Next Article in Journal
Proteases: Role and Function in Cancer
Previous Article in Journal
Profiling of Transcriptome-Wide N6-Methyladenosine (m6A) Modifications and Identifying m6A Associated Regulation in Sperm Tail Formation in Anopheles sinensis
Previous Article in Special Issue
Clinicopathological and Molecular Features of Secondary Cancer (Metastasis) to the Thyroid and Advances in Management
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Recent Advances in Thyroid Cancer Research

1
Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus, Denmark
2
Department of Microgravity and Translational Regenerative Medicine, University Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Pfälzer Str. 2, 39106 Magdeburg, Germany
3
Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
Int. J. Mol. Sci. 2022, 23(9), 4631; https://doi.org/10.3390/ijms23094631
Submission received: 13 April 2022 / Revised: 17 April 2022 / Accepted: 19 April 2022 / Published: 22 April 2022
(This article belongs to the Special Issue Recent Advances in Thyroid Cancer Research)
This Special Issue (SI) “Recent Advances in Thyroid Cancer Research” covers research articles and reviews in the field of thyroid cancer research. It includes publications reporting the results of cell biological studies, animal investigations and clinical studies.
The thyroid or glandula thyroidea is an endocrine gland located in the neck below the larynx in front of the trachea in mammals. In humans, it has the shape of a butterfly. It consists of two lobes connected by a narrow bridge called the isthmus. Thyroid carcinoma or thyroid cancer (TC) is a malignant tumor of the thyroid gland. It is the most common malignancy of the endocrine (hormonal) system, but is very rare, accounting for about 1% of all malignant tumors, however, it has been increasing in frequency in recent years.
The latest Global Cancer Observatory survey from 2020 reports that TC is responsible for 586,000 cancer cases worldwide [1]. The American Cancer Society published this year that there will be about 43,800 new cases of TC (11,860 in men and 31,940 in women) and about 2230 deaths from this type of tumour (1070 men and 1160 women) in the United States in 2022 [2].
TC is a malignant tumor of the glandular tissue of the thyroid gland. There are different types of thyroid cancer that arise from different cells in the thyroid gland. TC comprises several categories: (1) differentiated TC (DTC), including papillary (PTC), follicular (FTC), and Huerthle cell tumours, (2) medullary TC (MTC) and (3) anaplastic TC. The long-term survival rate of patients with differentiated TC is about 90%, whereas patients with poorly differentiated TC types show a long-term survival rate of below 10% due to their resistance to standard treatment options [3,4].
Standard therapy strategies for advanced and radioiodine-refractory TC are immunotherapy, chemotherapy and kinase inhibitors [5]. Unfortunately, the survival rate is still low, and therefore new research approaches with novel technologies are necessary.
This SI published five research articles [6,7,8,9,10]. Four studies investigated different types of TC cells (TCC) [6,7,9,10] and in three publications the cell cultures were combined with an animal study as tumor model [6,9,10]. Furthermore, one research article focused on PTC in a large South Eastern cohort of patients [8]. In addition, the SI covers two reviews [11,12].
Hong et al. investigated medullary thyroid cancer cells (MTC) [6]. The rationale was that mortalin inhibition suppressed human MTC cells in culture and in mouse xenografts and induced apoptosis and the downregulation of RET. Moreover, the agent MKT-077 inhibited mortalin and induced similar effects, but this compound is known to be toxic in animals. Different MKT-077 analogs (JG-98 and JG-194) inhibited propagation of TT and MZ-CRC-1 cells in 2D and 3D cultures. Both analogs also effectively suppressed the viability of TT and MZ-CRC-1 progenies resistant to vandetanib and cabozantinib. Moreover, JG-231 suppressed TT and MZ-CRC-1 xenografts in mice. These data suggest a potential of mortalin as a target for the design of a molecular therapy for MTC [6].
This SI also covered a study investigating human follicular TCC on the ISS in space [7]. A spaceflight induces various changes on the gene and protein expression level in human cells. Real microgravity (r-µg) altered many biological processes such as among others the cytoskeleton, extracellular matrix, focal adhesion, migration, proliferation, apoptosis, cell survival or growth behavior [13,14]. Using free-flow IEF to identify proteins under µg-conditions 235 proteins had been detected. 37 of this search appeared to be first identifications in human thyroid cells [15] and may serve as future targets. The CellBox-2 experiment focused on FTC-133 TCC flown to the ISS during the SpaceX CRS-13 cargo mission [7]. TCC grown on the ISS showed a similar secretory behavior as on Earth. Furthermore, the gene expression involved in processes such as proliferation, adhesion, growth and metastasis was suppressed in space [7]. In addition, a suppression of the NF-κB and ERK signaling pathway and an elevated angiopoietin 2 secretion after 10 d in orbit could be demonstrated [7]. These results are of clinical importance, but it has to be taken into account that the FTC-133 cell line is a stable platform for research on TC but does not fully represent the situation occurring in vivo.
Another study investigated FOXE1-induced transcriptional alterations in thyroid cells, not expressing endogenous FOXE1 [9]. The authors demonstrated the FOXE1-dependent regulation of macrophage chemotaxis by thyroid cells in vitro and in vivo. This study detected a link between FOXE1 and macrophages recruitment in the TCC microenvironment. The results indicate a function of this gene in the interaction of TCC and immune cells in relation to tumor development and progression [9].
Jiménez-Mora et al. demonstrated that the inhibition of V600EBRAF induces a cytoprotective autophagy through AMPK in TCC [10]. The paper focused on the mechanisms whereby V600EBRAF inhibition induced autophagy via activation of the LKB1-AMPK-ULK1 pathway. In addition, autophagy showed cell-protective properties and its blockage potentiates PLX4720-induced cell death of TCC carrying a V600EBRAF mutation, both in vitro and in vivo. This data proposed new treatment strategies to target the AMPK pathway and/or autophagy that can contribute to enhance the efficacy of V600EBRAF inhibitors and to overcome the acquired resistance to these compounds in TC [10].
A clinical study focused on the molecular mechanism and the clinical impact of ZNF677 expression in more than 1200 Middle Eastern papillary TC (PTC) and 15 metastatic tissues [8]. ZNF677 was frequently downregulated in primary PTC (13.6%, 168/1235) and the complete loss of ZNF677 expression was significantly associated with aggressive clinico-pathological markers such as extrathyroidal extension and distant metastases. Moreover, ZNF677 loss was an independent predictor of distant metastasis in PTC. In addition, the data showed that ZNF677 acts as a tumor suppressor, mediating its effect by inhibiting AKT phosphorylation. The authors demonstrated a key role for ZNF677 during carcinogenesis and metastasis formation in Middle Eastern PTC patients [8].
Finally, this SI also covered two reviews. The first review by Nguyen et al. summarized the current knowledge in clinicopathological and molecular features of secondary cancer (metastasis) to the thyroid and recent advances in the management of this finding [11]. This review provides clinicians with a comprehensive update regarding the clinical signs, diagnostic investigations, and current management of secondary cancer to the thyroid gland. Targeted therapies, such as multikinase inhibitors (MKIs) and immune checkpoint inhibitors might be effective in some of these patients.
The next review discussed the use of the MKIs lenvatinib, sorafenib and cabozantinib in RAI-refractory TC patients [12]. The authors reviewed the current knowledge of the MKIs in iodine-refractory DTC with focus on occurrence, mechanisms, and management of treatment-emergent hypertension (TE-HTN). The AE TE-HTN occurred during treatment with all compounds, but was well manageable as shown by the latest clinical trials. This was emphasized by the fact that lenvatinib was widely used as first-line drug of choice, despite its higher rate of TE-HTN [12].
Taken together, the seven excellent publications included in this SI demonstrate novel findings in the field of ‘Recent Advances in Thryoid Cancer Research’. The authors investigated several genes and molecular pathways which might serve as targets for future therapy in different TC types. They used cell culture and animal models or a large patient cohort to study molecular mechanisms to develop the rationale for a future targeted therapy.
I want to thank very much all the authors who contributed to this SI. I am convinced that the use of new molecular biological technologies together with a personalized medicine will increase the current knowledge of prevention, diagnosis and new therapies for TC. Moreover, novel approaches using OMICS technologies and Bioinformatics to search new proteins that may serve as new drug targets, will help to reduce the mortality of advanced metastatic TC types and secondary cancer to the thyroid.

Funding

D.G. was funded by Deutsches Zentrum für Luft-und Raumfahrt (DLR), BMWi project 50WB1924.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

I want to thank Herbert Schulz, Otto von Guericke University Magdeburg, Germany for his help with EndNote and important suggestions.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

AEAdverse effect
AKTProteinkinase B/AKT serine/threonine kinase 1
AMPK5′ adenosine monophosphate-activated protein kinase
BRAFv-Raf murine sarcoma viral oncogene homolog B1
DDay
ERKExtracellular-signal regulated kinases
FOXE1Forkhead box E1
FTCFollicular thyroid cancer
HTNHypertension
ISSInternational Space Station
LKB1Liver kinase B1
MCSMulticellular spheroids
MKIsMultikinase inhibitors
MTCMedullary thyroid cancer
mTORMammalian target of rapamycin
PTCPapillary thyroid cancer
r-µgReal microgravity
SISpecial Issue
TCThyroid cancer
TCCThyroid cancer cells
TE-HTNTreatment-emergent hypertension
2DTwo-dimensional
3DThree-dimensional
ULK1Unc-51 like autophagy activating kinase
ZNF677Zinc finger protein 677

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Key Statistics for Thyroid Cancer. How Common Is Thyroid Cancer? The American Cancer Society. Available online: https://www.cancer.org/cancer/thyroid-cancer/about/key-statistics.html (accessed on 11 April 2022).
  3. Ancker, O.V.; Kruger, M.; Wehland, M.; Infanger, M.; Grimm, D. Multikinase Inhibitor Treatment in Thyroid Cancer. Int. J. Mol. Sci. 2019, 21, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Patel, K.N.; Shaha, A.R. Poorly differentiated thyroid cancer. Curr. Opin. Otolaryngol. Head Neck. Surg. 2014, 22, 121–126. [Google Scholar] [CrossRef] [PubMed]
  5. Matrone, A.; Campopiano, M.C.; Nervo, A.; Sapuppo, G.; Tavarelli, M.; De Leo, S. Differentiated Thyroid Cancer, From Active Surveillance to Advanced Therapy: Toward a Personalized Medicine. Front. Endocrinol. 2019, 10, 884. [Google Scholar] [CrossRef] [PubMed]
  6. Hong, S.K.; Starenki, D.; Johnson, O.T.; Gestwicki, J.E.; Park, J.I. Analogs of the Heat Shock Protein 70 Inhibitor MKT-077 Suppress Medullary Thyroid Carcinoma Cells. Int. J. Mol. Sci. 2022, 23, 1063. [Google Scholar] [CrossRef] [PubMed]
  7. Melnik, D.; Kruger, M.; Schulz, H.; Kopp, S.; Wehland, M.; Bauer, J.; Baselet, B.; Vermeesen, R.; Baatout, S.; Corydon, T.J.; et al. The CellBox-2 Mission to the International Space Station: Thyroid Cancer Cells in Space. Int. J. Mol. Sci. 2021, 22, 8777. [Google Scholar] [CrossRef] [PubMed]
  8. Siraj, A.K.; Poyil, P.K.; Parvathareddy, S.K.; Alobaisi, K.; Ahmed, S.O.; Al-Sobhi, S.S.; Al-Dayel, F.; Al-Kuraya, K.S. Loss of ZNF677 Expression Is an Independent Predictor for Distant Metastasis in Middle Eastern Papillary Thyroid Carcinoma Patients. Int. J. Mol. Sci. 2021, 22, 7833. [Google Scholar] [CrossRef] [PubMed]
  9. Credendino, S.C.; De Menna, M.; Cantone, I.; Moccia, C.; Esposito, M.; Di Guida, L.; De Felice, M.; De Vita, G. FOXE1-Dependent Regulation of Macrophage Chemotaxis by Thyroid Cells In Vitro and In Vivo. Int. J. Mol. Sci. 2021, 22, 7666. [Google Scholar] [CrossRef] [PubMed]
  10. Jimenez-Mora, E.; Gallego, B.; Diaz-Gago, S.; Lasa, M.; Baquero, P.; Chiloeches, A. (V600E)BRAF Inhibition Induces Cytoprotective Autophagy through AMPK in Thyroid Cancer Cells. Int. J. Mol. Sci. 2021, 22, 6033. [Google Scholar] [CrossRef] [PubMed]
  11. Nguyen, M.; He, G.; Lam, A.K. Clinicopathological and Molecular Features of Secondary Cancer (Metastasis) to the Thyroid and Advances in Management. Int. J. Mol. Sci. 2022, 23, 3242. [Google Scholar] [CrossRef] [PubMed]
  12. Kaae, A.C.; Kreissl, M.C.; Kruger, M.; Infanger, M.; Grimm, D.; Wehland, M. Kinase-Inhibitors in Iodine-Refractory Differentiated Thyroid Cancer-Focus on Occurrence, Mechanisms, and Management of Treatment-Related Hypertension. Int. J. Mol. Sci. 2021, 22, 12217. [Google Scholar] [CrossRef] [PubMed]
  13. Grimm, D.; Schulz, H.; Kruger, M.; Cortes-Sanchez, J.L.; Egli, M.; Kraus, A.; Sahana, J.; Corydon, T.J.; Hemmersbach, R.; Wise, P.M.; et al. The Fight against Cancer by Microgravity: The Multicellular Spheroid as a Metastasis Model. Int. J. Mol. Sci. 2022, 23, 3073. [Google Scholar] [CrossRef] [PubMed]
  14. Ma, X.; Pietsch, J.; Wehland, M.; Schulz, H.; Saar, K.; Hubner, N.; Bauer, J.; Braun, M.; Schwarzwalder, A.; Segerer, J.; et al. Differential gene expression profile and altered cytokine secretion of thyroid cancer cells in space. FASEB J. 2014, 28, 813–835. [Google Scholar] [CrossRef]
  15. Pietsch, J.; Kussian, R.; Sickmann, A.; Bauer, J.; Weber, G.; Nissum, M.; Westphal, K.; Egli, M.; Grosse, J.; Schonberger, J.; et al. Application of free-flow IEF to identify protein candidates changing under microgravity conditions. Proteomics 2010, 10, 904–913. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Grimm, D. Recent Advances in Thyroid Cancer Research. Int. J. Mol. Sci. 2022, 23, 4631. https://doi.org/10.3390/ijms23094631

AMA Style

Grimm D. Recent Advances in Thyroid Cancer Research. International Journal of Molecular Sciences. 2022; 23(9):4631. https://doi.org/10.3390/ijms23094631

Chicago/Turabian Style

Grimm, Daniela. 2022. "Recent Advances in Thyroid Cancer Research" International Journal of Molecular Sciences 23, no. 9: 4631. https://doi.org/10.3390/ijms23094631

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop