1
|
Cherifi F, Awada A. Molecular oncology of iodine refractory thyroid cancer current therapies and perspective. Crit Rev Oncol Hematol 2025; 209:104679. [PMID: 40043925 DOI: 10.1016/j.critrevonc.2025.104679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 04/20/2025] Open
Abstract
Thyroid cancer (TC) is the most common endocrine malignancy. Most patients will be treated and cured by surgery but a low percentage will develop advanced disease. The treatment of advanced disease is at first the use of radioiodine treatment in differentiated cancer then at progression will rely on molecular alterations and consequently in targeted treatments. In this review, we will explore the most frequent molecular alterations of each histological subtype: differentiated thyroid cancer (DTC), anaplastic thyroid cancer (ATC), medullary thyroid cancers (MTC) and clinically tested and approved treatment. We will also report the clinical and preclinical perspective in this field.
Collapse
Affiliation(s)
- François Cherifi
- Oncology Medicine department, François Baclesse Center, Caen, France.
| | - Ahmad Awada
- Oncology Medicine Department of Chirec Cancer Institute, Université Libre de Bruxelles Brussels, Belgium
| |
Collapse
|
2
|
Cortas C, Charalambous H. Tyrosine Kinase Inhibitors for Radioactive Iodine Refractory Differentiated Thyroid Cancer. Life (Basel) 2023; 14:22. [PMID: 38255638 PMCID: PMC10817256 DOI: 10.3390/life14010022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/24/2024] Open
Abstract
Patients with differentiated thyroid cancer usually present with early-stage disease and undergo surgery followed by adjuvant radioactive iodine ablation, resulting in excellent clinical outcomes and prognosis. However, a minority of patients relapse with metastatic disease, and eventually develop radioactive iodine refractory disease (RAIR). In the past there were limited and ineffective options for systemic therapy for RAIR, but over the last ten to fifteen years the emergence of tyrosine kinase inhibitors (TKIs) has provided important new avenues of treatment for these patients, that are the focus of this review. Currently, Lenvatinib and Sorafenib, multitargeted TKIs, represent the standard first-line systemic treatment options for RAIR thyroid carcinoma, while Cabozantinib is the standard second-line treatment option. Furthermore, targeted therapies for patients with specific targetable molecular abnormalities include Latrectinib or Entrectinib for patients with NTRK gene fusions and Selpercatinib or Pralsetinib for patients with RET gene fusions. Dabrafenib plus Trametinib currently only have tumor agnostic approval in the USA for patients with BRAF V600E mutations, including thyroid cancer. Redifferentiation therapy is an area of active research, with promising initial results, while immunotherapy studies with checkpoint inhibitors in combination with tyrosine kinase inhibitors are underway.
Collapse
Affiliation(s)
| | - Haris Charalambous
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia 2006, Cyprus;
| |
Collapse
|
3
|
Martins RS, Jesus TT, Cardoso L, Soares P, Vinagre J. Personalized Medicine in Medullary Thyroid Carcinoma: A Broad Review of Emerging Treatments. J Pers Med 2023; 13:1132. [PMID: 37511745 PMCID: PMC10381735 DOI: 10.3390/jpm13071132] [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: 05/25/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Medullary thyroid carcinoma (MTC) arises from parafollicular cells in the thyroid gland, and although rare, it represents an aggressive type of thyroid cancer. MTC is recognized for its low mutational burden, with point mutations in RET or RAS genes being the most common oncogenic events. MTC can be resistant to cytotoxic chemotherapy, and multitarget kinase inhibitors (MKIs) have been considered a treatment option. They act by inhibiting the activities of specific tyrosine kinase receptors involved in tumor growth and angiogenesis. Several tyrosine kinase inhibitors are approved in the treatment of advanced MTC, including vandetanib and cabozantinib. However, due to the significant number of adverse events, debatable efficiency and resistance, there is a need for novel RET-specific TKIs. Newer RET-specific TKIs are expected to overcome previous limitations and improve patient outcomes. Herein, we aim to review MTC signaling pathways, the most recent options for treatment and the applications for personalized medicine.
Collapse
Affiliation(s)
- Rui Sousa Martins
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Ciências da Universidade do Porto (FCUP), 4169-007 Porto, Portugal
| | - Tito Teles Jesus
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Luís Cardoso
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Departamento de Endocrinologia, Diabetes e Metabolismo do Centro Hospitalar Universitário de Coimbra, 3000-075 Coimbra, Portugal
| | - Paula Soares
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - João Vinagre
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| |
Collapse
|
4
|
Derwich A, Sykutera M, Bromińska B, Rubiś B, Ruchała M, Sawicka-Gutaj N. The Role of Activation of PI3K/AKT/mTOR and RAF/MEK/ERK Pathways in Aggressive Pituitary Adenomas-New Potential Therapeutic Approach-A Systematic Review. Int J Mol Sci 2023; 24:10952. [PMID: 37446128 PMCID: PMC10341524 DOI: 10.3390/ijms241310952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Pituitary tumors (PT) are mostly benign, although occasionally they demonstrate aggressive behavior, invasion of surrounding tissues, rapid growth, resistance to conventional treatments, and multiple recurrences. The pathogenesis of PT is still not fully understood, and the factors responsible for its invasiveness, aggressiveness, and potential for metastasis are unknown. RAF/MEK/ERK and mTOR signaling are significant pathways in the regulation of cell growth, proliferation, and survival, its importance in tumorigenesis has been highlighted. The aim of our review is to determine the role of the activation of PI3K/AKT/mTOR and RAF/MEK/ERK pathways in the pathogenesis of pituitary tumors. Additionally, we evaluate their potential in a new therapeutic approach to provide alternative therapies and improved outcomes for patients with aggressive pituitary tumors that do not respond to standard treatment. We perform a systematic literature search using the PubMed, Embase, and Scopus databases (search date was 2012-2023). Out of the 529 screened studies, 13 met the inclusion criteria, 7 related to the PI3K/AKT/mTOR pathway, and 7 to the RAF/MEK/ERK pathway (one study was used in both analyses). Understanding the specific factors involved in PT tumorigenesis provides opportunities for targeted therapies. We also review the possible new targeted therapies and the use of mTOR inhibitors and TKI in PT management. Although the RAF/MEK/ERK and PI3K/AKT/mTOR pathways play a pivotal role in the complex signaling network along with many interactions, further research is urgently needed to clarify the exact functions and the underlying mechanisms of these signaling pathways in the pathogenesis of pituitary adenomas and their role in its invasiveness and aggressive clinical outcome.
Collapse
Affiliation(s)
- Aleksandra Derwich
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Monika Sykutera
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Barbara Bromińska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Nadia Sawicka-Gutaj
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| |
Collapse
|
5
|
Clinical Implications of mTOR Expression in Papillary Thyroid Cancer—A Systematic Review. Cancers (Basel) 2023; 15:cancers15061665. [PMID: 36980552 PMCID: PMC10046096 DOI: 10.3390/cancers15061665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Papillary thyroid cancer (PTC) comprises approximately 80% of all thyroid malignancies. Although several etiological factors, such as age, gender, and irradiation, are already known to be involved in the development of PTC, the genetics of cancerogenesis remain undetermined. The mTOR pathway regulates several cellular processes that are critical for tumorigenesis. Activated mTOR is involved in the development and progression of PTC. Therefore, we performed a systematic review of papers studying the expression of the mTOR gene and protein and its relationship with PTC risk and clinical outcome. A systematic literature search was performed using PubMed, Embase, and Scopus databases (the search date was 2012–2022). Studies investigating the expression of mTOR in the peripheral blood or tissue of patients with PTC were deemed eligible for inclusion. Seven of the 286 screened studies met the inclusion criteria for mTOR gene expression and four for mTOR protein expression. We also analyzed the data on mTOR protein expression in PTC. We analyzed the association of mTOR expression with papillary thyroid cancer clinicopathological features, such as the TNM stage, BRAF V600E mutation, sex distribution, lymph node and distant metastases, and survival prognosis. Understanding specific factors involved in PTC tumorigenesis provides opportunities for targeted therapies. We also reviewed the possible new targeted therapies and the use of mTOR inhibitors in PTC. This topic requires further research with novel techniques to translate the achieved results to clinical application.
Collapse
|
6
|
Cao Z, Zhang Z, Tang X, Liu R, Wu M, Wu J, Liu Z. Comprehensive analysis of tissue proteomics in patients with papillary thyroid microcarcinoma uncovers the underlying mechanism of lymph node metastasis and its significant sex disparities. Front Oncol 2022; 12:887977. [PMID: 36106120 PMCID: PMC9465038 DOI: 10.3389/fonc.2022.887977] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Background Lymph node metastasis (LNM) in papillary thyroid microcarcinoma (PTMC) is associated with an increased risk of recurrence and poor prognosis. Sex has been regarded as a critical risk factor for LNM. The present study aimed to investigate the molecular mechanisms underlying LNM and its significant sex disparities in PTMC development. Methods A direct data-independent acquisition (DIA) proteomics approach was used to identify differentially expressed proteins (DEPs) in PTMC tumorous tissues with or without LNM and from male and female patients with LNM. The functional annotation of DEPs was performed using bioinformatics methods. Furthermore, The Cancer Genome Atlas Thyroid Carcinoma (TCGA-THCA) dataset and immunohistochemistry (IHC) were used to validate selected DEPs. Results The proteomics profile in PTMC with LNM differed from that of PTMC without LNM. The metastasis-related DEPs were primarily enriched in categories associated with mitochondrial dysfunction and may promote tumor progression by activating oxidative phosphorylation and PI3K/AKT signaling pathways. Comparative analyses of these DEPs revealed downregulated expression of specific proteins with well-established links to tumor metastasis, such as SLC25A15, DIRAS2, PLA2R1, and MTARC1. Additionally, the proteomics profiles of male and female PTMC patients with LNM were dramatically distinguishable. An elevated level of ECM-associated proteins might be related to more LNM in male PTMC than in female PTMC patients. The upregulated expression levels of MMRN2 and NID2 correlated with sex disparities and showed a positive relationship with unfavorable variables, such as LNMs and poor prognosis. Conclusions The proteomics profiles of PTMC show significant differences associated with LNM and its sex disparities, which further expands our understanding of the functional networks and signaling pathways related to PTMC with LNM.
Collapse
Affiliation(s)
- Zhen Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zejian Zhang
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyue Tang
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Wu
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Jianqiang Wu, ; Ziwen Liu,
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Jianqiang Wu, ; Ziwen Liu,
| |
Collapse
|
7
|
Thyroid Carcinoma: Phenotypic Features, Underlying Biology and Potential Relevance for Targeting Therapy. Int J Mol Sci 2021; 22:ijms22041950. [PMID: 33669363 PMCID: PMC7920269 DOI: 10.3390/ijms22041950] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022] Open
Abstract
Thyroid carcinoma consists a group of phenotypically heterogeneous cancers. Recent advances in biological technologies have been advancing the delineation of genetic, epigenetic, and non-genetic factors that contribute to the heterogeneities of these cancers. In this review article, we discuss new findings that are greatly improving the understanding of thyroid cancer biology and facilitating the identification of novel targets for therapeutic intervention. We review the phenotypic features of different subtypes of thyroid cancers and their underlying biology. We discuss recent discoveries in thyroid cancer heterogeneities and the critical mechanisms contributing to the heterogeneity with emphases on genetic and epigenetic factors, cancer stemness traits, and tumor microenvironments. We also discuss the potential relevance of the intratumor heterogeneity in understanding therapeutic resistance and how new findings in tumor biology can facilitate designing novel targeting therapies for thyroid cancer.
Collapse
|
8
|
Maruei‐Milan R, Saravani M, Heidari Z, Asadi‐Tarani M, Salimi S. Effects of the
MTOR
and
AKT1
genes polymorphisms on papillary thyroid cancer development. IUBMB Life 2020; 72:2601-2610. [DOI: 10.1002/iub.2388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/01/2020] [Accepted: 09/13/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Rostam Maruei‐Milan
- Department of Clinical Biochemistry, School of Medicine Zahedan University of Medical Sciences Zahedan Iran
| | - Mohsen Saravani
- Department of Clinical Biochemistry, School of Medicine Zahedan University of Medical Sciences Zahedan Iran
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute Zahedan University of Medical Sciences Zahedan Iran
| | - Zahra Heidari
- Department of Internal Medicine Zahedan University of Medical Sciences Zahedan Iran
| | - Mina Asadi‐Tarani
- Department of Clinical Biochemistry, School of Medicine Zahedan University of Medical Sciences Zahedan Iran
| | - Saeedeh Salimi
- Department of Clinical Biochemistry, School of Medicine Zahedan University of Medical Sciences Zahedan Iran
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute Zahedan University of Medical Sciences Zahedan Iran
| |
Collapse
|
9
|
Ozdemir Kutbay N, Biray Avci C, Sarer Yurekli B, Caliskan Kurt C, Shademan B, Gunduz C, Erdogan M. Effects of metformin and pioglitazone combination on apoptosis and AMPK/mTOR signaling pathway in human anaplastic thyroid cancer cells. J Biochem Mol Toxicol 2020; 34:e22547. [PMID: 32589349 DOI: 10.1002/jbt.22547] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/28/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
Anaplastic cancer constitutes 1% of thyroid cancers, and it is one of the most aggressive cancers. Treatment options are external radiation therapy and/or chemotherapy. The success rate with these treatment modalities is not satisfactory. We aimed to evaluate the effects of metformin (MET) and pioglitazone (PIO) combination on apoptosis and AMP-activated protein kinase/mammalian target of rapamycin (mTOR) signaling pathway in human anaplastic thyroid cancer cells. In this study, we evaluated the effects of MET and PIO individually and the combination of the two drugs on the cellular lines SW1736 and C643 ATC. Genes contained in the mTOR signaling pathway were examined using human mTOR Signalization RT2 Profiler PCR Array. In C643 and SW1736 cell lines, IC50 doses of MET and PIO were found out as 17.69 mM, 11.64 mM, 27.12 µM, and 23.17 µM. Also, the combination of MET and PIO was determined as an additive according to isobologram analyses. We have found the downregulation of the expression levels of oncogenic genes: AKT3, CHUK, CDC42, EIF4E, HIF1A, IKBKB, ILK, MTOR, PIK3CA, PIK3CG, PLD1, PRKCA, and RICTOR genes, in the MET and PIO combination-treated cells. In addition, expression levels of tumor suppressor genes, DDIT4, DDIT4L, EIF4EBP1, EIF4EBP2, FKBP1A, FKBP8, GSK3B, MYO1C, PTEN, ULK1, and ULK2, were found to have increased significantly. The MET + PIO combination was first applied to thyroid cancer cells, and significant reductions in the level of oncogenic genes were detected. The decreases, particularly, in AKT3, DEPTOR, EIF4E, ILK, MTOR, PIK3C, and PRKCA expressions indicate that progression can be prevented in thyroid cancer cells and these genes could be selected as therapeutic targets.
Collapse
Affiliation(s)
- Nilufer Ozdemir Kutbay
- Department of Endocrinology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Cigir Biray Avci
- Department of Medical Biology, Ege University Medical School, Izmir, Turkey
| | - Banu Sarer Yurekli
- Department of Endocrinology, Ege University Medical School, Izmir, Turkey
| | | | - Behrouz Shademan
- Department of Medical Biology, Ege University Medical School, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Ege University Medical School, Izmir, Turkey
| | - Mehmet Erdogan
- Department of Endocrinology, Ege University Medical School, Izmir, Turkey
| |
Collapse
|
10
|
Yang X, Liu G, Li W, Zang L, Li D, Wang Q, Yu F, Xiang X. Silencing of zinc finger protein 703 inhibits medullary thyroid carcinoma cell proliferation in vitro and in vivo. Oncol Lett 2019; 19:943-951. [PMID: 31897207 PMCID: PMC6924197 DOI: 10.3892/ol.2019.11153] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 10/18/2019] [Indexed: 12/19/2022] Open
Abstract
Zinc finger protein 703 (ZNF703) is a new member of the zinc finger protein family of transcription factors that plays an important role during embryogenesis in metazoans. The overexpression of ZNF703 contributes to tumorigenesis and progression of a number of malignancies by activating the Akt/mammalian target of rapamycin (mTOR) signaling pathway. This pathway is activated in medullary thyroid cancer (MTC), but its mechanism of action is not yet fully understood. The aim of the present study was to examine the role of ZNF703 and its association with Akt/mTOR activation in MTC. The present study used the phosphorylation of Akt1 protein at serine 473 (pAkt473) as an indicator of signaling activation. Immunohistochemistry (IHC) staining and western blot analyses were performed in order to examine the expression of ZNF703 in 34 cases of MTC and 12 cases of corresponding normal thyroid tissues. ZNF703 expression in MTC was significantly higher compared with the corresponding normal thyroid tissues (P<0.05). Furthermore, expression of ZNF703 was associated with tumor size, lymph node metastasis and advanced stage of disease. IHC also demonstrated that the level of ZNF703 was positively correlated with p-Akt473 in the 34 cases of MTC. The human MTC cell line TT was selected for further investigation as TT cells exhibit Akt/mTOR activation. The biological effects of silencing ZNF703 in TT cells on proliferation and apoptosis, both in vitro and in vivo were investigated in the present study. ZNF703 silencing inhibited the proliferation of TT cells in vitro and inhibited xenograft tumor growth in vivo. These effects were accompanied by the substantial decrease of pAkt473 and the induction of p53 protein. These results demonstrate that ZNF703 may play a relevant role in MTC due to its association with the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaolin Yang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Geling Liu
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Weijuan Li
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Luyang Zang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Ding Li
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Qianqian Wang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Fang Yu
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Xiuxiu Xiang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
11
|
Kraft IL, Akshintala S, Zhu Y, Lei H, Derse-Anthony C, Dombi E, Steinberg SM, Lodish M, Waguespack SG, Kapustina O, Fox E, Balis FM, Merino MJ, Meltzer PS, Glod JW, Shern JF, Widemann BC. Outcomes of Children and Adolescents with Advanced Hereditary Medullary Thyroid Carcinoma Treated with Vandetanib. Clin Cancer Res 2018; 24:753-765. [PMID: 29187393 PMCID: PMC5815946 DOI: 10.1158/1078-0432.ccr-17-2101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/04/2017] [Accepted: 11/20/2017] [Indexed: 12/31/2022]
Abstract
Purpose: Vandetanib is well-tolerated in patients with advanced medullary thyroid carcinoma (MTC). Long-term outcomes and mechanisms of MTC progression have not been reported previously.Experimental Design: We monitored toxicities and disease status in patients taking vandetanib for hereditary, advanced MTC. Tumor samples were analyzed for molecular mechanisms of disease progression.Results: Seventeen patients [8 male, age 13 (9-17)* years] enrolled; 16 had a RET p.Met918Thr germline mutation. The duration of vandetanib therapy was 6.1 (0.1-9.7+)* years with treatment ongoing in 9 patients. Best response was partial response in 10, stable disease in 6, and progressive disease in one patient. Duration of response was 7.4 (0.6-8.7+)* and 4.9 (0.6-7.8+)* years in patients with PR and SD, respectively. Six patients died 2.0 (0.4-5.7)* years after progression. Median progression-free survival (PFS) was 6.7 years [95% confidence interval (CI): 2.3 years-undefined] and 5-year overall survival (OS) was 88.2% (95% CI: 60.6%-96.9%). Of 16 patients with a RET p.Met918Thr mutation, progression-free survival was 6.7 years (95% CI: 3.1-undefined) and 5-year overall survival was 93.8% (95% CI: 63.2%-99.1%). No patients terminated treatment because of toxicity. DNA sequencing of tissue samples (n = 11) identified an increase in copy number alterations across the genome as a potential mechanism of drug resistance [*median (range)].Conclusions: This study demonstrates that vandetanib is safe and results in sustained responses in children and adolescents with hereditary MTC. Our preliminary molecular data suggest that an increase in copy number abnormalities may be associated with tumor progression in hereditary MTC patients treated with vandetanib. Clin Cancer Res; 24(4); 753-65. ©2017 AACR.
Collapse
Affiliation(s)
- Ira L Kraft
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Srivandana Akshintala
- Department of Pediatrics, New York University Langone Medical Center, New York, New York
| | - Yuelin Zhu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Claudia Derse-Anthony
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., NCI Campus at Frederick, Frederick, Maryland
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Maya Lodish
- Developmental Endocrine Oncology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, Maryland
| | - Steven G Waguespack
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Elizabeth Fox
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Frank M Balis
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Maria J Merino
- Translational Surgical Pathology Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - John W Glod
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Jack F Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Brigitte C Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
12
|
Nozhat Z, Hedayati M, Pourhassan H. Signaling pathways in medullary thyroid carcinoma: therapeutic implications. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016. [DOI: 10.2217/ije-2016-0014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Medullary thyroid cancer (MTC) is the third most frequent thyroid cancer arising from thyroid parafollicular cells. Surgery is the first-line strategy in treatment of MTC but disease relapse and patient's death have been observed in approximately two out of three of MTC cases. Identification of molecular mechanisms and different signaling pathways has offered new insights for disease treatment. The development of tyrosine kinase inhibitors targeting these pathways has provided a promising landscape for prevention of progression in patients with advanced metastatic MTC. In this review article different altered molecular pathways implicated in the development of MTC and the therapeutic strategies based on targeting the identified signaling pathways have been summarized.
Collapse
Affiliation(s)
- Zahra Nozhat
- Cellular & Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Biotechnology Department, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular & Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hoda Pourhassan
- Clinical Instructor Faculty, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| |
Collapse
|
13
|
Asimomytis A, Karanikou M, Rodolakis A, Vaiopoulou A, Tsetsa P, Creatsas G, Stefos T, Antsaklis A, Patsouris E, Rassidakis GZ. mTOR downstream effectors, 4EBP1 and eIF4E, are overexpressed and associated with HPV status in precancerous lesions and carcinomas of the uterine cervix. Oncol Lett 2016; 12:3234-3240. [PMID: 27899988 PMCID: PMC5103924 DOI: 10.3892/ol.2016.5056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/07/2016] [Indexed: 11/17/2022] Open
Abstract
The present study aims to investigate the expression levels of two critical mammalian target of rapamycin (mTOR) downstream effectors, 4E binding protein 1 (4EBP1) and eukaryotic initiation factor 4E (eIF4E) proteins, in precancerous squamous intraepithelial lesions and cancer of the uterine cervix, and their association with human papilloma virus (HPV) infection status. Uterine cervical biopsies from 73 patients were obtained, including 40 fresh-frozen samples and 42 archival formalin-fixed, paraffin-embedded tissue specimens. Whole protein extracts were analyzed for the expression of 4EBP1 and eIF4E proteins using western blotting. In addition, distribution of 4EBP1 and eIF4E protein expression and 4EBP1 phosphorylation (P-4EBP1) were analyzed by immunohistochemistry in archival tissues and correlated with the degree of dysplasia. The presence of high-risk HPV (HR-HPV) types was assessed by polymerase chain reaction. Using western blot analysis, high expression levels of 4EBP1 and eIF4E were observed in all uterine cervical carcinomas, which significantly correlated with the degree of dysplasia. By immunohistochemistry, overexpression of 4EBP1 and eIF4E was detected in 20 of 21 (95%) and 17 of 21 (81%) samples, respectively, in patients with high-grade dysplasia and carcinomas, compared with 1 of 20 (5%) and 2 of 20 (10%) samples, respectively, in patients with low-grade lesions or normal histology. All 4EBP1-positive cases tested were also positive for P-4EBP1. Furthermore, overexpression of 4EBP1 and eIF4E significantly correlated with the presence of HR-HPV oncogenic types. The present study demonstrated that critical effectors of mTOR signaling, which control protein synthesis initiation, are overexpressed in cervical high-grade dysplasia and cancer, and their levels correlate with oncogenic HPV types. These findings may provide novel targets for investigational therapeutic approaches in patients with cancer of the uterine cervix.
Collapse
Affiliation(s)
- Aristidis Asimomytis
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - Maria Karanikou
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - Alexander Rodolakis
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Maternity Hospital, GR-11528 Athens, Greece
| | - Anna Vaiopoulou
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - Paraskevi Tsetsa
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Maternity Hospital, GR-11528 Athens, Greece
| | - George Creatsas
- Second Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Aretaieion Hospital, GR-11528 Athens, Greece
| | - Theodoros Stefos
- Department of Obstetrics and Gynecology, University of Ioannina, School of Medicine, GR-45110 Ioannina, Greece
| | - Aristidis Antsaklis
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Maternity Hospital, GR-11528 Athens, Greece
| | - Efstratios Patsouris
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece
| | - George Z Rassidakis
- First Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, GR-11527 Athens, Greece; Department of Pathology and Cytology, Karolinska University Hospital and Karolinska Institute, Solna 17176, Sweden
| |
Collapse
|
14
|
Craps J, Joris V, De Jongh B, Sonveaux P, Horman S, Lengelé B, Bertrand L, Many MC, Colin IM, Gérard AC. Involvement of mTOR and Regulation by AMPK in Early Iodine Deficiency-Induced Thyroid Microvascular Activation. Endocrinology 2016; 157:2545-59. [PMID: 27035650 DOI: 10.1210/en.2015-1911] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Iodine deficiency (ID) induces TSH-independent microvascular activation in the thyroid via the reactive oxygen species/nitric oxide-hypoxia-inducible factor-1α/vascular endothelial growth factor (VEGF) pathway. We hypothesized the additional involvement of mammalian target of rapamycin (mTOR) as a positive regulator of this pathway and AMP-activated protein kinase (AMPK) as a negative feedback regulator to explain the transient nature of ID-induced microvascular changes under nonmalignant conditions. mTOR and AMPK involvement was investigated using an in vitro model (human thyrocytes in primary cultures) and 2 murine models of goitrogenesis (normal NMRI and RET-PTC mice [a papillary thyroid cancer model]). In NMRI mice, ID had no effect on the phosphorylation of ribosomal S6 kinase (p70S6K), a downstream target of mTOR. However, rapamycin inhibited ID-induced thyroid blood flow and VEGF protein expression. In the RET-PTC model, ID strongly increased the phosphorylation of p70S6K, whereas rapamycin completely inhibited the ID-induced increase in p70S6K phosphorylation, thyroid blood flow, and VEGF-A expression. In vitro, although ID increased p70S6K phosphorylation, the ID-stimulated hypoxia-inducible factor/VEGF pathway was inhibited by rapamycin. Activation of AMPK by metformin inhibited ID effects both in vivo and in vitro. In AMPK-α1 knockout mice, the ID-induced increase in thyroid blood flow and VEGF-A protein expression persisted throughout the treatment, whereas both parameters returned to control values in wild-type mice after 4 days of ID. In conclusion, mTOR is required for early ID-induced thyroid microvascular activation. AMPK negatively regulates this pathway, which may account for the transient nature of ID-induced TSH-independent vascular effects under benign conditions.
Collapse
Affiliation(s)
- J Craps
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - V Joris
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - B De Jongh
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - P Sonveaux
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - S Horman
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - B Lengelé
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - L Bertrand
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - M-C Many
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - I M Colin
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - A-C Gérard
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| |
Collapse
|
15
|
Eukaryotic initiation factor 4E-binding protein 1 (4E-BP1): a master regulator of mRNA translation involved in tumorigenesis. Oncogene 2016; 35:4675-88. [DOI: 10.1038/onc.2015.515] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/11/2015] [Accepted: 12/11/2015] [Indexed: 01/17/2023]
|
16
|
Friday AJ, Henderson MA, Morrison JK, Hoffman JL, Keiper BD. Spatial and temporal translational control of germ cell mRNAs mediated by the eIF4E isoform IFE-1. J Cell Sci 2015; 128:4487-98. [PMID: 26542024 DOI: 10.1242/jcs.172684] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 11/02/2015] [Indexed: 11/20/2022] Open
Abstract
Regulated mRNA translation is vital for germ cells to produce new proteins in the spatial and temporal patterns that drive gamete development. Translational control involves the de-repression of stored mRNAs and their recruitment by eukaryotic initiation factors (eIFs) to ribosomes. C. elegans expresses five eIF4Es (IFE-1-IFE-5); several have been shown to selectively recruit unique pools of mRNA. Individual IFE knockouts yield unique phenotypes due to inefficient translation of certain mRNAs. Here, we identified mRNAs preferentially translated through the germline-specific eIF4E isoform IFE-1. Differential polysome microarray analysis identified 77 mRNAs recruited by IFE-1. Among the IFE-1-dependent mRNAs are several required for late germ cell differentiation and maturation. Polysome association of gld-1, vab-1, vpr-1, rab-7 and rnp-3 mRNAs relies on IFE-1. Live animal imaging showed IFE-1-dependent selectivity in spatial and temporal translation of germline mRNAs. Altered MAPK activation in oocytes suggests dual roles for IFE-1, both promoting and suppressing oocyte maturation at different stages. This single eIF4E isoform exerts positive, selective translational control during germ cell differentiation.
Collapse
Affiliation(s)
- Andrew J Friday
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Melissa A Henderson
- Department of Molecular Sciences, DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
| | - J Kaitlin Morrison
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Jenna L Hoffman
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Brett D Keiper
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
17
|
Positive mRNA Translational Control in Germ Cells by Initiation Factor Selectivity. BIOMED RESEARCH INTERNATIONAL 2015; 2015:327963. [PMID: 26357652 PMCID: PMC4556832 DOI: 10.1155/2015/327963] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/22/2015] [Indexed: 01/04/2023]
Abstract
Ultimately, the production of new proteins in undetermined cells pushes them to new fates. Other proteins hold a stem cell in a mode of self-renewal. In germ cells, these decision-making proteins are produced largely from translational control of preexisting mRNAs. To date, all of the regulation has been attributed to RNA binding proteins (RBPs) that repress mRNAs in many models of germ cell development (Drosophila, mouse, C. elegans, and Xenopus). In this review, we focus on the selective, positive function of translation initiation factors eIF4E and eIF4G, which recruit mRNAs to ribosomes upon derepression. Evidence now shows that the two events are not separate but rather are coordinated through composite complexes of repressors and germ cell isoforms of eIF4 factors. Strikingly, the initiation factor isoforms are themselves mRNA selective. The mRNP complexes of translation factors and RBPs are built on specific populations of mRNAs to prime them for subsequent translation initiation. Simple rearrangement of the partners causes a dormant mRNP to become synthetically active in germ cells when and where they are required to support gametogenesis.
Collapse
|
18
|
Yi YW, You K, Bae EJ, Kwak SJ, Seong YS, Bae I. Dual inhibition of EGFR and MET induces synthetic lethality in triple-negative breast cancer cells through downregulation of ribosomal protein S6. Int J Oncol 2015; 47:122-32. [PMID: 25955731 PMCID: PMC4735702 DOI: 10.3892/ijo.2015.2982] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/09/2015] [Indexed: 12/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) exhibits innate resistance to the EGFR inhibition despite high level expression of EGFR. Recently, we found that the proliferation of basal-like (BL) subtype TNBC cells is synergistically inhibited by combination of EGFR and PI3K/AKT inhibitors. On the contrary, TNBC cells of mesenchymal stem-like (MSL) subtype are resistant to these combinations. To identify potential synthetic lethal interaction of compounds for treatment of MSL subtype TNBC cells, we performed MTT screening of MDA-MB-231 cells with a small library of receptor tyrosine kinase inhibitors (RTKIs) in the presence of gefitinib, an EGFR inhibitor. We identified MET inhibitors as potent RTKIs that caused synthetic lethality in combination with gefitinib in MDA-MB-231 cells. We demonstrated that combination of a MET inhibitor SU11274 with various EGFR inhibitors resulted in synergistic suppression of cell viability (in MTT assay) and cell survival (in colony formation assay) of MSL subtype TNBC cells. We further demonstrated that SU11274 alone induced G2 arrest and gefitinib/SU11274 combination sustained the SU11274-induced G2 arrest in these cells. In addition, SU11274/gefitinib combination synergistically reduced the level of ribosomal protein S6 (RPS6) in MSL subtype TNBC cells. In addition, knockdown of RPS6 itself, in both HS578T and MDA-MB-231, markedly reduced the proliferation of these cells. Taken together, our data suggest that dual targeting of EGFR and MET inhibits the proliferation of MSL subtype TNBC cells through down-regulation of RPS6.
Collapse
Affiliation(s)
- Yong Weon Yi
- Department of Nanobiomedical Science and BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Kyusic You
- Department of Nanobiomedical Science and BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Edward Jeong Bae
- Department of Nanobiomedical Science and BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Sahng-June Kwak
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Yeon-Sun Seong
- Department of Nanobiomedical Science and BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Insoo Bae
- Department of Nanobiomedical Science and BK21 PLUS Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
19
|
Fallahi P, Mazzi V, Vita R, Ferrari SM, Materazzi G, Galleri D, Benvenga S, Miccoli P, Antonelli A. New therapies for dedifferentiated papillary thyroid cancer. Int J Mol Sci 2015; 16:6153-82. [PMID: 25789503 PMCID: PMC4394525 DOI: 10.3390/ijms16036153] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/14/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022] Open
Abstract
The number of thyroid cancers is increasing. Standard treatment usually includes primary surgery, thyroid-stimulating hormone suppressive therapy, and ablation of the thyroid remnant with radioactive iodine (RAI). Despite the generally good prognosis of thyroid carcinoma, about 5% of patients will develop metastatic disease, which fails to respond to RAI, exhibiting a more aggressive behavior. The lack of specific, effective and well-tolerated drugs, the scarcity of data about the association of multi-targeting drugs, and the limited role of radioiodine for dedifferentiated thyroid cancer, call for further efforts in the field of new drugs development. Rearranged during transfection (RET)/papillary thyroid carcinoma gene rearrangements, BRAF (B-RAF proto-oncogene, serine/threonine kinase) gene mutations, RAS (rat sarcoma) mutations, and vascular endothelial growth factor receptor 2 angiogenesis pathways are some of the known pathways playing a crucial role in the development of thyroid cancer. Targeted novel compounds have been demonstrated to induce clinical responses and stabilization of disease. Sorafenib has been approved for differentiated thyroid cancer refractory to RAI.
Collapse
Affiliation(s)
- Poupak Fallahi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi, 10, 56126 Pisa, Italy.
| | - Valeria Mazzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi, 10, 56126 Pisa, Italy.
| | - Roberto Vita
- Department of Clinical & Experimental Medicine, Section of Endocrinology, University of Messina, Piazza Pugliatti, 1, 98122 Messina, Italy.
| | - Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi, 10, 56126 Pisa, Italy.
| | - Gabriele Materazzi
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi, 10, 56126 Pisa, Italy.
| | - David Galleri
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi, 10, 56126 Pisa, Italy.
| | - Salvatore Benvenga
- Department of Clinical & Experimental Medicine, Section of Endocrinology, University of Messina, Piazza Pugliatti, 1, 98122 Messina, Italy.
| | - Paolo Miccoli
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi, 10, 56126 Pisa, Italy.
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi, 10, 56126 Pisa, Italy.
| |
Collapse
|
20
|
Manfredi GI, Dicitore A, Gaudenzi G, Caraglia M, Persani L, Vitale G. PI3K/Akt/mTOR signaling in medullary thyroid cancer: a promising molecular target for cancer therapy. Endocrine 2015; 48:363-70. [PMID: 25115638 DOI: 10.1007/s12020-014-0380-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 08/01/2014] [Indexed: 02/06/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is a central hub for the regulation of cell proliferation, apoptosis, cell cycle, metabolism, and angiogenesis. Several studies have recently suggested that the PI3K/Akt/mTOR signaling pathway is implicated in the pathogenesis and progression of neuroendocrine tumors. Medullary thyroid cancer (MTC) is a neuroendocrine tumor developing from the C cells of the thyroid. Mutations in the RET proto-oncogene are involved in the pathogenesis of several forms of MTC. The deregulation of the PI3K/Akt/mTOR pathway seems to contribute to the tumorigenic activity of RET proto-oncogene mutations. Targeting this pathway through specific inhibitors at simple or multiple sites may represent an attractive potential therapeutic approach for patients with advanced MTCs. The aim of this review is to examine the role of the PI3K/Akt/mTOR pathway in the development and progression of MTC and the new therapeutic options that target this signaling pathway.
Collapse
Affiliation(s)
- Gloria Irene Manfredi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | | | | | | | | |
Collapse
|
21
|
Moraitis D, Karanikou M, Liakou C, Dimas K, Tzimas G, Tseleni-Balafouta S, Patsouris E, Rassidakis GZ, Kouvaraki MA. SIN1, a critical component of the mTOR-Rictor complex, is overexpressed and associated with AKT activation in medullary and aggressive papillary thyroid carcinomas. Surgery 2014; 156:1542-8; discussion 1548-9. [PMID: 25456951 DOI: 10.1016/j.surg.2014.08.095] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 08/28/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Mammalian target of rapamycin (mTOR) forms 2 active complexes in the cell: the rapamycin-sensitive mTOR-Raptor (mTORC1) and the rapamycin-insensitive mTOR-Rictor (mTORC2). The latter activates AKT kinase, which promotes tumor cell survival and proliferation by multiple downstream targets. Mammalian stress-activated protein kinase interacting protein 1 (SIN1), an essential subunit of the mTORC2 complex, maintains the integrity of the complex and substrate specificity and regulates Akt activation. The role of mTOR-Rictor complex activation in thyroid carcinogenesis remains unknown. Therefore, we investigated expression patterns of Sin1 in the cells lines of thyroid carcinoma and tumors and their association with AKT activation, histologic type, and tumor aggressiveness. METHODS Tissue specimens from 42 patients with thyroid cancer, including follicular (5), papillary (18), medullary (16), and poorly differentiated (3) carcinomas were analyzed via immunohistochemistry for SIN1 expression and AKT phosphorylation at Ser473 residue (Ser473-p-AKT). Eight of 18 papillary carcinomas were aggressive histologic variants. In addition, expression of Sin1 and activation of AKT kinase were analyzed in fresh-frozen tissue samples (normal/tumor), primary cell cultures (papillary thyroid carcinoma [PTC]), and an established thyroid cancer cell line (medullary thyroid carcinoma) by Western blotting. RESULTS With immunohistochemistry, we found that Sin1 was overexpressed in medullary thyroid carcinomas and aggressive variants of papillary thyroid carcinoma compared with conventional papillary and follicular carcinomas (P < .001). Sin1 expression correlated with AKT activation in the entire study group (P = .002). Using Western blot analysis, we found that Sin1 and p-AKT were detected at a greater level in cultured primary cells from aggressive PTC compared with conventional PTC as well as in cell lines of medullary and anaplastic thyroid carcinoma. High expression levels of SIN1 were detected in papillary thyroid carcinomas compared with benign nodules in immunoblots in which we used fresh-frozen patient samples. Two of the Sin1 protein isoforms, p76 and p55, were detected predominantly in PTC samples. CONCLUSION Sin1, a critical factor of the mTORC2 complex is overexpressed in clinically aggressive thyroid cancer types and is associated strongly with activation of AKT kinase. Sin1-dependent AKT activation might represent a target for experimental therapy.
Collapse
Affiliation(s)
| | - Maria Karanikou
- First Department of Pathology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Chryssa Liakou
- First Department of Pathology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | | | - George Tzimas
- Department of Surgery, Hygeia Hospital, Athens, Greece
| | - Sofia Tseleni-Balafouta
- First Department of Pathology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Efstratios Patsouris
- First Department of Pathology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - George Z Rassidakis
- First Department of Pathology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece; Department of Pathology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | - Maria A Kouvaraki
- Department of Surgery, Hygeia Hospital, Athens, Greece; Department of Endocrine Surgery, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
22
|
Lyra J, Vinagre J, Batista R, Pinto V, Prazeres H, Rodrigues F, Eloy C, Sobrinho-Simões M, Soares P. mTOR activation in medullary thyroid carcinoma with RAS mutation. Eur J Endocrinol 2014; 171:633-40. [PMID: 25163725 DOI: 10.1530/eje-14-0389] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Rearranged during transfection (RET) mutations are well-known genetic events in sporadic and familial medullary thyroid carcinoma (FMTC). The presence of RAS mutations in sporadic cases, challenging the RET paradigm in these tumors, has been recently reported. We intend to evaluate mTOR pathway activation in RET- and RAS-mutated MTC. MATERIALS AND METHODS In this study, we analysed the presence of RET, H-RAS, and K-RAS mutations in a series of 87 MTCs (82 apparently sporadic and five FMTCs; five apparently sporadic MTCs were eventually found to be familial). We also evaluated mTOR activation--using the expression of its downstream effector phospho-S6 ribosomal protein (p-S6) and the expression of the mTOR inhibitor, phosphatase and tensin homologue deleted on chromosome 10 (PTEN)--by immunohistochemistry. RESULTS Our results revealed that RET mutations were present in 52.9% of the cases (46/87) and RAS mutations in 12.6% (11/87) of the whole series of MTCs and 14.3% of the 77 sporadic MTCs. The presence of RET and RAS mutations was mutually exclusive. RAS mutations were significantly associated with higher intensity of p-S6 expression (P=0.007), suggesting that the mTOR pathway is activated in such MTCs. We observed also an increased expression of p-S6 in invasive tumors (P=0.042) and in MTCs with lymph node metastases (P=0.046). Cytoplasmic PTEN expression was detected in 58.8% of the cases; cases WT for RAS showed a significantly lower expression of PTEN (P=0.045). CONCLUSIONS We confirmed the presence of RAS mutation in 14.3% of sporadic MTCs and report, for the first time, an association between such mutations and the activation of the mTOR pathway. The evaluation of the mTOR activation by pS6 expression may serve as an indicator of invasive MTC.
Collapse
Affiliation(s)
- Joana Lyra
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - João Vinagre
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - Rui Batista
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - Vasco Pinto
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - Hugo Prazeres
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - Fernando Rodrigues
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - Catarina Eloy
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - Manuel Sobrinho-Simões
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| | - Paula Soares
- Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal Medical FacultyUniversity of Porto, Al. Prof. Hernâni Monteiro, P-4200 Porto, PortugalInstitute of Molecular Pathology and Immunology of the University of Porto (Ipatimup) - Cancer BiologyRuaDr. Roberto Frias, s/n, 4200-465 Porto, PortugalMolecular Pathology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalEndocrinology Service of the Portuguese Institute of Oncology of Coimbra FGEPE, Av. Bissaya Barreto, 98, 3000-075 Coimbra, PortugalDepartment of PathologyHospital de S. João, Al. Prof. Hernâni Monteiro, P-4200 Porto, Portugal
| |
Collapse
|
23
|
Benvenga S, Koch CA. Molecular pathways associated with aggressiveness of papillary thyroid cancer. Curr Genomics 2014; 15:162-70. [PMID: 24955023 PMCID: PMC4064555 DOI: 10.2174/1389202915999140404100958] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 02/26/2014] [Accepted: 03/03/2014] [Indexed: 02/07/2023] Open
Abstract
The most common thyroid malignancy is papillary thyroid cancer (PTC). Mortality rates from PTC mainly depend on its aggressiveness. Geno- and phenotyping of aggressive PTC has advanced our understanding of treatment failures and of potential future therapies. Unraveling molecular signaling pathways of PTC including its aggressive forms will hopefully pave the road to reduce mortality but also morbidity from this cancer. The mitogen-activated protein kinase and the phosphatidylinositol 3-kinase signaling pathway as well as the family of RAS oncogenes and BRAF as a member of the RAF protein family and the aberrant expression of microRNAs miR-221, miR-222, and miR-146b all play major roles in tumor initiation and progression of aggressive PTC. Small molecule tyrosine kinase inhibitors targeting BRAF-mediated events, vascular endothelial growth factor receptors, RET/PTC rearrangements, and other molecular targets, show promising results to improve treatment of radioiodine resistant, recurrent, and aggressive PTC.
Collapse
Affiliation(s)
- Salvatore Benvenga
- Department of Clinical & Experimental Medicine, Section of Endocrinology, University of Messina, Messina, Italy
| | - Christian A Koch
- Division of Endocrinology, University of Mississippi Medical Center, Jackson, MS, USA ; GV (Sonny) Montgomery VA Medical Center, Jackson, MS, USA
| |
Collapse
|
24
|
Akeno N, Miller AL, Ma X, Wikenheiser-Brokamp KA. p53 suppresses carcinoma progression by inhibiting mTOR pathway activation. Oncogene 2014; 34:589-99. [PMID: 24469052 PMCID: PMC4112184 DOI: 10.1038/onc.2013.589] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/18/2013] [Accepted: 12/18/2013] [Indexed: 12/24/2022]
Abstract
Genetic alterations in human cancers and murine models indicate that Rb and p53 have critical tumor suppressive functions in retinoblastoma, a tumor of neural origin, and neuroendocrine tumors including small cell lung cancer and medullary thyroid cancer (MTC). Rb inactivation is the initiating lesion in retinoblastoma and current models propose that induction of apoptosis is a key p53 tumor suppressive function. Genetic studies in mice, however, indicate that other undefined p53 tumor suppressive functions are operative in vivo. How p53 loss cooperates with Rb inactivation to promote carcinogenesis is also not fully understood. In the current study, genetically engineered mice were generated to determine the role of Rb and p53 in MTC pathogenesis and test the hypothesis that p53 suppresses carcinogenesis by inhibiting mTOR signaling. Conditional Rb ablation resulted in thyroid tumors mimicking human MTC, and additional p53 loss led to rapid tumor progression. p53 suppressed tumorigenesis by inhibiting cell cycle progression, but did not induce apoptosis. On the contrary, p53 loss led to increased apoptosis that had to be overcome for tumor progression. mTOR activity was markedly increased in p53 deficient tumors and rapamycin treatment suppressed tumor cell growth identifying mTOR inhibition as a critical p53 tumor suppressive function. Rapamycin treatment did not result in AKT/MAPK activation providing evidence that this feedback mechanism operative in other cancers is not a general response to mTORC1 inhibition. Together, these studies provide mechanistic links between genetic alterations and aberrant signaling pathways critical in carcinogenesis, and identify essential Rb and p53 tumor suppressive functions in vivo.
Collapse
Affiliation(s)
- N Akeno
- Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - A L Miller
- Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - X Ma
- Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - K A Wikenheiser-Brokamp
- 1] Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA [2] Pulmonary Biology, Cincinnati Children's Hospital Medical Center and Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
25
|
Lim SM, Chang H, Yoon MJ, Hong YK, Kim H, Chung WY, Park CS, Nam KH, Kang SW, Kim MK, Kim SB, Lee SH, Kim HG, Na II, Kim YS, Choi MY, Kim JG, Park KU, Yun HJ, Kim JH, Cho BC. A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes. Ann Oncol 2013; 24:3089-94. [PMID: 24050953 DOI: 10.1093/annonc/mdt379] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- S M Lim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Samadi AK, Bazzill J, Zhang X, Gallagher R, Zhang H, Gollapudi R, Kindscher K, Timmermann B, Cohen MS. Novel withanolides target medullary thyroid cancer through inhibition of both RET phosphorylation and the mammalian target of rapamycin pathway. Surgery 2013; 152:1238-47. [PMID: 23158190 DOI: 10.1016/j.surg.2012.08.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 08/16/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND Despite development of current targeted therapies for medullary thyroid cancer (MTC), long-term survival remains unchanged. Recently isolated novel withanolide compounds from Solanaceae physalis are highly potent against MTCs. We hypothesize that these withanolides uniquely inhibit RET phosphorylation and the mammalian target of rapamycin (mTOR) pathway in MTC cells as a mechanism of antiproliferation and apoptosis. METHODS MTC cells were treated with novel withanolides and MTC-targeted drugs. In vitro studies assessed cell viability and proliferation (MTS; trypan blue assays), apoptosis (flow cytometry with Annexin V/PI staining; confirmed by Western blot analysis), long-term cytotoxic effects (clonogenic assay), and suppression of key regulatory proteins such as RET, Akt, and mTOR (by Western blot analysis). RESULTS The novel withanolides potently reduced MTC cell viability (half maximal inhibitory concentration [IC(50)], 270-2,850 nmol/L; 250-1,380 nmol/L for vandetanib; 360-1,640 nmol/L for cabozantinib) with induction of apoptosis at <1,000 nmol/L of drug. Unique from other targeted therapies, withanolides suppressed RET and Akt phosphorylation and protein expression (in a concentration- and time-dependent manner) as well as mTOR activity and translational activity of 4E-BP1 and protein synthesis mediated by p70S6kinase activation at IC(50) concentrations. CONCLUSION Novel withanolides from Physalis selectively and potently inhibit MTC cells in vitro. Unlike other MTC-targeted therapies, these compounds uniquely inhibit both RET kinase activity and the Akt/mTOR prosurvival pathway. Further translational studies are warranted to evaluate their clinical potential.
Collapse
Affiliation(s)
- Abbas K Samadi
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Giunti S, Antonelli A, Amorosi A, Santarpia L. Cellular signaling pathway alterations and potential targeted therapies for medullary thyroid carcinoma. Int J Endocrinol 2013; 2013:803171. [PMID: 23509459 PMCID: PMC3594951 DOI: 10.1155/2013/803171] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 01/08/2013] [Accepted: 01/10/2013] [Indexed: 12/12/2022] Open
Abstract
Parafollicular C-cell-derived medullary thyroid cancer (MTC) comprises 3% to 4% of all thyroid cancers. While cytotoxic treatments have been shown to have limited efficacy, targeted molecular therapies that inhibit rearranged during transfection (RET) and other tyrosine kinase receptors that are mainly involved in angiogenesis have shown great promise in the treatment of metastatic or locally advanced MTC. Multi-tyrosine kinase inhibitors such as vandetanib, which is already approved for the treatment of progressive MTC, and cabozantinib have shown distinct advantages with regard to rates of disease response and control. However, these types of tyrosine kinase inhibitor compounds are able to concurrently block several types of targets, which limits the understanding of RET as a specific target. Moreover, important resistances to tyrosine kinase inhibitors can occur, which limit the long-term efficacy of these treatments. Deregulated cellular signaling pathways and genetic alterations in MTC, particularly the activation of the RAS/mammalian target of rapamycin (mTOR) cascades and RET crosstalk signaling, are now emerging as novel and potentially promising therapeutic treatments for aggressive MTC.
Collapse
Affiliation(s)
- Serena Giunti
- Department of Pathology, Centro Oncologico Fiorentino, Sesto Fiorentino, 50019 Firenze, Italy
| | - Alessandro Antonelli
- Department of Internal Medicine, University of Pisa School of Medicine, 56100 Pisa, Italy
| | - Andrea Amorosi
- Department of Pathology, Centro Oncologico Fiorentino, Sesto Fiorentino, 50019 Firenze, Italy
| | - Libero Santarpia
- Translational Research Unit, Department of Oncology, Istituto Toscano Tumori, 59100 Prato, Italy
- *Libero Santarpia:
| |
Collapse
|
28
|
New molecular targeted therapy and redifferentiation therapy for radioiodine-refractory advanced papillary thyroid carcinoma: literature review. J Thyroid Res 2012; 2012:818204. [PMID: 23320248 PMCID: PMC3540819 DOI: 10.1155/2012/818204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/03/2012] [Indexed: 01/26/2023] Open
Abstract
Although the majority of papillary thyroid carcinoma could be successfully managed by complete surgical resection alone or resection followed by radioiodine ablation, a small proportion of patients may develop radioiodine-refractory progressive disease which is not amenable to surgery, local ablative treatment or other treatment modalities. The use of FDG-PET/CT scan for persistent/recurrent disease has improved the accuracy of restaging as well as cancer prognostication. Given that patients with RAI-refractory disease tend to do significantly worse than those with radioiodine-avid or non-progressive disease, an increasing number of phase I and II studies have been conducted to evaluate the efficacy of new molecular targeted drugs such as the tyrosine kinase inhibitors and redifferentiation drugs. The overall response rate of these drugs ranged between 0–53%, depending on whether the patients had been previously treated with these drugs, performance status and extent of disease. However, drug toxicity remains a major concern in administration of target therapies. Nevertheless, there are also ongoing phase III studies evaluating the efficacy of these new drugs. The aim of the review was to summarize and discuss the results of these targeted drugs and redifferentiation agents for patients with progressive, radioiodine-refractory papillary thyroid carcinoma.
Collapse
|
29
|
Nguyen SA, Walker D, Gillespie MB, Gutkind JS, Day TA. mTOR inhibitors and its role in the treatment of head and neck squamous cell carcinoma. Curr Treat Options Oncol 2012; 13:71-81. [PMID: 22282394 DOI: 10.1007/s11864-011-0180-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OPINION STATEMENT Head and neck squamous cell carcinomas (HNSCC) represent 6% of all cancers diagnosed each year in the United States, affecting approximately 43,000 new patients and resulting in approximately 12,000 deaths. Currently, three main rapalogs exist for the treatment of cancer: CCI-779 (temsirolimus), RAD001 (everolimus), and AP235373 (deforolimus). Clinicians managing HNSCC need to be aware of the three rapalogs. Extensive evidence has shown rapamycin-analogs to be effective agents in the treatment of a number of solid tumors. While extensive preclinical data suggests that HNSCC would be an appropriate tumor type to benefit from inhibition of the mTOR pathway, limited clinical data is yet available to support this. Numerous phase II trials evaluating mTOR inhibitors for use in HNSCC are currently recruiting patients.
Collapse
Affiliation(s)
- Shaun A Nguyen
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | |
Collapse
|