid carcinoma. We analyzed in detail this literature from both physiological and differential expression points of view. Based on explicit examples, we reviewed the progresses but also the discrepancies and limits trying to provide a critical approach of where this literature may lead. We also provide recommendations for future studies. The conclusions of this systematic analysis could be extended to other cancer types.
Collapse
Affiliation(s)
- Manuel Saiselet
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Jaime M Pita
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Alice Augenlicht
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Geneviève Dom
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Maxime Tarabichi
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Danai Fimereli
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Jacques E Dumont
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Vincent Detours
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium
| | - Carine Maenhaut
- Institute of Interdisciplinary Research (IRIBHM), University of Brussels, Brussels, Belgium.,WELBIO, School of Medicine, University of Brussels, Brussels, Belgium
| |
Collapse
|
13
|
Greco A, Auletta L, Orlandella FM, Iervolino PLC, Klain M, Salvatore G, Mancini M. Preclinical Imaging for the Study of Mouse Models of Thyroid Cancer. Int J Mol Sci 2017; 18:E2731. [PMID: 29258188 PMCID: PMC5751332 DOI: 10.3390/ijms18122731] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/05/2017] [Accepted: 12/08/2017] [Indexed: 12/23/2022] Open
Abstract
Thyroid cancer, which represents the most common tumors among endocrine malignancies, comprises a wide range of neoplasms with different clinical aggressiveness. One of the most important challenges in research is to identify mouse models that most closely resemble human pathology; other goals include finding a way to detect markers of disease that common to humans and mice and to identify the most appropriate and least invasive therapeutic strategies for specific tumor types. Preclinical thyroid imaging includes a wide range of techniques that allow for morphological and functional characterization of thyroid disease as well as targeting and in most cases, this imaging allows quantitative analysis of the molecular pattern of the thyroid cancer. The aim of this review paper is to provide an overview of all of the imaging techniques used to date both for diagnosis and theranostic purposes in mouse models of thyroid cancer.
Collapse
Affiliation(s)
- Adelaide Greco
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy.
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche-IBB, CNR, 80145 Napoli, Italy.
- CEINGE Biotecnologie Avanzate s.c.ar.l., 80131 Napoli, Italy.
| | | | | | | | - Michele Klain
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy.
| | - Giuliana Salvatore
- IRCCS S.D.N., 80134 Napoli, Italy.
- Dipartimento di Scienze Motorie e del Benessere, Università di Napoli Parthenope, 80133 Napoli, Italy.
| | - Marcello Mancini
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche-IBB, CNR, 80145 Napoli, Italy.
| |
Collapse
|
14
|
Rossfeld KK, Justiniano SE, Ding H, Gong L, Kothandaraman S, Sawant D, Saji M, Wright CL, Kirschner LS, Ringel MD, Tweedle MF, Phay JE. Biological Evaluation of a Fluorescent-Imaging Agent for Medullary Thyroid Cancer in an Orthotopic Model. J Clin Endocrinol Metab 2017; 102:3268-3277. [PMID: 28591772 PMCID: PMC5587064 DOI: 10.1210/jc.2017-00573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 06/02/2017] [Indexed: 02/06/2023]
Abstract
Context The primary and definitive treatment of medullary thyroid cancer (MTC) is surgical resection. Recurrent or residual disease is typically a result of incomplete surgical removal. Objective Our objective is to develop a compound that assists in intraoperative visualization of cancer, which would have the potential to improve surgical cure rates and outcomes. Results We report the biological characterization of Compound-17, which is labeled with IRdye800, allowing fluorescent visualization of MTC mouse models. We found that the agent has high affinity for two human MTC cell lines (TT and MZ-CRC1) in vitro and in vivo. We further tested the affinity of the compound in a newly developed MTC orthotopic xenograft model and found that Compound-17 produces fluorescent signals within MTC-derived orthotopic xenografts in comparison with a sequence-jumbled control compound and surrounding normal tissues. Conclusions Compound-17 is a unique and effective molecule for MTC identification that may have therapeutic potential.
Collapse
Affiliation(s)
- Kara K. Rossfeld
- Department of Surgery, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Steven E. Justiniano
- Division of Endocrinology Diabetes and Metabolism, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Haiming Ding
- Department of Radiology, Wright Center for Innovation in Biomedical Imaging, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Li Gong
- Department of Radiology, Wright Center for Innovation in Biomedical Imaging, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Shankaran Kothandaraman
- Department of Radiology, Wright Center for Innovation in Biomedical Imaging, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Dwitiya Sawant
- Department of Molecular Genetics, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Motoyasu Saji
- Division of Endocrinology Diabetes and Metabolism, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Chadwick L. Wright
- Department of Radiology, Wright Center for Innovation in Biomedical Imaging, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Lawrence S. Kirschner
- Division of Endocrinology Diabetes and Metabolism, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
- Molecular Biology and Cancer Genetics, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Matthew D. Ringel
- Division of Endocrinology Diabetes and Metabolism, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
- Molecular Biology and Cancer Genetics, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - Michael F. Tweedle
- Department of Radiology, Wright Center for Innovation in Biomedical Imaging, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| | - John E. Phay
- Department of Surgery, Ohio State University-Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
15
|
Arcolia V, Journe F, Wattier A, Leteurtre E, Renaud F, Gabius HJ, Remmelink M, Decaestecker C, Rodriguez A, Boutry S, Laurent S, Saussez S. Galectin-1 is a diagnostic marker involved in thyroid cancer progression. Int J Oncol 2017; 51:760-770. [PMID: 28677745 PMCID: PMC5564411 DOI: 10.3892/ijo.2017.4065] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/27/2017] [Indexed: 12/20/2022] Open
Abstract
Fine-needle aspiration (FNA) is the most commonly used pre-operative technique for diagnosis of malignant thyroid tumor. However, many benign lesions, with indeterminate diagnosis following FNA, are referred to surgery. Based on multifunctionality of the endogenous galectin-1, we aimed to assess its status for early diagnosis of thyroid cancer. Immunohistochemistry for galectin-1 and -3 was performed on a clinical series of 69 cases of thyroid lesions. Galectin-1 expression was further examined in two additional tissue microarrays (TMA) composed of 66 follicular adenomas and 66 papillary carcinomas in comparison to galectin-3 and cytokeratin-19 (CK19). In addition, a knockdown of galectin-1 in papillary (TPC-1) and anaplastic (8505C) thyroid cancer cell lines was achieved by lentiviral transduction for in vitro experiments. A murine orthotopic thyroid cancer model was used to investigate tumor growth and metastatic ability. Immunohistochemical analyses of galectin-1 and -3 in the series of 69 cases of thyroid lesions revealed that galectin-1 was completely absent in the epithelial compartment of all benign thyroid lesions. Levels of both galectins significantly increased in the cytoplasmic compartment of malignant thyroid cells. Galectin-1 expression in the TMA yielded an excellent specificity (97%), while galectin-3 and CK19 presented a higher sensitivity (>97%) in discriminating benign from malignant thyroid lesions. In vitro experiments revealed that migration was negatively affected in TPC-1 galectin-1 knockdown (KD) cells, and that proliferation and invasion capacity of 8505C cells decreased after galectin-1 KD. Moreover, an orthotopic mouse model displayed a lower rate of tumor development with galectin-1 KD thyroid anaplastic cancer cells than in the control. Our findings support the introduction of galectin-1 as a reliable diagnostic marker for thyroid carcinomas. Its involvement in cell proliferation, migration, invasion and tumor growth also intimate functional involvement of galectin-1 in the progression of thyroid carcinoma, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Vanessa Arcolia
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| | - Fabrice Journe
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| | - Aurore Wattier
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| | - Emmanuelle Leteurtre
- Université Lille, Inserm, CHU Lille, UMR-S 1172 - JPARC - Jean-Pierre Aubert Research Center, F-59000 Lille, France
| | - Florence Renaud
- Université Lille, Inserm, CHU Lille, UMR-S 1172 - JPARC - Jean-Pierre Aubert Research Center, F-59000 Lille, France
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig Maximilians University, D-80539 Munich, Germany
| | - Myriam Remmelink
- Department of Pathology, Hospital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Decaestecker
- Laboratory of Image, Signal Processing and Acoustics (LISA), Ecole Polytechnique de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandra Rodriguez
- Department of Oto-Rhino-Laryngology, CHU Saint-Pierre, Université Libre de Bruxelles, Brussels, Belgium
| | - Sébastien Boutry
- Center for Microscopy and Molecular Imaging (CMMI), Gosselies, Belgium
| | - Sophie Laurent
- Center for Microscopy and Molecular Imaging (CMMI), Gosselies, Belgium
| | - Sven Saussez
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| |
Collapse
|
16
|
Yin DT, Xu J, Lei M, Li H, Wang Y, Liu Z, Zhou Y, Xing M. Characterization of the novel tumor-suppressor gene CCDC67 in papillary thyroid carcinoma. Oncotarget 2016; 7:5830-41. [PMID: 26716505 PMCID: PMC4868724 DOI: 10.18632/oncotarget.6709] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023] Open
Abstract
Background Some studies showed an association of coiled-coil domain-containing (CCDC) genes with cancers. Our previous limited data specifically suggested a possible pathogenic role of CCDC67 in papillary thyroid cancer (PTC), but this has not been firmly established. The present study was to further investigate and establish this role of CCDC67 in PTC. Results The expression of CCDC67, both at mRNA and protein levels, was sharply down-regulated in PTC compared with normal thyroid tissues. Lower CCDC67 expression was significantly associated with aggressive tumor behaviors, such as advanced tumor stages and lymph node metastasis, as well as BRAF mutation. Introduced expression of CCDC67 in TPC-1 cells robustly inhibited cell proliferation, colony formation and migration, induced G1 phase cell cycle arrest, and increased cell apoptosis. Methods Primary PTC tumors and matched normal thyroid tissues were obtained from 200 unselected patients at the initial surgery for detection of CCDC67 mRNA and protein by RT-PCR and Western blotting analyses, respectively. Genomic DNA sequencing was performed to detect BRAF mutation in PTC tumors. Clinicopathological data were retrospectively reviewed for correlation analyses. PTC cell line TPC-1 with stable transfection of CCDC67 was used to investigate the functions of CCDC67. Conclusions This large study demonstrates down-regulation of CCDC67 in PTC, an inverse relationship between CCDC67 expression and PTC aggressiveness and BRAF mutation, and a robust inhibitory effect of CCDC67 on PTC cellular activities. These results are consistent with CCDC67 being a novel and impaired tumor suppressor gene in PTC, providing important prognostic and therapeutic implications for this cancer.
Collapse
Affiliation(s)
- De Tao Yin
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Jianhui Xu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Mengyuan Lei
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Hongqiang Li
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Yongfei Wang
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Zhen Liu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Mingzhao Xing
- Division of Endocrinology and Metabolism, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
17
|
Metastasis-associated MCL1 and P16 copy number alterations dictate resistance to vemurafenib in a BRAFV600E patient-derived papillary thyroid carcinoma preclinical model. Oncotarget 2016; 6:42445-67. [PMID: 26636651 PMCID: PMC4767444 DOI: 10.18632/oncotarget.6442] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 10/22/2015] [Indexed: 01/08/2023] Open
Abstract
BRAFV600E mutation exerts an essential oncogenic function in many tumors, including papillary thyroid carcinoma (PTC). Although BRAFV600E inhibitors are available, lack of response has been frequently observed. To study the mechanism underlying intrinsic resistance to the mutant BRAFV600E selective inhibitor vemurafenib, we established short-term primary cell cultures of human metastatic/recurrent BRAFV600E-PTC, intrathyroidal BRAFV600E-PTC, and normal thyroid (NT). We also generated an early intervention model of human BRAFV600E-PTC orthotopic mouse. We find that metastatic BRAFV600E-PTC cells elicit paracrine-signaling which trigger migration of pericytes, blood endothelial cells and lymphatic endothelial cells as compared to BRAFWT-PTC cells, and show a higher rate of invasion. We further show that vemurafenib therapy significantly suppresses these aberrant functions in non-metastatic BRAFV600E-PTC cells but lesser in metastatic BRAFV600E-PTC cells as compared to vehicle treatment. These results concur with similar folds of down-regulation of tumor microenvironment–associated pro-metastatic molecules, with no effects in BRAFWT-PTC and NT cells. Our early intervention preclinical trial shows that vemurafenib delays tumor growth in the orthotopic BRAFWT/V600E-PTC mice. Importantly, we identify high copy number gain of MCL1 (chromosome 1q) and loss of CDKN2A (P16, chromosome 9p) in metastatic BRAFV600E-PTC cells which are associated with resistance to vemurafenib treatment. Critically, we demonstrate that combined vemurafenib therapy with BCL2/MCL1 inhibitor increases metastatic BRAFV600E-PTC cell death and ameliorates response to vemurafenib treatment as compared to single agent treatment. In conclusion, short-term PTC and NT cultures offer a predictive model for evaluating therapeutic response in patients with PTC. Our PTC pre-clinical model suggests that combined targeted therapy might be an important therapeutic strategy for metastatic and refractory BRAFV600E-positive PTC.
Collapse
|
18
|
Greco A, Albanese S, Auletta L, Mirabelli P, Zannetti A, D'Alterio C, Di Maro G, Orlandella FM, Salvatore G, Soricelli A, Salvatore M. High-Frequency Ultrasound-Guided Injection for the Generation of a Novel Orthotopic Mouse Model of Human Thyroid Carcinoma. Thyroid 2016; 26:552-8. [PMID: 26844598 DOI: 10.1089/thy.2015.0511] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Thyroid carcinoma is the most common endocrine malignancy and has an increasing incidence. High-frequency ultrasound (HFUS) has a spatial resolution of 30 μm, which is a property that has been exploited for thyroid visualization and analysis in mice. The aim of this study was to generate a novel orthotopic mouse model of human follicular thyroid carcinoma (FTC) using an HFUS-guided injection system. METHODS Twenty Balb/C nude mice were injected in the right lobe of the thyroid with 2 × 10(6) FTC-133 cells using the microinjection HFUS-guided system, and 20 mice, used as a control, underwent surgical orthotopic implantation of 2 × 10(6) FTC-133 cells in the right lobe of the thyroid. All mice underwent HFUS imaging two weeks after cell injection; HFUS examinations and tumor volume (TV) measurements were repeated weekly. Micro-computed tomography was performed at different time points to determine whether lung metastasis had occurred. TVs were compared between the two models (surgical vs. HFUS-guided) using the Mann-Whitney U-test, and the Mantel-Cox log-rank test was applied to evaluate the death hazard. Hematoxylin and eosin analysis of formalin-fixed, paraffin-embedded mouse tissue was performed to validate the in vivo imaging results. RESULTS Of the HFUS-guided injected mice, 9/18 survived up to 40 days after the injection of tumor cells. Mice injected surgically had 100% mortality at day 29. Of 38 mice, 29 (14/18 HFUS, 15/20 surgical) showed metastasis in the salivary glands and lymph nodes, and 13 (10/18 HFUS, 3/20 surgical) also showed metastasis in the lungs, which was confirmed by histological analysis. In the surgical group, there was an evident, frequent (12/20 mice) involvement of the contralateral lobe of the thyroid, whereas this feature was only detected in 1/18 mice in the HFUS group. Statistical analysis showed the same pattern of growth in the two groups, and a significant hazard in the mice in the surgical group (p = 0.03). CONCLUSIONS This study demonstrated the technical feasibility of an HFUS-guided orthotopic mouse model of FTC. The HFUS-guided orthotopic model is easily reproducible and allows prolonged monitoring of the disease because the animals showed an increased survival rate.
Collapse
Affiliation(s)
- Adelaide Greco
- 1 Department of Advanced Biomedical Science, University of Naples Federico II , Naples, Italy
- 2 CEINGE Biotecnologie Avanzate , Naples, Italy
- 3 Institute of Biostructures and Bioimaging , CNR, Naples, Italy
| | - Sandra Albanese
- 1 Department of Advanced Biomedical Science, University of Naples Federico II , Naples, Italy
- 2 CEINGE Biotecnologie Avanzate , Naples, Italy
| | | | | | | | | | | | | | - Giuliana Salvatore
- 2 CEINGE Biotecnologie Avanzate , Naples, Italy
- 4 IRCCS SDN , Naples, Italy
- 6 Department of Motor Science and Wellness, Università Parthenope , Naples, Italy
| | - Andrea Soricelli
- 4 IRCCS SDN , Naples, Italy
- 6 Department of Motor Science and Wellness, Università Parthenope , Naples, Italy
| | | |
Collapse
|
19
|
Reeb AN, Ziegler A, Lin RY. Characterization of human follicular thyroid cancer cell lines in preclinical mouse models. Endocr Connect 2016; 5:47-54. [PMID: 26830329 PMCID: PMC5002955 DOI: 10.1530/ec-15-0114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/01/2016] [Indexed: 12/18/2022]
Abstract
Follicular thyroid cancer (FTC) is the second most common type of thyroid cancers. In order to develop more effective personalized therapies, it is necessary to thoroughly evaluate patient-derived cell lines in in vivo preclinical models before using them to test new, targeted therapies. This study evaluates the tumorigenic and metastatic potential of a panel of three human FTC cell lines (WRO, FTC-238, and TT1609-CO2) with defined genetic mutations in two in vivo murine models: an orthotopic thyroid cancer model to study tumor progression and a tail vein injection model to study metastasis. All cell lines developed tumors in the orthotopic model, with take rates of 100%. Notably, WRO-derived tumors grew two to four times faster than tumors arising from the FTC-238 and TT2609-CO2 cell lines. These results mirrored those of a tail vein injection model for lung metastasis: one hundred percent of mice injected with WRO cells in the tail vein exhibited aggressive growth of bilateral lung metastases within 35 days. In contrast, tail vein injection of FTC-238 or TT2609-CO2 cells did not result in lung metastasis. Together, our work demonstrates that these human FTC cell lines display highly varied tumorigenic and metastatic potential in vivo with WRO being the most aggressive cell line in both orthotopic and lung metastasis models. This information will be valuable when selecting cell lines for preclinical drug testing.
Collapse
Affiliation(s)
- Ashley N Reeb
- Department of Otolaryngology, Head and Neck SurgerySaint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Andrea Ziegler
- Department of Otolaryngology, Head and Neck SurgerySaint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Reigh-Yi Lin
- Department of Otolaryngology, Head and Neck SurgerySaint Louis University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
20
|
Sadow PM, Dias-Santagata D, Zheng Z, Lin DT, Le LP, Nucera C. Identification of insertions in PTEN and TP53 in anaplastic thyroid carcinoma with angiogenic brain metastasis. Endocr Relat Cancer 2015; 22:L23-8. [PMID: 26472209 PMCID: PMC4609031 DOI: 10.1530/erc-15-0198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Peter M Sadow
- Department of Pathology, WRN219, Harvard Medical School, Massachusetts General Hospital55 Fruit Street, Boston, Massachusetts, 02114USA Department of Pathology, WRN219, Harvard Medical School, Massachusetts General Hospital55 Fruit Street, Boston, Massachusetts, 02114USA
| | - Dora Dias-Santagata
- Department of Pathology, WRN219, Harvard Medical School, Massachusetts General Hospital55 Fruit Street, Boston, Massachusetts, 02114USA
| | - Zongli Zheng
- Department of Pathology, WRN219, Harvard Medical School, Massachusetts General Hospital55 Fruit Street, Boston, Massachusetts, 02114USA
| | - Derrick T Lin
- Department of Pathology, WRN219, Harvard Medical School, Massachusetts General Hospital55 Fruit Street, Boston, Massachusetts, 02114USA
| | - Long Phi Le
- Department of Pathology, WRN219, Harvard Medical School, Massachusetts General Hospital55 Fruit Street, Boston, Massachusetts, 02114USA
| | - Carmelo Nucera
- Department of Pathology, WRN219, Harvard Medical School, Massachusetts General Hospital55 Fruit Street, Boston, Massachusetts, 02114USA
| |
Collapse
|
21
|
Qasem E, Murugan AK, Al-Hindi H, Xing M, Almohanna M, Alswailem M, Alzahrani AS. TERT promoter mutations in thyroid cancer: a report from a Middle Eastern population. Endocr Relat Cancer 2015; 22:901-8. [PMID: 26354077 DOI: 10.1530/erc-15-0396] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/09/2015] [Indexed: 01/03/2023]
Abstract
Telomerase reverse transcriptase (TERT) promoter mutations C228T and C250T have recently been described in follicular cell-derived thyroid cancer (TC) in patients from North America and Europe. In this study, we explored whether these findings could be replicated in patients from a different ethnic group. We screened 17 benign thyroid adenomas and 265 TC samples from patients in the Middle East for these mutations by PCR and direct sequencing using DNA isolated from paraffin-embedded tumor tissues. None of the 17 benign adenomas harbored TERT promoter mutations. Of 265 TC, 34 (12.8%) harbored TERT promoter mutations, including 10/153 (6.5%) conventional papillary TC (CPTC), 8/57 (14.0%) follicular variant PTC, 9/30 (30%) tall cell variant PTC, 1/3 (30%) Hurthle cell thyroid cancer (HTC), 1/5 (20%) follicular TC, and 5/13 (38.5%) poorly differentiated TC. C250T mutation was present in only 6/265 (2.3%) cases, while C228T mutation was present in a total of 28/265 (10.6%) cases. These two mutations were mutually exclusive. TERT promoter mutations were significantly more common in older (≥45 years) than younger patients and were associated with larger tumour size, vascular invasion, higher TNM stage (stage III and IV), BRAF(V600E) mutation and persistent/recurrent disease at 6-12 months after initial treatment and at the last follow up. These associations were stronger in non-CPTC. Thus, this study on a large cohort of TC patients from Middle East demonstrates that TERT promoter mutations are relatively common, especially in the non-CPTC, and are associated with more aggressive histopathological features, BRAF(V600E) mutation, and disease persistence/recurrence than the WT TERT.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/ethnology
- Adenocarcinoma, Follicular/genetics
- Adenocarcinoma, Follicular/pathology
- Adenocarcinoma, Follicular/therapy
- Adenoma/ethnology
- Adenoma/genetics
- Adenoma/pathology
- Adenoma, Oxyphilic/ethnology
- Adenoma, Oxyphilic/genetics
- Adenoma, Oxyphilic/pathology
- Adenoma, Oxyphilic/therapy
- Adolescent
- Adult
- Age Factors
- Aged
- Arabs/genetics
- Carcinoma, Papillary/ethnology
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/pathology
- Carcinoma, Papillary/therapy
- Child
- Combined Modality Therapy
- DNA, Neoplasm/genetics
- Ethnicity/genetics
- Female
- Follow-Up Studies
- Humans
- Iodine Radioisotopes/therapeutic use
- Male
- Middle Aged
- Mutation, Missense
- Neoplasm Invasiveness
- Neoplasm Proteins/genetics
- Neoplasm Staging
- Point Mutation
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Proteins B-raf/genetics
- Recurrence
- Saudi Arabia/epidemiology
- Telomerase/genetics
- Thyroid Neoplasms/ethnology
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/therapy
- Thyroidectomy
- Treatment Outcome
- Tumor Burden
- Young Adult
Collapse
Affiliation(s)
- Ebtesam Qasem
- Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Avaniyapuram Kannan Murugan
- Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hindi Al-Hindi
- Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mingzhao Xing
- Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mai Almohanna
- Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Meshael Alswailem
- Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ali S Alzahrani
- Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA Department of Molecular OncologyKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of Pathology and Laboratory MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDepartment of MedicineKing Faisal Specialist Hospital and Research Centre, Riyadh, Saudi ArabiaDivision of Endocrinology and MetabolismJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Husain A, Hu N, Sadow PM, Nucera C. Expression of angiogenic switch, cachexia and inflammation factors at the crossroad in undifferentiated thyroid carcinoma with BRAF(V600E). Cancer Lett 2015; 380:577-585. [PMID: 26189429 DOI: 10.1016/j.canlet.2015.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 07/11/2015] [Accepted: 07/13/2015] [Indexed: 02/07/2023]
Abstract
Cachexia is the result of complex metabolic alterations which cause morbidity and mortality in patients with advanced cancers including undifferentiated (anaplastic) thyroid carcinoma (ATC). ATC is a lethal disease with limited therapeutic options and unclear etiology for cachexia. We hypothesize that the BRAF(V600E) oncoprotein triggers microvascular endothelial cell tubule formation (in vitro angiogenesis) by means of factors which play a crucial role in angiogenic switch, inflammation/immune response and cachexia. We use human ATC cells and applied multiplex ELISA assay to screen for and measure angiogenic/cachectic and pro-inflammatory factors in the ATC-derived secretome. We find that vemurafenib anti-BRAF(V600E) therapy significantly reduces secreted VEGFA, VEGFC and IL6 protein levels compared to vehicle-treated ATC cells. As a result, the secretome from vemurafenib-treated ATC cells inhibits microvascular endothelial cell-related in vitro angiogenesis. Furthermore, ATC clinical samples express VEGFA, VEGFC and IL6 proteins. Our results suggest that angiogenic/cachectic and pro-inflammatory/immune response factors could play a crucial role in BRAF(V600E)-positive human ATC aggressiveness. Understanding the extent to which microenvironment-associated angiogenic factors participate in cachexia and cancer metabolism in advanced thyroid cancers will reveal new biomarkers and foster novel therapeutic approaches.
Collapse
Affiliation(s)
- Amjad Husain
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Nina Hu
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Cancer Biology and Angiogenesis, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carmelo Nucera
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA; Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Cancer Biology and Angiogenesis, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Characterization of thyroid cancer cell lines in murine orthotopic and intracardiac metastasis models. Discov Oncol 2015; 6:87-99. [PMID: 25800363 DOI: 10.1007/s12672-015-0219-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/27/2015] [Indexed: 01/29/2023] Open
Abstract
Thyroid cancer incidence has been increasing over time, and it is estimated that ∼1950 advanced thyroid cancer patients will die of their disease in 2015. To combat this disease, an enhanced understanding of thyroid cancer development and progression as well as the development of efficacious, targeted therapies are needed. In vitro and in vivo studies utilizing thyroid cancer cell lines and animal models are critically important to these research efforts. In this report, we detail our studies with a panel of authenticated human anaplastic and papillary thyroid cancer (ATC and PTC) cell lines engineered to express firefly luciferase in two in vivo murine cancer models-an orthotopic thyroid cancer model as well as an intracardiac injection metastasis model. In these models, primary tumor growth in the orthotopic model and the establishment and growth of metastases in the intracardiac injection model are followed in vivo using an IVIS imaging system. In the orthotopic model, the ATC cell lines 8505C and T238 and the PTC cell lines K1/GLAG-66 and BCPAP had take rates >90 % with final tumor volumes ranging 84-214 mm(3) over 4-5 weeks. In the intracardiac model, metastasis establishment was successful in the ATC cell lines HTh74, HTh7, 8505C, THJ-16T, and Cal62 with take rates ≥70 %. Only one of the PTC cell lines tested (BCPAP) was successful in the intracardiac model with a take rate of 30 %. These data will be beneficial to inform the choice of cell line and model system for the design of future thyroid cancer studies.
Collapse
|
24
|
A versatile orthotopic nude mouse model for study of esophageal squamous cell carcinoma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:910715. [PMID: 25834829 PMCID: PMC4365327 DOI: 10.1155/2015/910715] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/20/2015] [Indexed: 12/29/2022]
Abstract
Increasing evidence indicates tumor-stromal interactions play a crucial role in cancer. An in vivo esophageal squamous cell carcinoma (ESCC) orthotopic animal model was developed with bioluminescence imaging established with a real-time monitoring platform for functional and signaling investigation of tumor-stromal interactions. The model was produced by injection of luciferase-labelled ESCC cells into the intraesophageal wall of nude mice. Histological examination indicates this orthotopic model is highly reproducible with 100% tumorigenesis among the four ESCC cell lines tested. This new model recapitulates many clinical and pathological properties of human ESCC, including esophageal luminal stricture by squamous cell carcinoma with nodular tumor growth, adventitia invasion, lymphovascular invasion, and perineural infiltration. It was tested using an AKT shRNA knockdown of ESCC cell lines and the in vivo tumor suppressive effects of AKT knockdown were observed. In conclusion, this ESCC orthotopic mouse model allows investigation of gene functions of cancer cells in a more natural tumor microenvironment and has advantages over previous established models. It provides a versatile platform with potential application for metastasis and therapeutic regimen testing.
Collapse
|
25
|
Smith N, Nucera C. Personalized therapy in patients with anaplastic thyroid cancer: targeting genetic and epigenetic alterations. J Clin Endocrinol Metab 2015; 100:35-42. [PMID: 25347569 PMCID: PMC4283016 DOI: 10.1210/jc.2014-2803] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Anaplastic thyroid cancer (ATC) is the most lethal of all thyroid cancers and one of the most aggressive human carcinomas. In the search for effective treatment options, research toward targeted, personalized therapies is proving to be a path with great potential. As we gain a deeper understanding of the genetic (eg, BRAF(V600E), PIK3CA, TP53, hTERT mutations, etc) and epigenetic (eg, histone methylation, histone de-acetylation, microRNA regulatory circuits, etc) alterations driving ATC, we are able to find targets when developing novel therapies to improve the lives of patients. Beyond development, we can look into the effectiveness of already approved targeted therapies (eg, anti-BRAF(V600E) selective inhibitors, tyrosine kinase inhibitors, histone deacetylase inhibitors, inhibitors of DNA methylation, etc) to potentially test in ATC after learning the molecular mechanisms that aid in tumor progression. DESIGN We performed a literature analysis in Medline through the PubMed web site for studies published between 2003 and 2014 using the following main keywords: anaplastic thyroid cancer, genetic and epigenetic alterations. OBJECTIVE Here, we outlined the common pathways that are altered in ATC, including the BRAF(V600E)/ERK1/2-MEK1/2 and PI3K-AKT pathways. We then examined the current research looking into personalized, potential targeted therapies in ATC, mentioning those that have been tentatively advanced into clinical trials and those with the potential to reach that stage. We also reviewed side effects of the current and potential targeted therapies used in patients with advanced thyroid cancer. CONCLUSIONS DNA and RNA next-generation sequencing analysis will be fundamental to unraveling a precise medicine and therapy in patients with ATC. Indeed, given the deep biological heterogeneity/complexity and high histological grade of this malignancy and its tumor microenvironment, personalized therapeutic approaches possibly based on the use of combinatorial targeted therapy will provide a rational approach when finding the optimal way to improve treatments for patients with ATC.
Collapse
Affiliation(s)
- Neal Smith
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Cancer Biology and Angiogenesis, Department of Pathology, Center for Vascular Biology Research, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
| | | |
Collapse
|
26
|
Borre PV, Gunda V, McFadden DG, Sadow PM, Varmeh S, Bernasconi M, Parangi S. Combined BRAF(V600E)- and SRC-inhibition induces apoptosis, evokes an immune response and reduces tumor growth in an immunocompetent orthotopic mouse model of anaplastic thyroid cancer. Oncotarget 2014; 5:3996-4010. [PMID: 24994118 PMCID: PMC4147301 DOI: 10.18632/oncotarget.2130] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/24/2014] [Indexed: 01/09/2023] Open
Abstract
Anaplastic (ATC) and refractory papillary thyroid cancer (PTC) lack effective treatments. Inhibition of either oncogenic BRAF or SRC has marked anti-tumor effects in mouse models of thyroid cancer, however, neither drug induces notable apoptosis. Here we report that the SRC-inhibitor dasatinib further sensitizes BRAFV600E-positive thyroid cancer cells to the BRAFV600E-inhibitor PLX4720. Combined treatment with PLX4720 and dasatinib synergistically inhibited proliferation and reduced migration in PTC and ATC cells. Whereas PLX4720 did not induce robust apoptosis in thyroid cancer cells, combined treatment with dasatinib induced apoptosis in 4 of 6 lines. In an immunocompetent orthotopic mouse model of ATC, combined PLX4720 and dasatinib treatment significantly reduced tumor volume relative to PLX4720 treatment alone. Immune cell infiltration was increased by PLX4720 treatment and this effect was maintained in mice treated with both PLX4720 and dasatinib. Further, combined treatment significantly increased caspase 3 cleavage in vivo relative to control or either treatment alone. In conclusion, combined PLX4720 and dasatinib treatment induces apoptosis, increases immune cell infiltration and reduces tumor volume in a preclinical model of ATC, suggesting that the combination of these FDA-approved drugs may have potential for the treatment of patients with ATC or refractory PTC.
Collapse
Affiliation(s)
- Pierre Vanden Borre
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Viswanath Gunda
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David G. McFadden
- Thyroid Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Peter M. Sadow
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shohreh Varmeh
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maria Bernasconi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Parenti R, Salvatorelli L, Magro G. Anaplastic Thyroid Carcinoma: Current Treatments and Potential New Therapeutic Options with Emphasis on TfR1/CD71. Int J Endocrinol 2014; 2014:685396. [PMID: 25097549 PMCID: PMC4102021 DOI: 10.1155/2014/685396] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/15/2014] [Accepted: 06/17/2014] [Indexed: 12/24/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is one of the most aggressive human cancers. Actually, ATC is refractory to conventional therapies, including surgery, chemotherapy, radiotherapy, and radioiodine ((131)I) therapy. Accordingly, genetic and molecular characterizations of ATC have been frequently and periodically reviewed in order to identify potential biological markers exploitable for target therapy. This review briefly focuses on main molecular events that characterize ATC and provides an update about preclinical studies. In addition, the overexpression of transferrin receptor 1 (TfR1/CD71) by neoplastic cells of ATC is emphasized in that it could represent a potential therapeutic target. In this regard, new therapeutic approaches based on the use of monoclonal or recombinant antibodies, or transferrin-gallium-TfR1/CD71 molecular complexes, or lastly small interfering RNAs (siRNAs) are proposed.
Collapse
Affiliation(s)
- Rosalba Parenti
- Department of Bio-Medical Sciences, Physiology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- *Rosalba Parenti:
| | - Lucia Salvatorelli
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
| | - Gaetano Magro
- Department G.F. Ingrassia, Section of Anatomic Pathology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
| |
Collapse
|