1
|
Dutta S, Knauf JA. Development of animal models to study aggressive thyroid cancers. Eur Thyroid J 2025; 14:e240361. [PMID: 39874138 PMCID: PMC11825169 DOI: 10.1530/etj-24-0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 01/30/2025] Open
Abstract
The development of mouse models for thyroid cancer has significantly advanced over the years, enhancing our understanding of thyroid tumorigenesis, molecular pathways and treatment responses. The earliest mouse models of thyroid cancer relied on hormone, radiation or chemical carcinogenesis to induce tumors. However, as our understanding of the genetic alterations driving thyroid cancer has expanded, more sophisticated genetic engineering techniques have been employed to create models with thyroid-specific expression of these driver mutations. While driver mutations can initiate tumorigenesis, they are often insufficient to sustain cancer progression and invasion, which significantly limits their usefulness in studying advanced thyroid cancers. Recent studies exploring the genomic landscape of advanced thyroid cancer have identified several cooperating mutations, which are secondary genetic alterations that work alongside driver mutations to promote thyroid tumor progression. Indeed, mice with a combination of oncogenic drivers and common cooperating alterations have been developed, demonstrating that these alterations function in conjunction with the oncogenic driver to promote the progression to advanced thyroid cancer. These models provide important preclinical tools to explore how cooperating alterations influence the response to therapies, particularly those targeting the oncogenic driver. This review will focus on recent publications that broaden the scope of advanced thyroid cancer models by combining thyroid-specific oncogenic driver expression with various cooperating mutations.
Collapse
Affiliation(s)
- Shovan Dutta
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jeffrey A Knauf
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Otolaryngology-Head & Neck Surgery, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Tiedje V, Greenberg J, Qin T, Im SY, Krishnamoorthy GP, Boucai L, Xu B, French JD, Sherman EJ, Ho AL, de Stanchina E, Socci ND, Jin J, Ghossein R, Knauf JA, Koche RP, Fagin JA. Loss of tumor cell MHC Class II drives insensitivity of BRAF-mutant anaplastic thyroid cancers to MAPK inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635086. [PMID: 40093098 PMCID: PMC11908150 DOI: 10.1101/2025.01.27.635086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Cancer cells present neoantigens dominantly through MHC class I (MHCI) to drive tumor rejection through cytotoxic CD8+ T-cells. There is growing recognition that a subset of tumors express MHC class II (MHCII), causing recognition of antigens by TCRs of CD4+ T-cells that contribute to the anti-tumor response. We find that mouse Braf V600E -driven anaplastic thyroid cancers (ATC) respond markedly to the RAF + MEK inhibitors dabrafenib and trametinib (dab/tram) and that this is associated with upregulation of MhcII in cancer cells and increased CD4+ T-cell infiltration. A subset of recurrent tumors lose MhcII expression due to silencing of Ciita , the master transcriptional regulator of MhcII, despite preserved interferon gamma signal transduction, which can be rescued by EZH2 inhibition. Orthotopically-implanted Ciita -/- and H2-Ab1 -/- ATC cells into immune competent mice become unresponsive to the MAPK inhibitors. Moreover, depletion of CD4+, but not CD8+ T-cells, also abrogates response to dab/tram. These findings implicate MHCII-driven CD4+ T cell activation as a key determinant of the response of Braf-mutant ATCs to MAPK inhibition.
Collapse
|
3
|
Zhang D, Zhang W, Liu H, Liu P, Li C, Liu Y, Han J, Zhu G. Recent advances in the treatment of non-small cell lung cancer with MET inhibitors. Front Chem 2024; 12:1501844. [PMID: 39720556 PMCID: PMC11666382 DOI: 10.3389/fchem.2024.1501844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024] Open
Abstract
Recently, research into the oncogenic driver genes associated with non-small cell lung cancer (NSCLC) has advanced significantly, leading to the development and clinical application of an increasing number of approved therapeutic agents. Among these, small molecule inhibitors that target mesenchymal-epithelial transition (MET) have demonstrated successful application in clinical settings. Currently, three categories of small molecule MET inhibitors, characterized by distinct binding patterns to the MET kinase region, have been developed: types Ia/Ib, II, and III. This review thoroughly examines MET's structure and its crucial role in NSCLC initiation and progression, explores discovery strategies for MET inhibitors, and discusses advancements in understanding resistance mechanisms. These insights are anticipated to enhance the development of a new generation of MET inhibitors characterized by high efficiency, selectivity, and low toxicity, thereby offering additional therapeutic alternatives for patients diagnosed with NSCLC.
Collapse
Affiliation(s)
- Dongna Zhang
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Wenying Zhang
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - He Liu
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Pan Liu
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Chunxin Li
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yangyang Liu
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jicheng Han
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of integrative medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Guangze Zhu
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
4
|
Cirello V, Gambale C, Nikitski AV, Masaki C, Roque J, Colombo C. Poorly differentiated thyroid carcinoma: molecular, clinico-pathological hallmarks and therapeutic perspectives. Panminerva Med 2024; 66:155-173. [PMID: 38576304 DOI: 10.23736/s0031-0808.23.05040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Poorly differentiated thyroid carcinoma (PDTC) is a rare and extremely aggressive tumor, accounting for about 2-15% of all thyroid cancer. PDTC has a distinct biological behavior compared to well-differentiated and anaplastic thyroid carcinoma and, in last years, it has been classified as a separate entity from both anatomopathological and clinical points of view. Nevertheless, there is still a lack of consensus among clinicians regarding inclusion criteria and definition of PDTC that affects its diagnosis and clinical management. Due to its rarity and difficulty in classification compared to other tumors, very few studies are available to date and series often include different histotypes in addition to PDTC. This review focuses on main studies concerning PDTC summarizing the evolution in the definition of its diagnosis criteria, clinicopathological features, management, and outcome. The data available confirm that the pathological evaluation and classification of PDTC are crucial and should therefore be standardized. Since the clinical presentation and prognosis of PDTC may vary widely depending on the different stage of the disease at diagnosis, the patient's management may differ in treatment and should be tailored to each patient. Finally, this review discusses advances in molecular insights of PDTC that, together with the implementation of both in vitro and in vivo models, will provide valuable insights into biological mechanisms of progression, metastasis, and invasion of this aggressive thyroid carcinoma. Further studies on larger, carefully selected series are needed to better assess the peculiar features of PDTC and to better define its management by focusing on the best diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Valentina Cirello
- Endocrine Oncology Unit, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Carla Gambale
- Department of Clinical and Experimental Medicine, Endocrine Unit, University Hospital of Pisa, Pisa, Italy
| | - Alyaksandr V Nikitski
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Chie Masaki
- Department of Surgery, Ito Hospital, Tokyo, Japan
| | - João Roque
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
| | - Carla Colombo
- Endocrine Oncology Unit, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy -
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
5
|
Landa I, Cabanillas ME. Genomic alterations in thyroid cancer: biological and clinical insights. Nat Rev Endocrinol 2024; 20:93-110. [PMID: 38049644 DOI: 10.1038/s41574-023-00920-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 12/06/2023]
Abstract
Tumours can arise from thyroid follicular cells if they acquire driver mutations that constitutively activate the MAPK signalling pathway. In addition, a limited set of additional mutations in key genes drive tumour progression towards more aggressive and less differentiated disease. Unprecedented insights into thyroid tumour biology have come from the breadth of thyroid tumour sequencing data from patients and the wide range of mutation-specific mechanisms identified in experimental models, in combination with the genomic simplicity of thyroid cancers. This knowledge is gradually being translated into refined strategies to stratify, manage and treat patients with thyroid cancer. This Review summarizes the biological underpinnings of the genetic alterations involved in thyroid cancer initiation and progression. We also provide a rationale for and discuss specific examples of how to implement genomic information to inform both recommended and investigational approaches to improve thyroid cancer prognosis, redifferentiation strategies and targeted therapies.
Collapse
Affiliation(s)
- Iñigo Landa
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
6
|
Califano I, Smulever A, Jerkovich F, Pitoia F. Advances in the management of anaplastic thyroid carcinoma: transforming a life-threatening condition into a potentially treatable disease. Rev Endocr Metab Disord 2024; 25:123-147. [PMID: 37648897 DOI: 10.1007/s11154-023-09833-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Anaplastic thyroid cancer (ATC) is an infrequent thyroid tumor that usually occurs in elderly patients. There is often a history of previous differentiated thyroid cancer suggesting a biological progression. It is clinically characterized by a locally invasive cervical mass of rapid onset. Metastases are found at diagnosis in 50% of patients. Due to its adverse prognosis, a prompt diagnosis is crucial. In patients with unresectable or metastatic disease, multimodal therapy (chemotherapy and external beam radiotherapy) has yielded poor outcomes with 12-month overall survival of less than 20%. Recently, significant progress has been made in understanding the oncogenic pathways of ATC, leading to the identification of BRAF V600E mutations as the driver oncogene in nearly 40% of cases. The combination of the BRAF inhibitor dabrafenib (D) and MEK inhibitor trametinib (T) showed outstanding response rates in BRAF-mutated ATC and is now considered the standard of care in this setting. Recently, it was shown that neoadjuvant use of DT followed by surgery achieved 24-month overall survival rates of 80%. Although these approaches have changed the management of ATC, effective therapies are still needed for patients with BRAF wild-type ATC, and high-quality evidence is lacking for most aspects of this neoplasia. Additionally, in real-world settings, timely access to multidisciplinary care, molecular testing, and targeted therapies continues to be a challenge. Health policies are warranted to ensure specialized treatment for ATC.The expanding knowledge of ATC´s molecular biology, in addition to the ongoing clinical trials provides hope for the development of further therapeutic options.
Collapse
Affiliation(s)
- Inés Califano
- Endocrinology Division, Instituto de Oncología AH Roffo, University of Buenos Aires, Buenos Aires, Argentina.
| | - Anabella Smulever
- Endocrinology Division, Instituto de Investigaciones Médicas A. Lanari, University of Buenos Aires, Buenos Aires, Argentina
| | - Fernando Jerkovich
- Endocrinology Division, Hospital de Clínicas, University of Buenos Aires, Buenos Aires, Argentina
| | - Fabian Pitoia
- Endocrinology Division, Hospital de Clínicas, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
Lee M, Morris LGT. Genetic alterations in thyroid cancer mediating both resistance to BRAF inhibition and anaplastic transformation. Oncotarget 2024; 15:36-48. [PMID: 38275291 PMCID: PMC10812235 DOI: 10.18632/oncotarget.28544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/08/2023] [Indexed: 01/27/2024] Open
Abstract
A subset of thyroid cancers present at advanced stage or with dedifferentiated histology and have limited response to standard therapy. Tumors harboring the BRAF V600E mutation may be treated with BRAF inhibitors; however, tumor response is often short lived due to multiple compensatory resistance mechanisms. One mode of resistance is the transition to an alternative cell state, which on rare occasions can correspond to tumor dedifferentiation. DNA sequencing and RNA expression profiling show that thyroid tumors that dedifferentiate after BRAF inhibition are enriched in known genetic alterations that mediate resistance to BRAF blockade, and may also drive tumor dedifferentiation, including mutations in the PI3K/AKT/MTOR (PIK3CA, MTOR), MAP/ERK (MET, NF2, NRAS, RASA1), SWI/SNF chromatin remodeling complex (ARID2, PBRM1), and JAK/STAT pathways (JAK1). Given these findings, recent investigations have evaluated the efficacy of dual-target therapies; however, continued lack of long-term tumor control illustrates the complex and multifactorial nature of these compensatory mechanisms. Transition to an immune-suppressed state is another correlate of BRAF inhibitor resistance and tumor dedifferentiation, suggesting a possible role for concurrent targeted therapy with immunotherapy. Investigations into combined targeted and immunotherapy are ongoing, but early results with checkpoint inhibitors, viral therapies, and CAR T-cells suggest enhanced anti-tumor immune activity with these combinations.
Collapse
Affiliation(s)
- Mark Lee
- Department of Otolaryngology-Head and Neck Surgery, New York Presbyterian Hospital, New York, NY 10032, USA
| | - Luc GT Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
8
|
Cleere EF, Crotty TJ, Hintze JM, Fitzgerald CWR, Kinsella J, Lennon P, Timon CVI, Woods RSR, Shine NP, O'Neill JP. The role of surgery for anaplastic thyroid carcinoma in the era of targeted therapeutics: A scoping review. Laryngoscope Investig Otolaryngol 2023; 8:1673-1684. [PMID: 38130255 PMCID: PMC10731486 DOI: 10.1002/lio2.1172] [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: 08/06/2023] [Accepted: 10/06/2023] [Indexed: 12/23/2023] Open
Abstract
Background Questions exist regarding patient selection for surgery in anaplastic thyroid carcinoma (ATC), particularly with the advent of neoadjuvant-targeted therapeutics. The present scoping review sought to evaluate what extent of surgical resection should be performed in ATC. Methods A scoping review was carried out in accordance with Joanna Briggs Institute and the preferred reporting items for systematic reviews and meta-analyses extension for scoping reviews (PRISMA-ScR) protocols. Included studies were required to provide clear description of the surgery performed for ATC. Results The final search identified 6901 articles. Ultimately only 15 articles including 1484 patients met inclusion criteria. A total of 765 patients (51.5%) underwent attempted curative intent surgery. The approach to resection of adjacent tissues varied between studies. Eight studies considered laryngeal ± pharyngeal resection (8/15, 53.3%), eight studies (53.3%) considered tracheal resection and again eight studies (53.3%) considered esophageal resection. More extensive resections increased morbidity without improving overall survival (OS) (<9 months in the 12 studies using a combination of surgery and chemoradiotherapy). In the three studies utilizing targeted therapy in addition to surgery, OS was notably improved while surgical resection following neoadjuvant therapy was less extensive. Conclusions There is no clear agreement in the literature regarding the limits of surgical resection in locoregionally advanced ATC. A definition of surgically resectable disease will be required to guide surgical decision making in ATC, particularly with the potential to reduce tumor burden using neoadjuvant targeted treatment in suitable patients. Level of evidence III.
Collapse
Affiliation(s)
- Eoin F. Cleere
- Department of Otolaryngology Head and Neck Surgery, Beaumont HospitalDublinIreland
- The Royal College of Surgeons in IrelandDublinIreland
| | - Thomas J. Crotty
- Department of Otolaryngology Head and Neck Surgery, St James's HospitalDublinIreland
| | - Justin M. Hintze
- Department of Otolaryngology Head and Neck Surgery, St James's HospitalDublinIreland
| | | | - John Kinsella
- Department of Otolaryngology Head and Neck Surgery, St James's HospitalDublinIreland
| | - Paul Lennon
- Department of Otolaryngology Head and Neck Surgery, St James's HospitalDublinIreland
| | - Conrad V. I. Timon
- Department of Otolaryngology Head and Neck Surgery, St James's HospitalDublinIreland
| | - Robbie S. R. Woods
- Department of Otolaryngology Head and Neck Surgery, Beaumont HospitalDublinIreland
| | - Neville P. Shine
- Department of Otolaryngology Head and Neck Surgery, Beaumont HospitalDublinIreland
| | - James P. O'Neill
- Department of Otolaryngology Head and Neck Surgery, Beaumont HospitalDublinIreland
- The Royal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW To summarize recent developments in the diagnosis and management of patients with anaplastic thyroid cancer (ATC). RECENT FINDINGS An updated edition of the Classification of Endocrine and Neuroendocrine Tumors was released by the World Health Organization (WHO), in which squamous cell carcinoma of the thyroid are now a subtype of ATC. Broader access to next generation sequencing has allowed better understanding of the molecular mechanisms driving ATC and improved prognostication. BRAF-targeted therapies revolutionized the treatment of advanced/metastatic BRAFV600E -mutated ATC, offering significant clinical benefit and allowing better locoregional control of disease through the neoadjuvant approach. However, inevitable development of resistance mechanisms represents a major challenge. Addition of immunotherapy to BRAF/MEK inhibition has shown very promising results and significant improvement in survival outcomes. SUMMARY Major advancements took place in the characterization and management of ATC in recent years, especially in patients with a BRAF V600E mutation. Still, no curative treatment is available, and options are limited once resistance to currently available BRAF-targeted therapies develops. Additionally, there is still a need for more effective treatments for patients without a BRAF mutation.
Collapse
Affiliation(s)
- Sarah Hamidi
- Department of Endocrine Neoplasia and Hormonal Disorders
| | - Anastasios Maniakas
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
10
|
Fagin JA, Krishnamoorthy GP, Landa I. Pathogenesis of cancers derived from thyroid follicular cells. Nat Rev Cancer 2023; 23:631-650. [PMID: 37438605 PMCID: PMC10763075 DOI: 10.1038/s41568-023-00598-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
The genomic simplicity of differentiated cancers derived from thyroid follicular cells offers unique insights into how oncogenic drivers impact tumour phenotype. Essentially, the main oncoproteins in thyroid cancer activate nodes in the receptor tyrosine kinase-RAS-BRAF pathway, which constitutively induces MAPK signalling to varying degrees consistent with their specific biochemical mechanisms of action. The magnitude of the flux through the MAPK signalling pathway determines key elements of thyroid cancer biology, including differentiation state, invasive properties and the cellular composition of the tumour microenvironment. Progression of disease results from genomic lesions that drive immortalization, disrupt chromatin accessibility and cause cell cycle checkpoint dysfunction, in conjunction with a tumour microenvironment characterized by progressive immunosuppression. This Review charts the genomic trajectories of these common endocrine tumours, while connecting them to the biological states that they confer.
Collapse
Affiliation(s)
- James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Gnana P Krishnamoorthy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iñigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
11
|
Leandro-García LJ, Landa I. Mechanistic Insights of Thyroid Cancer Progression. Endocrinology 2023; 164:bqad118. [PMID: 37503738 PMCID: PMC10403681 DOI: 10.1210/endocr/bqad118] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
Differentiated thyroid cancers (DTCs) are primarily initiated by mutations that activate the MAPK signaling cascade, typically at BRAF or RAS oncoproteins. DTCs can evolve to more aggressive forms, specifically, poorly differentiated (PDTC) and anaplastic thyroid cancers (ATC), by acquiring additional genetic alterations which deregulate key pathways. In this review, we focused on bona fide mutations involved in thyroid cancer progression for which consistent mechanistic data exist. Here we summarized the relevant literature, spanning approximately 2 decades, highlighting genetic alterations that are unquestionably enriched in PDTC/ATC. We describe the relevant functional data obtained in multiple in vitro and in vivo thyroid cancer models employed to study genetic alterations in the following genes and functional groups: TP53, effectors of the PI3K/AKT pathway, TERT promoter, members of the SWI/SNF chromatin remodeling complex, NF2, and EIF1AX. In addition, we briefly discuss other genetic alterations that are selected in aggressive thyroid tumors but for which mechanistic data is still either limited or nonexistent. Overall, we argue for the importance conveyed by preclinical studies for the clinical translation of genomic knowledge of thyroid cancers.
Collapse
Affiliation(s)
- Luis Javier Leandro-García
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Iñigo Landa
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Ruiz EM, Alhassan SA, Errami Y, Abd Elmageed ZY, Fang JS, Wang G, Brooks MA, Abi-Rached JA, Kandil E, Zerfaoui M. A Predictive Model of Adaptive Resistance to BRAF/MEK Inhibitors in Melanoma. Int J Mol Sci 2023; 24:8407. [PMID: 37176114 PMCID: PMC10178962 DOI: 10.3390/ijms24098407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The adaptive acquisition of resistance to BRAF and MEK inhibitor-based therapy is a common feature of melanoma cells and contributes to poor patient treatment outcomes. Leveraging insights from a proteomic study and publicly available transcriptomic data, we evaluated the predictive capacity of a gene panel corresponding to proteins differentially abundant between treatment-sensitive and treatment-resistant cell lines, deciphering predictors of treatment resistance and potential resistance mechanisms to BRAF/MEK inhibitor therapy in patient biopsy samples. From our analysis, a 13-gene signature panel, in both test and validation datasets, could identify treatment-resistant or progressed melanoma cases with an accuracy and sensitivity of over 70%. The dysregulation of HMOX1, ICAM, MMP2, and SPARC defined a BRAF/MEK treatment-resistant landscape, with resistant cases showing a >2-fold risk of expression of these genes. Furthermore, we utilized a combination of functional enrichment- and gene expression-derived scores to model and identify pathways, such as HMOX1-mediated mitochondrial stress response, as potential key drivers of the emergence of a BRAF/MEK inhibitor-resistant state in melanoma cells. Overall, our results highlight the utility of these genes in predicting treatment outcomes and the underlying mechanisms that can be targeted to reduce the development of resistance to BRAF/MEK targeted therapy.
Collapse
Affiliation(s)
- Emmanuelle M. Ruiz
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Solomon A. Alhassan
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Youssef Errami
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Zakaria Y. Abd Elmageed
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of Louisiana, Monroe, LA 71203, USA
| | - Jennifer S. Fang
- Department of Cell and Molecular Biology, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA
| | - Guangdi Wang
- Department of Chemistry, RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Margaret A. Brooks
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Joe A. Abi-Rached
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Emad Kandil
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Mourad Zerfaoui
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
13
|
Branigan GP, Casado-Medrano V, O’Neill AB, Ricarte-Filho JC, Massoll N, Salwen M, Spangler Z, Scheerer M, Williamson EK, Bauer AJ, Franco AT. Development of Novel Murine BRAF V600E-Driven Papillary Thyroid Cancer Cell Lines for Modeling of Disease Progression and Preclinical Evaluation of Therapeutics. Cancers (Basel) 2023; 15:879. [PMID: 36765847 PMCID: PMC9913801 DOI: 10.3390/cancers15030879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 02/04/2023] Open
Abstract
The Cancer Genome Atlas study in thyroid cancer exposed the genomic landscape of ~500 PTCs and revealed BRAFV600E-mutant tumors as having different prognosis, contrasting indolent cases and those with more invasive disease. Here, we describe the generation and characterization of six novel BRAFV600E-driven papillary thyroid cancer (PTC) cell lines established from a BrafV600E+/-/Pten+/-/TPO-Cre mouse model that spontaneously develop thyroid tumors. The novel cell lines were obtained from animals representing a range of developmental stages and both sexes, with the goal of establishing a heterogeneous panel of PTC cell lines sharing a common driver mutation. These cell lines recapitulate the genetics and diverse histopathological features of BRAFV600E-driven PTC, exhibiting differing degrees of growth, differentiation, and invasive potential that may help define mechanisms of pathogenesis underlying the heterogeneity present in the patient population. We demonstrate that these cell lines can be used for a variety of in vitro applications and can maintain the potential for in vivo transplantation into immunocompetent hosts. We believe that these novel cell lines will provide powerful tools for investigating the molecular basis of thyroid cancer progression and will lead to the development of more personalized diagnostic and treatment strategies for BRAFV600E-driven PTC.
Collapse
Affiliation(s)
- Grace Purvis Branigan
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Victoria Casado-Medrano
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alison B. O’Neill
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Julio C. Ricarte-Filho
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicole Massoll
- Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Madeleine Salwen
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Zachary Spangler
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Michele Scheerer
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Edward K. Williamson
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew J. Bauer
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Aime T. Franco
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Schubert L, Mariko ML, Clerc J, Huillard O, Groussin L. MAPK Pathway Inhibitors in Thyroid Cancer: Preclinical and Clinical Data. Cancers (Basel) 2023; 15:cancers15030710. [PMID: 36765665 PMCID: PMC9913385 DOI: 10.3390/cancers15030710] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Thyroid cancer is the most common endocrine cancer, with a good prognosis in most cases. However, some cancers of follicular origin are metastatic or recurrent and eventually become radioiodine refractory thyroid cancers (RAIR-TC). These more aggressive cancers are a clinical concern for which the therapeutic arsenal remains limited. Molecular biology of these tumors has highlighted a hyper-activation of the Mitogen-Activated Protein Kinases (MAPK) pathway (RAS-RAF-MEK-ERK), mostly secondary to the BRAFV600E hotspot mutation occurring in about 60% of papillary cancers and 45% of anaplastic cancers. Therapies targeting the different protagonists of this signaling pathway have been tested in preclinical and clinical models: first and second generation RAF inhibitors and MEK inhibitors. In clinical practice, dual therapies with a BRAF inhibitor and a MEK inhibitor are being recommended in anaplastic cancers with the BRAFV600E mutation. Concerning RAIR-TC, these inhibitors can be used as anti-proliferative drugs, but their efficacy is inconsistent due to primary or secondary resistance. A specific therapeutic approach in thyroid cancers consists of performing a short-term treatment with these MAPK pathway inhibitors to evaluate their capacity to redifferentiate a refractory tumor, with the aim of retreating the patients by radioactive iodine therapy in case of re-expression of the sodium-iodide symporter (NIS). In this work, we report data from recent preclinical and clinical studies on the efficacy of MAPK pathway inhibitors and their resistance mechanisms. We will also report the different preclinical and clinical studies that have investigated the redifferentiation with these therapies.
Collapse
Affiliation(s)
- Louis Schubert
- Department of Endocrinology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, 75014 Paris, France
| | - Mohamed Lamine Mariko
- Department of Endocrinology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, 75014 Paris, France
| | - Jérôme Clerc
- Department of Nuclear Medicine, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, 75014 Paris, France
| | - Olivier Huillard
- Institut du Cancer Paris CARPEM, Department of Medical Oncology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Lionel Groussin
- Department of Endocrinology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, 75014 Paris, France
- Correspondence:
| |
Collapse
|
15
|
Li Q, Tie Y, Alu A, Ma X, Shi H. Targeted therapy for head and neck cancer: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:31. [PMID: 36646686 PMCID: PMC9842704 DOI: 10.1038/s41392-022-01297-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/27/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Head and neck cancer (HNC) is malignant, genetically complex and difficult to treat and is the sixth most frequent cancer, with tobacco, alcohol and human papillomavirus being major risk factors. Based on epigenetic data, HNC is remarkably heterogeneous, and treatment remains challenging. There is a lack of significant improvement in survival and quality of life in patients with HNC. Over half of HNC patients experience locoregional recurrence or distal metastasis despite the current multiple traditional therapeutic strategies and immunotherapy. In addition, resistance to chemotherapy, radiotherapy and some targeted therapies is common. Therefore, it is urgent to explore more effective and tolerable targeted therapies to improve the clinical outcomes of HNC patients. Recent targeted therapy studies have focused on identifying promising biomarkers and developing more effective targeted therapies. A well understanding of the pathogenesis of HNC contributes to learning more about its inner association, which provides novel insight into the development of small molecule inhibitors. In this review, we summarized the vital signaling pathways and discussed the current potential therapeutic targets against critical molecules in HNC, as well as presenting preclinical animal models and ongoing or completed clinical studies about targeted therapy, which may contribute to a more favorable prognosis of HNC. Targeted therapy in combination with other therapies and its limitations were also discussed.
Collapse
Affiliation(s)
- Qingfang Li
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Tie
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Abstract
Anaplastic thyroid cancer (ATC) is one of the most lethal of all cancers. It is more common in women and occurs primarily in older patients. ATC has a median overall survival of 3-5 months and a nearly 100% disease-specific mortality. It is known to spread rapidly to locoregional structures as well as outside the neck to distant sites, hence ATC is always considered stage IV. With better understanding of the disease at a molecular level, the introduction of newer treatment strategies has been possible and is part of the multimodal (surgery, radiation, and systemic therapy) therapeutic approach. However, there is extensive work needed to achieve better survival outcomes.
Collapse
Affiliation(s)
- Sarika N Rao
- Mayo Clinic School of Medicine, Dual Appointment in the Divisions of Endocrinology/Metabolism and Hematology/Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Robert C Smallridge
- Mayo Clinic School of Medicine, Division of Endocrinology/Metabolism, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| |
Collapse
|
17
|
Fuziwara CS, Kimura ET. Using CRISPR/Cas9 to Edit a Thyroid Cancer Cell Line. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1429:73-84. [PMID: 37486517 DOI: 10.1007/978-3-031-33325-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Thyroid cancer is the most prevalent endocrine malignancy, comprising multiple types of cancer, with distinct clinical-pathological characteristics. The oncogenesis of thyroid cancer is related to genetic alterations in MAPK signaling that induce proliferation and modulate noncoding genes, such as microRNAs and long noncoding RNAs. In this context, CRISPR/Cas9 emerges as a potential tool to modify gene sequence and modulate gene expression in thyroid cancer cell lines. In this chapter, we explore some of the current studies in which researchers have applied CRISPR/Cas9 in vitro to investigate thyroid cancer biology (Fig. 5.1).
Collapse
Affiliation(s)
- Cesar Seigi Fuziwara
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Edna Teruko Kimura
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
18
|
Wang JR, Montierth M, Xu L, Goswami M, Zhao X, Cote G, Wang W, Iyer P, Dadu R, Busaidy NL, Lai SY, Gross ND, Ferrarotto R, Lu C, Gunn GB, Williams MD, Routbort M, Zafereo ME, Cabanillas ME. Impact of Somatic Mutations on Survival Outcomes in Patients With Anaplastic Thyroid Carcinoma. JCO Precis Oncol 2022; 6:e2100504. [PMID: 35977347 PMCID: PMC10530586 DOI: 10.1200/po.21.00504] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/02/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Anaplastic thyroid carcinoma (ATC) uniformly present with aggressive disease, but the mutational landscape of tumors varies. We aimed to determine whether tumor mutations affect survival outcomes in ATC. MATERIALS AND METHODS Patients who underwent mutation sequencing using targeted gene panels between 2005 and 2019 at a tertiary referral center were included. Associations between mutation status and survival outcomes were assessed using Cox proportional hazards models. RESULTS A total of 202 patients were included, where 122 died of ATC (60%). The median follow-up was 31 months (interquartile range, 18-45 months). The most common mutations were in TP53 (59%), BRAF (41%), TERT promoter (37%), and the RAS gene family (22%). Clinicopathologic characteristics and overall survival (OS) significantly correlated with mutations in BRAFV600E and RAS, which were mutually exclusive. The BRAFV600E mutation was associated with the presence of a papillary thyroid carcinoma precursor and significantly better OS (median OS: 24 months). RAS-mutated patients more commonly presented without cervical lymph node involvement but had the worst OS (median OS: 6 months). Tumors that were wild-type for both BRAF and RAS were enriched for NF1 mutations and harbored intermediate prognosis (median OS: 15 months). In multivariate analyses, RAS mutations were associated with a more than 2.5-fold higher risk of death (adjusted hazard ratio, 2.64; 95% CI, 1.66 to 4.20) compared with BRAFV600E. In patients treated with BRAF-directed therapy (n = 60), disease progression occurred in 48% of patients (n = 29). The median progression-free survival was 14 months. The presence of a TP53 mutation was independently associated with reduced progression-free survival in BRAFV600E-mutated patients treated with BRAF-directed therapy (adjusted hazard ratio, 2.89; 95% CI, 1.35 to 6.21). CONCLUSION Mutation analysis provides prognostic information in ATC and should be incorporated into routine clinical care.
Collapse
Affiliation(s)
- Jennifer Rui Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Matthew Montierth
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Li Xu
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Maitrayee Goswami
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Xiao Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Gilbert Cote
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Priyanka Iyer
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Ramona Dadu
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Neil D Gross
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Renata Ferrarotto
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Charles Lu
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Gary Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Michelle D Williams
- Department of Pathology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Mark Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Mark E Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center; Houston, TX
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX
| |
Collapse
|
19
|
Zhang Y, Xing Z, Liu T, Tang M, Mi L, Zhu J, Wu W, Wei T. Targeted therapy and drug resistance in thyroid cancer. Eur J Med Chem 2022; 238:114500. [DOI: 10.1016/j.ejmech.2022.114500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022]
|
20
|
To KKW, Cho WCS. Mesenchymal Epithelial Transition Factor (MET): A Key Player in Chemotherapy Resistance and an Emerging Target for Potentiating Cancer Immunotherapy. Curr Cancer Drug Targets 2022; 22:269-285. [PMID: 35255791 DOI: 10.2174/1568009622666220307105107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 11/22/2022]
Abstract
The MET protein is a cell surface receptor tyrosine kinase predominately expressed in epithelial cells. Upon binding of its only known ligand, hepatocyte growth factor (HGF), MET homodimerizes, phosphorylates, and stimulates intracellular signalling to drive cell proliferation. Amplification or hyperactivation of MET is frequently observed in various cancer types and it is associated with poor response to conventional and targeted chemotherapy. More recently, emerging evidence also suggests that MET/HGF signalling may play an immunosuppressive role and it could confer resistance to cancer immunotherapy. In this review, we summarized the preclinical and clinical evidence of MET's role in drug resistance to conventional chemotherapy, targeted therapy, and immunotherapy. Previous clinical trials investigating MET-targeted therapy in unselected or MET-overexpressing cancers yielded mostly unfavourable results. More recent clinical studies focusing on MET exon 14 alterations and MET amplification have produced encouraging treatment responses to MET inhibitor therapy. The translational relevance of MET inhibitor therapy to overcome drug resistance in cancer patients is discussed.
Collapse
Affiliation(s)
- Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| |
Collapse
|
21
|
Jannin A, Escande A, Al Ghuzlan A, Blanchard P, Hartl D, Chevalier B, Deschamps F, Lamartina L, Lacroix L, Dupuy C, Baudin E, Do Cao C, Hadoux J. Anaplastic Thyroid Carcinoma: An Update. Cancers (Basel) 2022; 14:cancers14041061. [PMID: 35205809 PMCID: PMC8869821 DOI: 10.3390/cancers14041061] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 01/13/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare and undifferentiated form of thyroid cancer. Its prognosis is poor: the median overall survival (OS) of patients varies from 4 to 10 months after diagnosis. However, a doubling of the OS time may be possible owing to a more systematic use of molecular tests for targeted therapies and integration of fast-track dedicated care pathways for these patients in tertiary centers. The diagnostic confirmation, if needed, requires an urgent biopsy reread by an expert pathologist with additional immunohistochemical and molecular analyses. Therapeutic management, defined in multidisciplinary meetings, respecting the patient's choice, must start within days following diagnosis. For localized disease diagnosed after primary surgical treatment, adjuvant chemo-radiotherapy is recommended. In the event of locally advanced or metastatic disease, the prognosis is very poor. Treatment should then involve chemotherapy or targeted therapy and decompressive cervical radiotherapy. Here we will review current knowledge on ATC and provide perspectives to improve the management of this deadly disease.
Collapse
Affiliation(s)
- Arnaud Jannin
- Department of Endocrinology, Diabetology, Metabolism and Nutrition, Lille University Hospital, 59000 Lille, France; (A.J.); (B.C.); (C.D.C.)
- H. Warembourg School of Medicine, University of Lille, 59000 Lille, France;
| | - Alexandre Escande
- H. Warembourg School of Medicine, University of Lille, 59000 Lille, France;
- Academic Radiation Oncology Department, Oscar Lambret Center, 59000 Lille, France
| | - Abir Al Ghuzlan
- Cancer Medical Pathology and Biology Department, Institute Gustave Roussy, 94805 Villejuif, France;
| | - Pierre Blanchard
- Department of Radiation Oncology, Institute Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France;
| | - Dana Hartl
- Département d’Anesthésie, Chirurgie et Interventionnel (DACI), Institute Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France;
| | - Benjamin Chevalier
- Department of Endocrinology, Diabetology, Metabolism and Nutrition, Lille University Hospital, 59000 Lille, France; (A.J.); (B.C.); (C.D.C.)
- H. Warembourg School of Medicine, University of Lille, 59000 Lille, France;
| | - Frédéric Deschamps
- Department of Head and Neck Oncology, Institute Gustave Roussy, Université Paris Saclay, 94805 Paris, France;
| | - Livia Lamartina
- Cancer Medicine Department, Institute Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France; (L.L.); (E.B.)
| | - Ludovic Lacroix
- Department of Medical Oncology, Institute Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France;
| | | | - Eric Baudin
- Cancer Medicine Department, Institute Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France; (L.L.); (E.B.)
| | - Christine Do Cao
- Department of Endocrinology, Diabetology, Metabolism and Nutrition, Lille University Hospital, 59000 Lille, France; (A.J.); (B.C.); (C.D.C.)
| | - Julien Hadoux
- Cancer Medicine Department, Institute Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France; (L.L.); (E.B.)
- Correspondence: ; Tel.: +33-142116361
| |
Collapse
|
22
|
Lee M, Untch BR, Xu B, Ghossein R, Han C, Kuo F, Valero C, Nadeem Z, Patel N, Makarov V, Dogan S, Wong RJ, Sherman EJ, Ho AL, Chan TA, Fagin JA, Morris LGT. Genomic and Transcriptomic Correlates of Thyroid Carcinoma Evolution after BRAF Inhibitor Therapy. Mol Cancer Res 2022; 20:45-55. [PMID: 34635506 PMCID: PMC8738128 DOI: 10.1158/1541-7786.mcr-21-0442] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/12/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Targeted inhibition of BRAF V600E achieves tumor control in a subset of advanced thyroid tumors. Nearly all tumors develop resistance, and some have been observed to subsequently undergo dedifferentiation. The molecular alterations associated with thyroid cancer dedifferentiation in the setting of BRAF inhibition are unknown. We analyzed targeted next-generation sequencing data from 639 advanced, recurrent and/or metastatic thyroid carcinomas, including 15 tumors that were treated with BRAF inhibitor drugs and had tissue sampled during or posttreatment, 8 of which had matched pretherapy samples. Pre- and posttherapy tissues from one additional patient were profiled with whole-exome sequencing and RNA expression profiling. Mutations in genes comprising the SWI/SNF chromatin remodeling complex and the PI3K-AKT-mTOR, MAPK, and JAK-STAT pathways all increased in prevalence across more dedifferentiated thyroid cancer histologies. Of 7 thyroid cancers that dedifferentiated after BRAF inhibition, 6 had mutations in these pathways. These mutations were mostly absent from matched pretreatment samples and were rarely detected in tumors that did not dedifferentiate. Additional analyses in one of the vemurafenib-treated tumors before and after anaplastic transformation revealed the emergence of an oncogenic PIK3CA mutation, activation of ERK signaling, dedifferentiation, and development of an immunosuppressive tumor microenvironment. These findings validate earlier preclinical data implicating these genetic pathways in resistance to BRAF inhibitors, and suggest that genetic alterations mediating acquired drug resistance may also promote thyroid tumor dedifferentiation. IMPLICATIONS: The possibility that thyroid cancer dedifferentiation may be attributed to selective pressure applied by BRAF inhibitor-targeted therapy should be investigated further.
Collapse
Affiliation(s)
- Mark Lee
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian R Untch
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Catherine Han
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fengshen Kuo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina Valero
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zaineb Nadeem
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neal Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - James A Fagin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luc G T Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
23
|
Hou P, Wang YA. Conquering oncogenic KRAS and its bypass mechanisms. Theranostics 2022; 12:5691-5709. [PMID: 35966590 PMCID: PMC9373815 DOI: 10.7150/thno.71260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022] Open
Abstract
Aberrant activation of KRAS signaling is common in cancer, which has catalyzed heroic drug development efforts to target KRAS directly or its downstream signaling effectors. Recent works have yielded novel small molecule drugs with promising preclinical and clinical activities. Yet, no matter how a cancer is addicted to a specific target - cancer's genetic and biological plasticity fashions a variety of resistance mechanisms as a fait accompli, limiting clinical benefit of targeted interventions. Knowledge of these mechanisms may inform combination strategies to attack both oncogenic KRAS and subsequent bypass mechanisms.
Collapse
Affiliation(s)
- Pingping Hou
- Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103, USA.,Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA.,Lead contact
| | - Y Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
24
|
Chakraborty S, Carnazza M, Jarboe T, DeSouza N, Li XM, Moscatello A, Geliebter J, Tiwari RK. Disruption of Cell-Cell Communication in Anaplastic Thyroid Cancer as an Immunotherapeutic Opportunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:33-66. [PMID: 34888843 DOI: 10.1007/978-3-030-83282-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thyroid cancer incidence is increasing at an alarming rate, almost tripling every decade. About 44,280 new cases of thyroid cancer (12,150 in men and 32,130 in women) are estimated to be diagnosed in 2021, with an estimated death toll of around 2200. Although most thyroid tumors are treatable and associated with a favorable outcome, anaplastic thyroid cancer (ATC) is extremely aggressive with a grim prognosis of 6-9 months post-diagnosis. A large contributing factor to this aggressive nature is that ATC is completely refractory to mainstream therapies. Analysis of the tumor microenvironment (TME) associated with ATC can relay insight to the pathological realm that encompasses tumors and aids in cancer progression and proliferation. The TME is defined as a complex niche that surrounds a tumor and involves a plethora of cellular components whose secretions can modulate the environment in order to favor tumor progression. The cellular heterogeneity of the TME contributes to its dynamic function due to the presence of both immune and nonimmune resident, infiltrating, and interacting cell types. Associated immune cells discussed in this chapter include macrophages, dendritic cells (DCs), natural killer (NK) cells, and tumor-infiltrating lymphocytes (TILs). Nonimmune cells also play a role in the establishment and proliferation of the TME, including neuroendocrine (NE) cells, adipocytes, endothelial cells (ECs), mesenchymal stem cells (MSCs), and fibroblasts. The dynamic nature of the TME contributes greatly to cancer progression.Recent work has found ATC tissues to be defined by a T cell-inflamed "hot" tumor immune microenvironment (TIME) as evidenced by presence of CD3+ and CD8+ T cells. These tumor types are amenable to immune checkpoint blockade (ICB) therapy. This therapeutic avenue, as of 2021, has remained unexplored in ATC. New studies should seek to explore the therapeutic feasibility of a combination therapy, through the use of a small molecule inhibitor with ICB in ATC. Screening of in vitro model systems representative of papillary, anaplastic, and follicular thyroid cancer explored the expression of 29 immune checkpoint molecules. There are higher expressions of HVEM, BTLA, and CD160 in ATC cell lines when compared to the other TC subtypes. The expression level of HVEM was more than 30-fold higher in ATC compared to the others, on average. HVEM is a member of tumor necrosis factor (TNF) receptor superfamily, which acts as a bidirectional switch through interaction with BTLA, CD160, and LIGHT, in a cis or trans manner. Given the T cell-inflamed hot TIME in ATC, expression of HVEM on tumor cells was suggestive of a possibility for complex crosstalk of HVEM with inflammatory cytokines. Altogether, there is emerging evidence of a T cell-inflamed TIME in ATC along with the expression of immune checkpoint proteins HVEM, BTLA, and CD160 in ATC. This can open doors for combination therapies using small molecule inhibitors targeting downstream effectors of MAPK pathway and antagonistic antibodies targeting the HVEM/BTLA axis as a potentially viable therapeutic avenue for ATC patients. With this being stated, the development of adaptive resistance to targeted therapies is inevitable; therefore, using a combination therapy that targets the TIME can serve as a preemptive tactic against the characteristic therapeutic resistance that is seen in ATC. The dynamic nature of the TME, including the immune cells, nonimmune cells, and acellular components, can serve as viable targets for combination therapy in ATC. Understanding the complex interactions of these associated cells and the paradigm in which their secretions and components can serve as immunomodulators are critical points of understanding when trying to develop therapeutics specifically tailored for the anaplastic thyroid carcinoma microenvironment.
Collapse
Affiliation(s)
- Sanjukta Chakraborty
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA.,Weill Cornell Medicine, New York, NY, USA
| | - Michelle Carnazza
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Tara Jarboe
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Nicole DeSouza
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Xiu-Min Li
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | | | - Jan Geliebter
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Raj K Tiwari
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
25
|
Pani F, Yasuda Y, Di Dalmazi G, Chalan P, Gabrielson K, Adamo L, Sabini E, Mariotti S, Caturegli P. Pre-existing Thyroiditis Ameliorates Papillary Thyroid Cancer: Insights From a New Mouse Model. Endocrinology 2021; 162:6332851. [PMID: 34331442 PMCID: PMC8389179 DOI: 10.1210/endocr/bqab144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 01/27/2023]
Abstract
Papillary thyroid cancer (PTC) often co-occurs with Hashimoto's thyroiditis, an association that has long been reported in clinical studies yet remains controversial. Some studies, in fact, have suggested a protective effect of thyroiditis while others have not. We generated a mouse model where PTC and thyroiditis develop in a predictable manner, combining the oncogenic drive of the BRAFv600E mutation (inducible by tamoxifen) to the thyroiditis susceptibility of the NOD.H2h4 strain (inducible by iodine). A total of 113 NOD.H2h4_TPO-CRE-ER_BRAFV600E mice (50 followed throughout lifetime and 63 sacrificed at 16 weeks post tamoxifen) were used to determine whether the PTC phenotype differs when thyroiditis precedes or coincides with the onset of PTC. Mice with pre-existing thyroiditis lived longer (median survival of 28.2 weeks post tamoxifen) than those with concomitant (25.6 weeks) or no (24.5 weeks) thyroiditis (P < 0.01 by Laplace regression). PTC developed less frequently (33%) in the pre-existing thyroiditis group than the concomitant (100%) or no (100%) thyroiditis groups (P < 0.001 by chi-squared) and showed less aggressive histopathological features. The intratumoral mononuclear cell infiltration was more prominent in mice with pre-existing thyroiditis (P = 0.002 vs the other groups) and sustained by a significant expansion of effector memory CD8 + T cells and CD19 + B cells. These findings shed light on the controversial PTC-thyroiditis association and emphasize the contribution of intratumoral T and B lymphocytes to the evolution of PTC.
Collapse
Affiliation(s)
- Fabiana Pani
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Yoshinori Yasuda
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Giulia Di Dalmazi
- Division of Endocrinology, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Paulina Chalan
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, Pathology and Oncology and Environmental Health Engineering Johns Hopkins School of Medicine and Bloomberg School of Public Health, Baltimore, MD, USA
| | - Luigi Adamo
- Division of Cardiology, Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Elena Sabini
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stefano Mariotti
- Retired from Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Patrizio Caturegli
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Correspondence: Patrizio Caturegli, MD, MPH, Johns Hopkins Pathology, Ross Building, Room 656, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Valvo V, Iesato A, Kavanagh TR, Priolo C, Zsengeller Z, Pontecorvi A, Stillman IE, Burke SD, Liu X, Nucera C. Fine-Tuning Lipid Metabolism by Targeting Mitochondria-Associated Acetyl-CoA-Carboxylase 2 in BRAFV600E Papillary Thyroid Carcinoma. Thyroid 2021; 31:1335-1358. [PMID: 33107403 PMCID: PMC8558082 DOI: 10.1089/thy.2020.0311] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: BRAFV600E acts as an ATP-dependent cytosolic kinase. BRAFV600E inhibitors are widely available, but resistance to them is widely reported in the clinic. Lipid metabolism (fatty acids) is fundamental for energy and to control cell stress. Whether and how BRAFV600E impacts lipid metabolism regulation in papillary thyroid carcinoma (PTC) is still unknown. Acetyl-CoA carboxylase (ACC) is a rate-limiting enzyme for de novo lipid synthesis and inhibition of fatty acid oxidation (FAO). ACC1 and ACC2 genes encode distinct isoforms of ACC. The aim of our study was to determine the relationship between BRAFV600E and ACC in PTC. Methods: We performed RNA-seq and DNA copy number analyses in PTC and normal thyroid (NT) in The Cancer Genome Atlas samples. Validations were performed by using assays on PTC-derived cell lines of differing BRAF status and a xenograft mouse model derived from a heterozygous BRAFWT/V600E PTC-derived cell line with knockdown (sh) of ACC1 or ACC2. Results:ACC2 mRNA expression was significantly downregulated in BRAFV600E-PTC vs. BRAFWT-PTC or NT clinical samples. ACC2 protein levels were downregulated in BRAFV600E-PTC cell lines vs. the BRAFWT/WT PTC cell line. Vemurafenib increased ACC2 (and to a lesser extent ACC1) mRNA levels in PTC-derived cell lines in a BRAFV600E allelic dose-dependent manner. BRAFV600E inhibition increased de novo lipid synthesis rates, and decreased FAO due to oxygen consumption rate (OCR), and extracellular acidification rate (ECAR), after addition of palmitate. Only shACC2 significantly increased OCR rates due to FAO, while it decreased ECAR in BRAFV600E PTC-derived cells vs. controls. BRAFV600E inhibition synergized with shACC2 to increase intracellular reactive oxygen species production, leading to increased cell proliferation and, ultimately, vemurafenib resistance. Mice implanted with a BRAFWT/V600E PTC-derived cell line with shACC2 showed significantly increased tumor growth after vemurafenib treatment, while vehicle-treated controls, or shGFP control cells treated with vemurafenib showed stable tumor growth. Conclusions: These findings suggest a potential link between BRAFV600E and lipid metabolism regulation in PTC. BRAFV600E downregulates ACC2 levels, which deregulates de novo lipid synthesis, FAO due to OCR, and ECAR rates. ShACC2 may contribute to vemurafenib resistance and increased tumor growth. ACC2 rescue may represent a novel molecular strategy for overcoming resistance to BRAFV600E inhibitors in refractory PTC.
Collapse
Affiliation(s)
- Veronica Valvo
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI), Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Asumi Iesato
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI), Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Taylor R. Kavanagh
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carmen Priolo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Alfredo Pontecorvi
- Department of Medicine, Agostino Gemelli Medical School, UCSC, Rome, Italy
| | - Isaac E. Stillman
- Department of Pathology; Harvard Medical School, Boston, Massachusetts, USA
| | - Suzanne D. Burke
- Department of Medicine; Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaowen Liu
- Department of Emergency Medicine; Harvard Medical School, Boston, Massachusetts, USA
| | - Carmelo Nucera
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI), Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Center for Vascular Biology Research (CVBR); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Address correspondence to: Carmelo Nucera, MD, PhD, Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI) Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Office: RN270K, 99 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
27
|
Johansson K, Stenman A, Paulsson JO, Wang N, Ihre-Lundgren C, Zedenius J, Juhlin CC. Development of metastatic poorly differentiated thyroid cancer from a sub-centimeter papillary thyroid carcinoma in a young patient with a germline MET mutation - association or random chance? Thyroid Res 2021; 14:19. [PMID: 34389035 PMCID: PMC8364030 DOI: 10.1186/s13044-021-00110-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/21/2021] [Indexed: 11/10/2022] Open
Abstract
Background Thyroid cancer dedifferentiation is an unusual observation among young patients and is poorly understood, although a recent correlation to DICER1 gene mutations has been proposed. Case presentation A 28-year old patient presented with a sub-centimeter cytology-verified primary papillary thyroid carcinoma (PTC) and a synchronous lateral lymph node metastasis. Following surgery, histopathology confirmed a 9 mm oxyphilic PTC and a synchronous metastasis of poorly differentiated thyroid carcinoma (PDTC). Extensive molecular examinations of both lesions revealed wildtype DICER1 sequences, but identified a somatic ETV6-NTRK3 gene fusion and a MET germline variant (c.1076G > A, p.Arg359Gln). MET is an established oncogene known to be overexpressed in thyroid cancer, and this specific alteration was not reported as a single nucleotide polymorphism (SNP), suggestive of a mutation. Both the primary PTC and the metastatic PDTC displayed strong MET immunoreactivity. A validation cohort of 50 PTCs from young patients were analyzed using quantitative real-time PCR, revealing significantly higher MET gene expression in tumors than normal thyroid controls, a finding which was particularly pronounced in BRAF V600E mutated cases. No additional tumors apart from the index case harbored the p.Arg359Gln MET mutation. Transfecting PTC cell lines MDA-T32 and MDA-T41 with a p.Arg359Gln MET plasmid construct revealed no obvious effects on cellular migratory or invasive properties, whereas overexpression of wildtype MET stimulated invasion. Conclusions The question of whether the observed MET mutation in any way influenced the dedifferentiation of a primary PTC into a PDTC metastasis remains to be established. Moreover, our data corroborate earlier studies, indicating that MET is aberrantly expressed in PTC and may influence the invasive behavior of these tumors. Supplementary Information The online version contains supplementary material available at 10.1186/s13044-021-00110-4.
Collapse
Affiliation(s)
- Klara Johansson
- Department of Oncology-Pathology, Karolinska Institutet, BioClinicum J6:20, Visionsgatan 4, SE-17164, Solna, Sweden
| | - Adam Stenman
- Department of Oncology-Pathology, Karolinska Institutet, BioClinicum J6:20, Visionsgatan 4, SE-17164, Solna, Sweden.,Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Johan O Paulsson
- Department of Oncology-Pathology, Karolinska Institutet, BioClinicum J6:20, Visionsgatan 4, SE-17164, Solna, Sweden
| | - Na Wang
- Department of Oncology-Pathology, Karolinska Institutet, BioClinicum J6:20, Visionsgatan 4, SE-17164, Solna, Sweden.,Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Ihre-Lundgren
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jan Zedenius
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, BioClinicum J6:20, Visionsgatan 4, SE-17164, Solna, Sweden. .,Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
28
|
Luo H, Xia X, Kim GD, Liu Y, Xue Z, Zhang L, Shu Y, Yang T, Chen Y, Zhang S, Chen H, Zhang W, Li R, Tang H, Dong B, Fu X, Cheng W, Zhang W, Yang L, Peng Y, Dai L, Hu H, Jiang Y, Gong C, Hu Y, Zhu J, Li Z, Caulin C, Wei T, Park J, Xu H. Characterizing dedifferentiation of thyroid cancer by integrated analysis. SCIENCE ADVANCES 2021; 7:7/31/eabf3657. [PMID: 34321197 PMCID: PMC8318367 DOI: 10.1126/sciadv.abf3657] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/10/2021] [Indexed: 02/05/2023]
Abstract
Understanding of dedifferentiation, an indicator of poo prognosis for patients with thyroid cancer, has been hampered by imprecise and incomplete characterization of its heterogeneity and its attributes. Using single-cell RNA sequencing, we explored the landscape of thyroid cancer at single-cell resolution with 46,205 cells and delineated its dedifferentiation process and suppressive immune microenvironment. The developmental trajectory indicated that anaplastic thyroid cancer (ATC) cells were derived from a small subset of papillary thyroid cancer (PTC) cells. Moreover, a potential functional role of CREB3L1 on ATC development was revealed by integrated analyses of copy number alteration and transcriptional regulatory network. Multiple genes in differentiation-related pathways (e.g., EMT) were involved as the downstream targets of CREB3L1, increased expression of which can thus predict higher relapse risk of PTC. Collectively, our study provided insights into the heterogeneity and molecular evolution of thyroid cancer and highlighted the potential driver role of CREB3L1 in its dedifferentiation process.
Collapse
Affiliation(s)
- Han Luo
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuyang Xia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Gyeong Dae Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yang Liu
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhinan Xue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Lingyun Zhang
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China.,Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Yang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Shouyue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Haining Chen
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weihan Zhang
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruicen Li
- Health Promotion Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huairong Tang
- Health Promotion Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Birong Dong
- National Clinical Research Center for Geriatrics, Chengdu, Sichuan, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Wei Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Hongbo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yong Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yiguo Hu
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Jingqiang Zhu
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhihui Li
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Carlos Caulin
- Department of Otolaryngology-Head & Neck Surgery and University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Tao Wei
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.
| | - Heng Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China. .,Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Jiménez-Mora E, Gallego B, Díaz-Gago S, Lasa M, Baquero P, Chiloeches A. V600EBRAF Inhibition Induces Cytoprotective Autophagy through AMPK in Thyroid Cancer Cells. Int J Mol Sci 2021; 22:ijms22116033. [PMID: 34204950 PMCID: PMC8199856 DOI: 10.3390/ijms22116033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
The dysregulation of autophagy is important in the development of many cancers, including thyroid cancer, where V600EBRAF is a main oncogene. Here, we analyse the effect of V600EBRAF inhibition on autophagy, the mechanisms involved in this regulation and the role of autophagy in cell survival of thyroid cancer cells. We reveal that the inhibition of V600EBRAF activity with its specific inhibitor PLX4720 or the depletion of its expression by siRNA induces autophagy in thyroid tumour cells. We show that V600EBRAF downregulation increases LKB1-AMPK signalling and decreases mTOR activity through a MEK/ERK-dependent mechanism. Moreover, we demonstrate that PLX4720 activates ULK1 and increases autophagy through the activation of the AMPK-ULK1 pathway, but not by the inhibition of mTOR. In addition, we find that autophagy blockade decreases cell viability and sensitize thyroid cancer cells to V600EBRAF inhibition by PLX4720 treatment. Finally, we generate a thyroid xenograft model to demonstrate that autophagy inhibition synergistically enhances the anti-proliferative and pro-apoptotic effects of V600EBRAF inhibition in vivo. Collectively, we uncover a new role of AMPK in mediating the induction of cytoprotective autophagy by V600EBRAF inhibition. In addition, these data establish a rationale for designing an integrated therapy targeting V600EBRAF and the LKB1-AMPK-ULK1-autophagy axis for the treatment of V600EBRAF-positive thyroid tumours.
Collapse
Affiliation(s)
- Eva Jiménez-Mora
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Beatriz Gallego
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Sergio Díaz-Gago
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Marina Lasa
- Departamento de Bioquímica-Instituto de Investigaciones Biomédicas “Alberto Sols”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28029 Madrid, Spain;
| | - Pablo Baquero
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Antonio Chiloeches
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
- Correspondence:
| |
Collapse
|
30
|
When the MET receptor kicks in to resist targeted therapies. Oncogene 2021; 40:4061-4078. [PMID: 34031544 DOI: 10.1038/s41388-021-01835-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 02/04/2023]
Abstract
Although targeted therapies have increased the life expectancy of patients with druggable molecular alterations directly involved in tumor development, the efficacy of these therapies is limited by acquired resistances leading to treatment failure. Most targeted therapies, including ones exploiting therapeutic antibodies and kinase inhibitors, are directed against receptor tyrosine kinases (RTKs) or major signaling hubs. Resistances to these therapies arise when inhibition of these targets is bypassed through activation of alternative signaling pathways. In recent years, activation of the receptor tyrosine kinase MET has been shown to promote resistance to various targeted therapies. This casts MET as important actor in resistance. In this review, we describe how the MET receptor triggers resistance to targeted therapies against RTKs such as EGFR, VEGFR, and HER2 and against signaling hubs such as BRAF. We also describe how MET can be its own resistance factor, as illustrated by on-target resistance of lung tumors harboring activating mutations causing MET exon 14 skipping. Interestingly, investigation of all these situations reveals functional physiological relationships between MET and the target of the therapy to which the cancer becomes resistant, suggesting that resistance stems from preexisting mechanisms. Identification of MET as a resistance factor opens the way to co-treatment strategies that are being tested in current clinical trials.
Collapse
|
31
|
Bible KC, Kebebew E, Brierley J, Brito JP, Cabanillas ME, Clark TJ, Di Cristofano A, Foote R, Giordano T, Kasperbauer J, Newbold K, Nikiforov YE, Randolph G, Rosenthal MS, Sawka AM, Shah M, Shaha A, Smallridge R, Wong-Clark CK. 2021 American Thyroid Association Guidelines for Management of Patients with Anaplastic Thyroid Cancer. Thyroid 2021; 31:337-386. [PMID: 33728999 PMCID: PMC8349723 DOI: 10.1089/thy.2020.0944] [Citation(s) in RCA: 336] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background: Anaplastic thyroid cancer (ATC) is a rare but highly lethal form of thyroid cancer. Since the guidelines for the management of ATC by the American Thyroid Association were first published in 2012, significant clinical and scientific advances have occurred in the field. The aim of these guidelines is to inform clinicians, patients, and researchers on published evidence relating to the diagnosis and management of ATC. Methods: The specific clinical questions and topics addressed in these guidelines were based on prior versions of the guidelines, stakeholder input, and input of the Task Force members (authors of the guideline). Relevant literature was reviewed, including serial PubMed searches supplemented with additional articles. The American College of Physicians Guideline Grading System was used for critical appraisal of evidence and grading strength of recommendations. Results: The guidelines include the diagnosis, initial evaluation, establishment of treatment goals, approaches to locoregional disease (surgery, radiotherapy, targeted/systemic therapy, supportive care during active therapy), approaches to advanced/metastatic disease, palliative care options, surveillance and long-term monitoring, and ethical issues, including end of life. The guidelines include 31 recommendations and 16 good practice statements. Conclusions: We have developed evidence-based recommendations to inform clinical decision-making in the management of ATC. While all care must be individualized, such recommendations provide, in our opinion, optimal care paradigms for patients with ATC.
Collapse
Affiliation(s)
- Keith C. Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Electron Kebebew
- Stanford University, School of Medicine, Stanford, California, USA
| | - James Brierley
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Juan P. Brito
- Division of Diabetes, Endocrinology, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Maria E. Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Antonio Di Cristofano
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Robert Foote
- Department of Radiation Oncology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Thomas Giordano
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jan Kasperbauer
- Department of Otolaryngology, Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Kate Newbold
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, United Kingdom
| | - Yuri E. Nikiforov
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gregory Randolph
- Division of Thyroid and Parathyroid Endocrine Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - M. Sara Rosenthal
- Program for Bioethics and Markey Cancer Center Oncology Ethics Program, Departments Internal Medicine, Pediatrics and Behavioral Science, University of Kentucky, Lexington, Kentucky, USA
| | - Anna M. Sawka
- Division of Endocrinology, Department of Medicine, University Health Network and University of Toronto, Toronto, Canada
| | - Manisha Shah
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Ashok Shaha
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | |
Collapse
|
32
|
Chen B, Cai T, Huang C, Zang X, Sun L, Guo S, Wang Q, Chen Z, Zhao Y, Han Z, Xu R, Xu W, Wang M, Shen B, Zhu W. G6PD-NF-κB-HGF Signal in Gastric Cancer-Associated Mesenchymal Stem Cells Promotes the Proliferation and Metastasis of Gastric Cancer Cells by Upregulating the Expression of HK2. Front Oncol 2021; 11:648706. [PMID: 33718248 PMCID: PMC7952978 DOI: 10.3389/fonc.2021.648706] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Tumor-associated stromal cells have been widely recognized for their tumor-promoting capability involving paracrine signaling. However, the underlying mechanism and the effects of the molecules in the glycolysis pathway in gastric cancer-associated mesenchymal stem cells (GCMSCs) and gastric cancer cells on tumor progression remain unclear. Methods: The expression of hepatocyte growth factor (HGF) in GCMSCs and bone marrow mesenchymal stem cells (BMMSCs) was detected by enzyme-linked immunosorbent assay (ELISA). The effect of HGF derived from GCMSCs on the proliferation, metastasis, and HK2 expression of gastric cancer cells was evaluated in vitro and in vivo. The effects of G6PD on the production of HGF in mesenchymal stem cells (MSCs) were analyzed by immunoblotting. Results: HGF derived from GCMSCs promoted glycolysis, proliferation, and metastasis of gastric cancer by upregulating c-Myc-HK2 signal. The progression of the disease induced by GCMSCs decelerated in the absence of HK2. The expression of G6PD activated NF-κB signaling and stimulated the production of HGF in GCMSCs. Blocking HGF derived from GCMSCs decreased proliferation, metastasis, and angiogenesis of gastric cancer cells in vivo. Conclusions: GCMSCs highly expressed G6PD and facilitated the progression of gastric cancer through the G6PD-NF-κB-HGF axis coordinates. Blocking HGF derived from GCMSCs is a potential new therapeutic target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Bin Chen
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Tuo Cai
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chao Huang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xueyan Zang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Sun
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shuwei Guo
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qianqian Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhihong Chen
- Department of Surgery, Zhenjiang First People's Hospital, Zhenjiang, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhiqiang Han
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Rongman Xu
- Department of Clinical Laboratory, Haian People's Hospital, Haian, China
| | - Wenrong Xu
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Mei Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
33
|
Miller KC, Chintakuntlawar AV. Molecular-Driven Therapy in Advanced Thyroid Cancer. Curr Treat Options Oncol 2021; 22:24. [PMID: 33569661 DOI: 10.1007/s11864-021-00822-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 01/02/2023]
Abstract
OPINION STATEMENT With a growing understanding of the biologic drivers of different thyroid cancers, there is an ongoing revolution in the treatment of aggressive and advanced disease variants. This includes matching patients with specific point mutations or gene fusions to targeted therapies (e.g., selective RET inhibitors), delineating patients who are likely to respond to immune checkpoint inhibition (i.e., PD-L1-positive tumors) and even priming responses to traditional therapies such as radioactive iodine (via concomitant MAPK pathway inhibition). There is also a growing role for genomics in the prognostication of thyroid tumors to aid the adjudication of appropriate treatments. Taking stock of the current state of the field, recent successes should be celebrated, but there still remains a long road ahead to improve outcomes for patients, particularly for radioactive-iodine refractory differentiated thyroid cancer and anaplastic thyroid cancer. In this review, we summarize findings from recent clinical trials and highlight promising preclinical data supporting molecular-driven therapy in advanced thyroid cancer. Ultimately, enrollment in clinical trials remains paramount to the advancement of thyroid cancer care.
Collapse
Affiliation(s)
- Kevin C Miller
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
34
|
Han Y, Yu X, Yin Y, Lv Z, Jia C, Liao Y, Sun H, Liu T, Cong L, Fei Z, Fu D, Cong X, Qu S. Identification of Potential BRAF Inhibitor Joint Therapy Targets in PTC based on WGCAN and DCGA. J Cancer 2021; 12:1779-1791. [PMID: 33613767 PMCID: PMC7890315 DOI: 10.7150/jca.51551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/28/2020] [Indexed: 01/08/2023] Open
Abstract
As the most common mutation in papillary thyroid cancer (PTC), B-type Raf kinase V600E mutation (BRAFV600E ) has become an important target for the clinical treatment of PTC. However, the clinical application still faces the problem of resistance to BRAF inhibitors (BRAFi). Therefore, exploring BRAFV600E-associated prognostic factors to providing potential joint targets is important for combined targeted therapy with BRAFi. In this study, we combined transcript data and clinical information from 199 BRAF wild-type (BRAFWT ) patients and 283 BRAFV600E mutant patients collected from The Cancer Genome Atlas (TCGA), and screened 455 BRAFV600E- associated genes through differential analysis and weighted gene co-expression network analysis. Based on these BRAFV600E -associated genes, we performed functional enrichment analysis and co-expression differential analysis and constructed a core co-expression network. Next, genes in the differential co-expression network were used to predict drugs for therapy in the crowd extracted expression of differential signatures (CREEDS) database, and the key genes were selected based on the hub co-expression network through survival analyses and receiver operating characteristic (ROC) curve analyses. Finally, we obtained eight BRAFV600E -associated biomarkers with both prognostic and diagnostic values as potential BRAFi joint targets, including FN1, MET, SLC34A2, NGEF, TBC1D2, PLCD3, PROS1, and NECTIN4. Among these genes, FN1, MET, PROS1, and TBC1D2 were validated through GEO database. Two novel biomarkers, PROS1 and TBC1D2, were further validated by qRT-PCR experiment. Besides, we obtained four potential targeted drugs that could be used in combination with BRAFi to treat PTC, including MET inhibitor, ERBB3 inhibitor, anti-NaPi2b antibody-drug conjugate, and carboplatin through literature review. The study provided potential drug targets for combination therapy with BRAFi for PTC to overcome the drug resistance for BRAFi.
Collapse
Affiliation(s)
- YaLi Han
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| | - XiaQing Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - YuZhen Yin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhongwei Lv
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - ChengYou Jia
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| | - Yina Liao
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| | - Hongyan Sun
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Tie Liu
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Lele Cong
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - ZhaoLiang Fei
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xianling Cong
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Shen Qu
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| |
Collapse
|
35
|
Wang J, Pollard K, Calizo A, Pratilas CA. Activation of Receptor Tyrosine Kinases Mediates Acquired Resistance to MEK Inhibition in Malignant Peripheral Nerve Sheath Tumors. Cancer Res 2020; 81:747-762. [PMID: 33203698 DOI: 10.1158/0008-5472.can-20-1992] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/08/2020] [Accepted: 11/12/2020] [Indexed: 11/16/2022]
Abstract
Malignant peripheral nerve sheath tumors often arise in patients with neurofibromatosis type 1 and are among the most treatment-refractory types of sarcoma. Overall survival in patients with relapsed disease remains poor, and thus novel therapeutic approaches are needed. NF1 is essential for negative regulation of RAS activity and is altered in about 90% of malignant peripheral nerve sheath tumors (MPNST). A complex interplay of upstream signaling and parallel RAS-driven pathways characterizes NF1-driven tumorigenesis, and inhibiting more than one RAS effector pathway is therefore necessary. To devise potential combination therapeutic strategies, we identified actionable alterations in signaling that underlie adaptive and acquired resistance to MEK inhibitor (MEKi). Using a series of proteomic, biochemical, and genetic approaches in an in vitro model of MEKi resistance provided a rationale for combination therapies. HGF/MET signaling was elevated in the MEKi-resistant model. HGF overexpression conferred resistance to MEKi in parental cells. Depletion of HGF or MET restored sensitivity of MEKi-resistant cells to MEKi. Finally, a combination of MEK and MET inhibition demonstrated activity in models of MPNST and may therefore be effective in patients with MPNST harboring genetic alterations in NF1. SIGNIFICANCE: This study demonstrates that MEKi plus MET inhibitor may delay or prevent a novel mechanism of acquired MEKi resistance, with clinical implications for MPNST patients harboring NF1 alterations.
Collapse
Affiliation(s)
- Jiawan Wang
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kai Pollard
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ana Calizo
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine A Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
36
|
French JD. Immunotherapy for advanced thyroid cancers - rationale, current advances and future strategies. Nat Rev Endocrinol 2020; 16:629-641. [PMID: 32839578 DOI: 10.1038/s41574-020-0398-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
In the past decade, the field of cancer immunotherapy has been revolutionized by immune checkpoint blockade (ICB) technologies. Success across a broad spectrum of cancers has led to a paradigm shift in therapy for patients with advanced cancer. Early data are now accumulating in progressive thyroid cancers treated with single-agent ICB therapies and combination approaches that incorporate ICB technologies. This Review discusses our current knowledge of the immune response in thyroid cancers, the latest and ongoing immune-based approaches, and the future of immunotherapies in thyroid cancer. Physiologically relevant preclinical mouse models and human correlative research studies will inform development of the next stage of immune-based therapies for patients with advanced thyroid cancer.
Collapse
Affiliation(s)
- Jena D French
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Denver, Aurora, CO, USA.
- University of Colorado Cancer Center, University of Colorado Denver, Aurora, CO, USA.
| |
Collapse
|
37
|
Yuan J, Dong X, Yap J, Hu J. The MAPK and AMPK signalings: interplay and implication in targeted cancer therapy. J Hematol Oncol 2020; 13:113. [PMID: 32807225 PMCID: PMC7433213 DOI: 10.1186/s13045-020-00949-4] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is characterized as a complex disease caused by coordinated alterations of multiple signaling pathways. The Ras/RAF/MEK/ERK (MAPK) signaling is one of the best-defined pathways in cancer biology, and its hyperactivation is responsible for over 40% human cancer cases. To drive carcinogenesis, this signaling promotes cellular overgrowth by turning on proliferative genes, and simultaneously enables cells to overcome metabolic stress by inhibiting AMPK signaling, a key singular node of cellular metabolism. Recent studies have shown that AMPK signaling can also reversibly regulate hyperactive MAPK signaling in cancer cells by phosphorylating its key components, RAF/KSR family kinases, which affects not only carcinogenesis but also the outcomes of targeted cancer therapies against the MAPK signaling. In this review, we will summarize the current proceedings of how MAPK-AMPK signalings interplay with each other in cancer biology, as well as its implications in clinic cancer treatment with MAPK inhibition and AMPK modulators, and discuss the exploitation of combinatory therapies targeting both MAPK and AMPK as a novel therapeutic intervention.
Collapse
Affiliation(s)
- Jimin Yuan
- Department of Urology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Geriatric Department, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Xiaoduo Dong
- Shenzhen People's Hospital, 1017 Dongmen North Road, Shenzhen, 518020, China
| | - Jiajun Yap
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Jiancheng Hu
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore.
| |
Collapse
|
38
|
Lin B, Lu B, Hsieh IY, Liang Z, Sun Z, Yi Y, Lv W, Zhao W, Li J. Synergy of GSK-J4 With Doxorubicin in KRAS-Mutant Anaplastic Thyroid Cancer. Front Pharmacol 2020; 11:632. [PMID: 32477122 PMCID: PMC7239034 DOI: 10.3389/fphar.2020.00632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Anaplastic thyroid cancer is the most aggressive thyroid cancer and has a poor prognosis. At present, there is no effective treatment for it. Methods Here, we used different concentrations of GSK-J4 or a combination of GSK-J4 and doxorubicin to treat human Cal-62, 8505C, and 8305C anaplastic thyroid cancer (ATC) cell lines. The in vitro experiments were performed using cell viability assays, cell cycle assays, annexin-V/PI binding assays, Transwell migration assays, and wound-healing assays. Tumor xenograft models were used to observe effects in vivo. Results The half maximal inhibitory concentration (IC50) of GSK-J4 in Cal-62 cells was 1.502 μM, and as the dose of GSK-J4 increased, more ATC cells were blocked in the G2-M and S stage. The combination of GSK-J4 and doxorubicin significantly increased the inhibitory effect on proliferation, especially in KRAS-mutant ATC cells in vivo (inhibition rate 38.0%) and in vitro (suppresses rate Fa value 0.624, CI value 0.673). The invasion and migration abilities of the KRAS-mutant cell line were inhibited at a low concentration (p < 0.05). Conclusions The combination of GSK-J4 with doxorubicin in KRAS-mutant ATC achieved tumor-suppressive effects at a low dose. The synergy of the combination of GSK-J4 and doxorubicin may make it an effective chemotherapy regimen for KRAS-mutant ATC.
Collapse
Affiliation(s)
- Bo Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bing Lu
- Institute of Urology of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - I-Yun Hsieh
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Liang
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zicheng Sun
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Yi
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Weiming Lv
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
39
|
Pozdeyev N, Rose MM, Bowles DW, Schweppe RE. Molecular therapeutics for anaplastic thyroid cancer. Semin Cancer Biol 2020; 61:23-29. [PMID: 31991166 DOI: 10.1016/j.semcancer.2020.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Anaplastic thyroid cancer (ATC) represents one of the most lethal human cancers and although this tumor type is rare, ATC accounts for the majority of deaths from thyroid cancer. Due to the rarity of ATC, a comprehensive genomic characterization of this tumor type has been challenging, and thus the development of new therapies has been lacking. To date, there is only one mutation-driven targeted therapy for BRAF-mutant ATC. Recent genomic studies have used next generation sequencing to define the genetic landscape of ATC in order to identify new therapeutic targets. Together, these studies have confirmed the role of oncogenic mutations of MAPK pathway as key drivers of differentiated thyroid cancer (BRAF, RAS), and that additional genetic alterations in the PI3K pathway, TP53, and the TERT promoter are necessary for anaplastic transformation. Recent novel findings have linked the high mutational burden associated with ATC with mutations in the Mismatch Repair (MMR) pathway and overactivity of the AID/APOBEC family of cytidine deaminases. Additional novel mutations include cell cycle genes, SWI/SNF chromatin remodeling complex, and histone modification genes. Mutations in RAC1 were also identified in ATC, which have important implications for BRAF-directed therapies. In this review, we summarize these novel findings and the new genetic landscape of ATC. We further discuss the development of therapies targeting these pathways that are being tested in clinical and preclinical studies.
Collapse
Affiliation(s)
- Nikita Pozdeyev
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA; Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, Aurora, CO, USA
| | - Madison M Rose
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA
| | - Daniel W Bowles
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA.
| |
Collapse
|
40
|
The Aryl Hydrocarbon Receptor Is Expressed in Thyroid Carcinoma and Appears to Mediate Epithelial-Mesenchymal-Transition. Cancers (Basel) 2020; 12:cancers12010145. [PMID: 31936153 PMCID: PMC7016998 DOI: 10.3390/cancers12010145] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 12/22/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) is expected to promote initiation, progression and invasion of cancer cells regulating proliferation, differentiation, gene expression, inflammation, cell motility and migration. Furthermore, an immunosuppressant function of AhR has been recognized. This study evaluated AhR expression and its role in thyroid cancer progression. AhR expression was assessed by qPCR in 107 thyroid cancer samples (90 PTCs, 11 MTCs, 6 ATCs), and by immunohistochemistry in 41 PTCs. To estimate receptor activation, the expression of target genes CYP1A1 and CYP1B1 was measured. AhR functional effects were evaluated in kynurenine-stimulated FTC-133 and BcPap cell lines by analyzing the expression of genes involved in EMT and cell motility. AhR mRNA expression resulted significantly higher in all the analyzed thyroid cancer samples compared to normal thyroid and a statistically significant correlation with CYP1B1 was detected. Kynurenine-stimulated FTC-133 and BcPap showed the activation of a specific AhR-driven EMT program characterized by E-cadherin decrease and SLUG, N-cadherin and fibronectin increase, resulting in boost of cell motility and invasion. This study confirmed the importance of the IDO1-Kyn-AhR pathway in thyroid cancer tumorigenesis, suggesting an AhR pivotal role in mediating an immunosuppressive microenvironment and favoring the acquisition of a mesenchymal phenotype that could promote invasiveness and metastasis.
Collapse
|
41
|
Garcia C, Cormier F, Groussin L. Genetics of Human Thyroid Cancer Cell Lines-Letter. Clin Cancer Res 2019; 25:6882. [PMID: 31732664 DOI: 10.1158/1078-0432.ccr-19-2107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/11/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Cyril Garcia
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Hôpital d'Instruction des Armées BEGIN, Saint-Mandé, France
| | - Françoise Cormier
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lionel Groussin
- INSERM Unité 1016, Institut Cochin, Paris, France. .,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Endocrinology, APHP, Cochin Hospital, Paris, France
| |
Collapse
|
42
|
Landa I, Pozdeyev N, Knauf JA, Haugen BR, Fagin JA, Schweppe RE. Genetics of Human Thyroid Cancer Cell Lines-Response. Clin Cancer Res 2019; 25:6883-6884. [PMID: 31732665 DOI: 10.1158/1078-0432.ccr-19-2531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Iñigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikita Pozdeyev
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, Aurora, Colorado
| | - Jeffrey A Knauf
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bryan R Haugen
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, Aurora, Colorado
| | - James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|
43
|
Duan H, Li Y, Hu P, Gao J, Ying J, Xu W, Zhao D, Wang Z, Ye J, Lizaso A, He Y, Wu H, Liang Z. Mutational profiling of poorly differentiated and anaplastic thyroid carcinoma by the use of targeted next-generation sequencing. Histopathology 2019; 75:890-899. [PMID: 31230400 DOI: 10.1111/his.13942] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/18/2019] [Indexed: 02/04/2023]
Abstract
AIMS To characterise the mutational profiles of poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) and to identify markers with potential diagnostic, prognostic and therapeutic significance. METHODS AND RESULTS Targeted next-generation sequencing with a panel of 18 thyroid carcinoma-related genes was performed on tissue samples from 41 PDTC and 25 ATC patients. Genetic alterations and their correlations with clinicopathological factors, including survival outcomes, were also analysed. Our results showed that ATC had significantly higher mutation rates of BRAF, TP53, TERT and PIK3CA than PDTC (P = 0.005, P = 0.007, P = 0.005, and P = 0.033, respectively). Nine (69%) ATC cases with papillary thyroid carcinoma (PTC) components harboured BRAF mutations, all of which coexisted with a late mutation event (TP53, TERT, or PIK3CA). Nine cases with oncogenic fusion (six RET cases, one NTRK1 case, one ALK case, and one PPARG case) were identified in 41 PDTCs, whereas only one case with oncogenic fusion (NTRK1) was found among 25 ATCs. Moreover, all six cases of RET fusion were found in PDTC with PTC components, accounting for 33%. In PDTC/ATC patients, concurrent TERT and PIK3CA mutations were associated with poor overall survival after adjustment for TNM stage (P = 0.001). CONCLUSIONS ATC with PTC components is typically characterised by a BRAF mutation with a late mutation event, whereas PDTC with PTC components is more closely correlated with RET fusion. TERT and concurrent PIK3CA mutations predict worse overall survival in PDTC/ATC patients.
Collapse
Affiliation(s)
- Huanli Duan
- Department of Pathology, Molecular Pathology Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Pathology, National Cancer Centre/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peizhen Hu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jie Gao
- Department of Pathology, Molecular Pathology Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Centre/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanni Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Danhui Zhao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhe Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Junyi Ye
- Burning Rock Biotech, Guangzhou, China
| | | | - Yangzhige He
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huanwen Wu
- Department of Pathology, Molecular Pathology Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhiyong Liang
- Department of Pathology, Molecular Pathology Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Nikitski AV, Rominski SL, Condello V, Kaya C, Wankhede M, Panebianco F, Yang H, Altschuler DL, Nikiforov YE. Mouse Model of Thyroid Cancer Progression and Dedifferentiation Driven by STRN-ALK Expression and Loss of p53: Evidence for the Existence of Two Types of Poorly Differentiated Carcinoma. Thyroid 2019; 29:1425-1437. [PMID: 31298630 PMCID: PMC6797076 DOI: 10.1089/thy.2019.0284] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Thyroid tumor progression from well-differentiated cancer to poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) involves step-wise dedifferentiation associated with loss of iodine avidity and poor outcomes. ALK fusions, typically STRN-ALK, are found with higher incidence in human PDTC compared with well-differentiated cancer and, as previously shown, can drive the development of murine PDTC. The aim of this study was to evaluate thyroid cancer initiation and progression in mice with concomitant expression of STRN-ALK and inactivation of the tumor suppressor p53 (Trp53) in thyroid follicular cells. Methods: Transgenic mice with thyroid-specific expression of STRN-ALK and biallelic p53 loss were generated and aged on a regular diet or with methimazole and sodium perchlorate goitrogen treatment. Development and progression of thyroid tumors were monitored by using ultrasound imaging, followed by detailed histological and immunohistochemical evaluation. Gene expression analysis was performed on selected tumor samples by using RNA-Seq and quantitative RT-PCR. Results: In mice treated with goitrogen, the first thyroid cancers appeared at 6 months of age, reaching 86% penetrance by the age of 12 months, while a similar rate (71%) of tumor occurrence in mice on regular diet was observed by 18 months of age. Histological examination revealed well-differentiated papillary thyroid carcinomas (PTC) (n = 26), PDTC (n = 21), and ATC (n = 8) that frequently coexisted in the same thyroid gland. The tumors were frequently lethal and associated with the development of lung metastasis in 24% of cases. Histological and immunohistochemical characteristics of these cancers recapitulated tumors seen in humans. Detailed analysis of PDTC revealed two tumor types with distinct cell morphology and immunohistochemical characteristics, designated as PDTC type 1 (PDTC1) and type 2 (PDTC2). Gene expression analysis showed that PDTC1 tumors retained higher expression of thyroid differentiation genes including Tg and Slc5a5 (Nis) as compared with PDTC2 tumors. Conclusions: In this study, we generated a new mouse model of multistep thyroid cancer dedifferentiation with evidence of progression from PTC to PDTC and ATC. Further, PDTC in these mice showed two distinct histologic appearances correlated with levels of expression of thyroid differentiation and iodine metabolism genes, suggesting a possibility of existence of two PDTC types with different functional characteristics and potential implication for therapeutic approaches to these tumors.
Collapse
MESH Headings
- Anaplastic Lymphoma Kinase/genetics
- Animals
- Antithyroid Agents/toxicity
- Calmodulin-Binding Proteins/genetics
- Cell Dedifferentiation/genetics
- Cell Differentiation/genetics
- Disease Models, Animal
- Disease Progression
- Membrane Proteins/genetics
- Methimazole/toxicity
- Mice
- Mice, Knockout
- Mice, Transgenic
- Nerve Tissue Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Perchlorates/toxicity
- RNA-Seq
- Sodium Compounds/toxicity
- Symporters/genetics
- Thyroglobulin/genetics
- Thyroid Cancer, Papillary/chemically induced
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/pathology
- Thyroid Carcinoma, Anaplastic/chemically induced
- Thyroid Carcinoma, Anaplastic/genetics
- Thyroid Carcinoma, Anaplastic/pathology
- Thyroid Neoplasms/chemically induced
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/pathology
- Transcriptome
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
| | - Susan L. Rominski
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vincenzo Condello
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Cihan Kaya
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mamta Wankhede
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Hong Yang
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel L. Altschuler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yuri E. Nikiforov
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Address correspondence to: Yuri E. Nikiforov, MD, PhD, Department of Pathology, University of Pittsburgh, CLB Room 8031, 3477 Euler Way, Pittsburgh, PA 15213
| |
Collapse
|
45
|
Knippler CM, Saji M, Rajan N, Porter K, La Perle KMD, Ringel MD. MAPK- and AKT-activated thyroid cancers are sensitive to group I PAK inhibition. Endocr Relat Cancer 2019; 26:699-712. [PMID: 31146260 PMCID: PMC7062234 DOI: 10.1530/erc-19-0188] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 12/25/2022]
Abstract
The number of individuals who succumb to thyroid cancer has been increasing and those who are refractory to standard care have limited therapeutic options, highlighting the importance of developing new treatments for patients with aggressive forms of the disease. Mutational activation of MAPK signaling, through BRAF and RAS mutations and/or gene rearrangements, and activation of PI3K signaling, through mutational activation of PIK3CA or loss of PTEN, are well described in aggressive thyroid cancer. We previously reported overactivation and overexpression of p21-activated kinases (PAKs) in aggressive human thyroid cancer invasive fronts and determined that PAK1 functionally regulated thyroid cancer cell migration. We reported mechanistic crosstalk between the MAPK and PAK pathways that are BRAF-dependent but MEK independent, suggesting that PAK and MEK inhibition might be synergistic. In the present study, we tested this hypothesis. Pharmacologic inhibition of group I PAKs using two PAK kinase inhibitors, G-5555 or FRAX1036, reduced thyroid cancer cell viability, cell cycle progression and migration and invasion, with greater potency for G-5555. Combination of G-5555 with vemurafenib was synergistic in BRAFV600E-mutated thyroid cancer cell lines. Finally, G-5555 restrained thyroid size of BRAFV600E-driven murine papillary thyroid cancer by >50% (P < 0.0001) and reduced carcinoma formation (P = 0.0167), despite maintenance of MAPK activity. Taken together, these findings suggest both that group I PAKs may be a new therapeutic target for thyroid cancer and that PAK activation is functionally important for BRAFV600E-mediated thyroid cancer development.
Collapse
Affiliation(s)
- Christina M. Knippler
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University Wexner Medical Center and Arthur G. James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Motoyasu Saji
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University Wexner Medical Center and Arthur G. James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Neel Rajan
- College of Arts and Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Kyle Porter
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, USA
| | - Krista M. D. La Perle
- Department of Veterinary Biosciences, Comparative Pathology & Mouse Phenotyping Shared Resource, The Ohio State University, Columbus, Ohio, USA
| | - Matthew D. Ringel
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University Wexner Medical Center and Arthur G. James Comprehensive Cancer Center, Columbus, Ohio, USA
- To whom correspondence should be addressed: Matthew D. Ringel, MD, Ralph W. Kurtz Professor of Medicine, Director, Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University College of Medicine & Comprehensive Cancer Center, McCampbell Hall, Room 565, 1581 Dodd Drive, Columbus, OH 43210, Tel: 614-685-3333,
| |
Collapse
|
46
|
Garcia C, Buffet C, El Khattabi L, Rizk-Rabin M, Perlemoine K, Ragazzon B, Bertherat J, Cormier F, Groussin L. MET overexpression and activation favors invasiveness in a model of anaplastic thyroid cancer. Oncotarget 2019; 10:2320-2334. [PMID: 31040922 PMCID: PMC6481343 DOI: 10.18632/oncotarget.26798] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 03/04/2019] [Indexed: 11/25/2022] Open
Abstract
In thyroid cancers, MET receptor overexpression has been associated with higher risk of metastatic progression. In this study, it was shown that the anaplastic thyroid cancer (ATC)-derived TTA1 cell line overexpressed MET. By using FISH and relative quantification by qPCR, it was demonstrated that this overexpression resulted from a MET amplification with more than 20 copies. As expected, MET overexpression led to its constitutive activation and upregulated signaling towards the MAPK, PI3K/AKT, STAT3 and NF-κB pathways. Since the usual feature of MET-amplified cell lines is the "MET addiction" for their cell proliferation, the effect of the highly selective ATP competitive MET inhibitor PHA665752 was analyzed. While PHA665752 strongly inhibited the MAPK pathway, it did not reduce cell proliferation in TTA1 cells (IC50 = 4100 nM). This resistance to PHA665752 of the TTA1 cell line was demonstrated to be related to EGFR-MET functional cross-talk and PI3K/AKT and NF-κB signaling. Nevertheless, PHA665752 suppressed the anchorage-independent growth capacity of the TTA1 cell line and reduced cell migration and invasion in a transwell assay. The role of activated MET in these neoplastic properties of the TTA1 cells was also proved with si-MET-RNA targeting. Thus, this work highlights the TTA1 cell line as the first model of MET amplification in an ATC cell line, which leads to MET constitutive activation and underlies its neoplastic properties. Besides being a useful model for MET inhibitors screening, the TTA1 cell line also supports the argument for searching for MET amplification in ATC, as it could have therapeutic implications.
Collapse
Affiliation(s)
- Cyril Garcia
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Hôpital d'Instruction des Armées BEGIN, Saint-Mandé, France
| | - Camille Buffet
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Laila El Khattabi
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Cytogenetics Laboratory, APHP, Cochin Hospital, Paris, France
| | - Marthe Rizk-Rabin
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Karine Perlemoine
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bruno Ragazzon
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jérôme Bertherat
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Endocrinology, APHP, Cochin Hospital, Paris, France
| | - Françoise Cormier
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lionel Groussin
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Endocrinology, APHP, Cochin Hospital, Paris, France
| |
Collapse
|
47
|
Zhang Y, Dong J, Shi R, Feng L, Li Y, Cheng C, Zhang L, Song B, Bi Y, Huang H, Kong P, Guo J, Liu J. Mps1 is associated with the BRAF V600E mutation and predicts poor outcome in patients with colorectal cancer. Oncol Lett 2019; 17:2809-2817. [PMID: 30854056 PMCID: PMC6365956 DOI: 10.3892/ol.2019.9924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/08/2018] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) with the V600E mutation of B-Raf proto-oncogene serine/threonine kinase (BRAFV600E) mutation is insensitive to chemotherapy and is indicative of a poor patient prognosis. Although BRAF inhibitors have a marked effect on malignant melanoma harboring the BRAFV600E mutation, they have a limited effect on patients with CRC with the same BRAF mutation. A previous study identified a novel gene, monopolar spindle protein kinase 1 (Mps1), a downstream target of BRAFV600E only, rather than of wild-type BRAF as well, which contributes to tumorigenesis in melanoma. In the present study, the incidence of BRAFV600E in patients with CRC was identified and the correlation of Mps1, phospho-extracellular-signal-regulated kinase (p-ERK) and BRAFV600E was investigated. The results indicated that the mutation rate of BRAFV600E was 5.2% in CRC. Poorly differentiated tumors and mucinous tumors have a significantly higher incidence of BRAFV600E compared with well-differentiated tumors and non-mucinous tumors (P<0.05). Kaplan-Meier survival analysis indicated that the survival rate was markedly lower in patients with BRAFV600E compared with in patients with wild-type BRAF (BRAFWT). The expression of p-ERK and Mps1 in CRC with BRAFV600E was significantly higher compared with in CRC with BRAFWT (P<0.05), and their expression is associated with cancer classification, degree of differentiation and lymph node transfusion (P<0.05). In addition p-ERK expression was positively correlated with Mps1 expression, with a contingency coefficient of 0.679 (P=0.002). In conclusion, the results of the present study indicated that Mps1 was significantly associated with BRAFV600E/p-ERK and may serve a crucial function in the development of CRC. The results of the present study raise the possibility that targeting the oncogenic BRAF and Mps1, particularly when in conjunction, could provide promising therapeutic opportunities for the treatment of CRC.
Collapse
Affiliation(s)
- Yanyan Zhang
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jinyao Dong
- Endoscopy Center, Shanxi Cancer Hospital, Taiyuan, Shanxi 30013, P.R. China
| | - Ruyi Shi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Cell Biology and Genetics, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Liguo Feng
- Department of General Surgery, Taiyuan Municipal No. 2 People's Hospital, Taiyuan, Shanxi 030002, P.R. China
| | - Yike Li
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Caixia Cheng
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Pathology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ling Zhang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Bin Song
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Oncology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yanghui Bi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - He Huang
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Pengzhou Kong
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jiansheng Guo
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jing Liu
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
48
|
Landa I, Knauf JA. Mouse Models as a Tool for Understanding Progression in Braf V600E-Driven Thyroid Cancers. Endocrinol Metab (Seoul) 2019; 34:11-22. [PMID: 30784243 PMCID: PMC6435851 DOI: 10.3803/enm.2019.34.1.11] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/12/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
Abstract
The development of next generation sequencing (NGS) has led to marked advancement of our understanding of genetic events mediating the initiation and progression of thyroid cancers. The NGS studies have confirmed the previously reported high frequency of mutually-exclusive oncogenic alterations affecting BRAF and RAS proto-oncogenes in all stages of thyroid cancer. Initially identified by traditional sequencing approaches, the NGS studies also confirmed the acquisition of alterations that inactivate tumor protein p53 (TP53) and activate phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) in advanced thyroid cancers. Novel alterations, such as those in telomerase reverse transcriptase (TERT) promoter and mating-type switching/sucrose non-fermenting (SWI/SNF) complex, are also likely to promote progression of the BRAFV600E-driven thyroid cancers. A number of genetically engineered mouse models (GEMM) of BRAFV600E-driven thyroid cancer have been developed to investigate thyroid tumorigenesis mediated by oncogenic BRAF and to explore the role of genetic alterations identified in the genomic analyses of advanced thyroid cancer to promote tumor progression. This review will discuss the various GEMMs that have been developed to investigate oncogenic BRAFV600E-driven thyroid cancers.
Collapse
Affiliation(s)
- Iñigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey A Knauf
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
49
|
Saini S, Maker AV, Burman KD, Prabhakar BS. Molecular aberrations and signaling cascades implicated in the pathogenesis of anaplastic thyroid cancer. Biochim Biophys Acta Rev Cancer 2018; 1872:188262. [PMID: 30605717 DOI: 10.1016/j.bbcan.2018.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 01/16/2023]
Abstract
Anaplastic Thyroid Cancer (ATC) accounts for >40% thyroid cancer-related deaths and has a dismal prognosis. In the past decade, significant efforts have been made towards understanding the pathogenesis of this disease and developing novel therapeutics. Unfortunately, effective treatment is still lacking and a more thorough understanding of ATC pathogenesis may provide new opportunities to improve ATC therapeutics. This review provides insights into ATC clinical presentation and pathology, and the putative role of genetic aberrations and alterations in molecular signaling pathways in ATC pathogenesis. We reviewed prevalent mutations, chromosomal abnormalities and fusions, epigenetic alterations and dysregulations in ATC, and highlighted several signaling cascades which appeared to be integral to ATC pathogenesis. Moreover, these features offer insights into de-differentiated, aggressive and drug-resistant phenotype of ATC, and thus may help in exploring potential new molecular targets for developing novel therapeutics.
Collapse
Affiliation(s)
- Shikha Saini
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, United States
| | - Ajay V Maker
- Department of Surgery, Division of Surgical Oncology, University of Illinois-College of Medicine, Chicago, IL, United States
| | - Kenneth D Burman
- Medstar Washington Hospital Medical Center, Washington, DC, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, United States; Jesse Brown VA Medical Center, Chicago, IL, United States.
| |
Collapse
|
50
|
Damalanka VC, Han Z, Karmakar P, O’Donoghue AJ, La Greca F, Kim T, Pant SM, Helander J, Klefström J, Craik CS, Janetka JW. Discovery of Selective Matriptase and Hepsin Serine Protease Inhibitors: Useful Chemical Tools for Cancer Cell Biology. J Med Chem 2018; 62:480-490. [DOI: 10.1021/acs.jmedchem.8b01536] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Vishnu C. Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, 63110, United States
| | - Zhenfu Han
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, 63110, United States
| | - Partha Karmakar
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, 63110, United States
| | - Anthony J. O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California, 92093, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, 94158, United States
| | - Florencia La Greca
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, 94158, United States
| | - Tommy Kim
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, 63110, United States
| | - Shishir M. Pant
- Cancer Cell Circuitry Laboratory, Research Programs Unit/Translational Cancer Biology & Medicum, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014 Helsinki, Finland
| | - Jonathan Helander
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, 63110, United States
| | - Juha Klefström
- Cancer Cell Circuitry Laboratory, Research Programs Unit/Translational Cancer Biology & Medicum, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014 Helsinki, Finland
| | - Charles S. Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, 94158, United States
| | - James W. Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, 63110, United States
| |
Collapse
|