Next Article in Journal
Treatment for Viral Hepatitis as Secondary Prevention for Hepatocellular Carcinoma
Next Article in Special Issue
Anti-Aging Effects of a Serum Based on Coconut Oil Combined with Deer Antler Stem Cell Extract on a Mouse Model of Skin Aging
Previous Article in Journal
Impaired Skeletal Muscle Development and Regeneration in Transglutaminase 2 Knockout Mice
Previous Article in Special Issue
Evaluation of Immunomodulatory Responses and Changed Wound Healing in Type 2 Diabetes—A Study Exploiting Dermal Fibroblasts from Diabetic and Non-Diabetic Human Donors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

GLUT1, GLUT3 Expression and 18FDG-PET/CT in Human Malignant Melanoma: What Relationship Exists? New Insights and Perspectives

1
Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy
2
Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy
3
Section of Ginecology and Obstetrics, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy
4
Section of Legal Medicine, Department of Interdisciplinary Medicine, Bari Policlinico Hospital, University of Bari, “Aldo Moro”, 70124 Bari, Italy
*
Author to whom correspondence should be addressed.
These Authors contributed to the same extent.
Cells 2021, 10(11), 3090; https://doi.org/10.3390/cells10113090
Submission received: 14 October 2021 / Revised: 3 November 2021 / Accepted: 7 November 2021 / Published: 9 November 2021
(This article belongs to the Special Issue Molecular and Cell Basis of Skin Diseases and Aging)

Abstract

:
Background: Malignant melanoma is the most aggressive of skin cancers and the 19th most common cancer worldwide, with an estimated age-standardized incidence rate of 2.8–3.1 per 100,000; although there have been clear advances in therapeutic treatment, the prognosis of MM patients with Breslow thickness greater than 1 mm is still quite poor today. The study of how melanoma cells manage to survive and proliferate by consuming glucose has been partially addressed in the literature, but some rather interesting results are starting to be present. Methods: A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and a search of PubMed and Web of Sciences (WoS) databases was performed until 27 September 2021 using the terms: glucose transporter 1 and 3 and GLUT1/3 in combination with each of the following: melanoma, neoplasm and immunohistochemistry. Results: In total, 46 records were initially identified in the literature search, of which six were duplicates. After screening for eligibility and inclusion criteria, 16 publications were ultimately included. Conclusions: the results discussed regarding the role and expression of GLUT are still far from definitive, but further steps toward understanding and stopping this mechanism have, at least in part, been taken. New studies and new discoveries should lead to further clarification of some aspects since the various mechanisms of glucose uptake by neoplastic cells are not limited to the transporters of the GLUT family alone.

Graphical Abstract

1. Introduction

Malignant melanoma is the most aggressive of skin cancers and the 19th most common cancer worldwide, with an estimated age-standardized incidence rate of 2.8–3.1 per 100,000 [1]. Although the advent of new targeted molecular therapies (directed against BRAF mutations) and immunotherapy have made it possible to improve the survival and clinical outcome of affected patients, even today the diagnosis of melanoma still has a strong impact on the patient’s life, and survival rates are strictly dependent on independent prognostic markers, above all, Breslow thickness [2,3]. In the diagnostics, staging, restaging and follow-up of patients suffering from various neoplasms, including melanoma, positron emission tomography/CT with the use of 18 fluoro-deoxy-glucose is considered the “gold standard” investigation [4]. The underlying physical principle relates to the notion that proliferating cells modify their glucose metabolism so as to allow them to “feed” through the process of aerobic glycolysis (Warburg effect), surviving and maintaining the neoplastic clone in expansion [5]. Against this background, some works in the literature have shed some light on the potential relationships between glucose uptake, visualization in FDG-PET/CT and predominantly immunohistochemical expression of glucose transporters 1 and 3: that they have a high affinity for glucose and can operate in an insulin-independent manner. In this paper, we conduct a detailed review of the existing works in the literature on this topic, outlining the state of the art in this field and, finally, consider future viable paths.

2. Materials and Methods

A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A search of PubMed and Web of Sciences (WoS) databases was performed until 27 September 2021 using the terms: glucose transporter 1 and 3 and GLUT1/3 in combination with each of the following: melanoma, neoplasm and immunohistochemistry. Only articles in English were selected. Eligible articles were assessed according to the Oxford Centre for Evidence-Based Medicine 2011 guidelines [6]. Review articles, meta-analyses, observational studies and letters to the editor were included. Other potentially relevant articles were identified by manually checking the references of the included literature.
An independent extraction of articles was performed by two investigators according to the inclusion criteria. Disagreement was resolved by discussion between the two review authors. Since the study designs, participants, treatment measures and reported outcomes varied markedly, we focused on describing the different approaches of the authors regarding the expression of GLUT1 and GLUT3 in malignant melanoma, analyzed the techniques (mainly immunohistochemistry) used in the works examined and, finally, analyzed the state of the art, imagining what the future perspectives may be.

3. Results

In total, 46 records were initially identified in the literature search, of which six were duplicates. After screening for eligibility and inclusion criteria, 16 publications were ultimately included (Figure 1). The study and features are summarized in Table 1. Most of the publications were original and/or research articles (n = 12), followed by reviews with or without metanalysis (n = 2), a comparative study (n = 1) and a case–control retrospective study (n = 1). All studies included were rated as level 4 or 5 evidence for clinical research as detailed in the Oxford Centre for Evidence-Based Medicine 2011 guidelines [6].

4. Discussion

Melanoma continues to be the most aggressive cutaneous malignancy of all [1,23], featuring different incidence and prevalence rates on the various continents, but which tend to remain rather stable [23]. Classically, MM has always been distinguished as cutaneous or mucosal according to the site of onset, but, in recent years, research has made it possible to study in greater depth the mechanisms of the genetic–molecular landscape of both types, offering a better understanding of the points of contact and divergence [7,8]. It has long been known that glucose transporters (GLUT1-14) belong to the family of structurally related proteins that mediate the energy-independent transport of glucose across the plasma membrane. Among the various types, the most historically studied have been GLUT1, expressed mostly in erythrocytes, endothelial cells of the blood–brain barrier and placental cells and GLUT3, also present in various tissues [9,10]. Different works have shown that the basic principle on which the use of FDG-PET/CT is based is the increased energy requirement of cancer cells, with a consequent increase in the expression of GLUT1 and the neoplasm capacity to proliferate, survive and, finally, metastasize. This mechanism has been well studied and characterized in neoplasms of the breast, pancreas, cervix, endometrium, lung, mesothelium, colon, bladder, thyroid, bone, soft tissues and oral cavity [10,11,12]. Conversely, there are conflicting results regarding the role played by GLUT3 in various malignancies [13]. In the field of melanocytic pathology, we probed the role of GLUT1/3 in mediating glucose uptake and correlated findings with the clinical outcome.
In 2002, Wachsberger et al. demonstrated, by Western immunoblotting performed on 31 melanoma biopsies, a wide variability in the expression of both GLUT1 and HK and proposed the explanation that a broad spectrum of transporter activities could differently influence the response of patients to the same therapeutic treatment [14]. Three years later, Yamada et al. [15] conducted a study using four human melanoma cell lines (SK-MEL 23, SK-MEL 24 and G361) and one mouse (B16): the authors demonstrated that factors such as cell viability and rate of cell proliferation, as well as HK expression, were directly responsible for an increased glucose uptake at FDG-PET/CT. However, there did not appear to be any correlation between GLUT1 expression and the degree of glucose uptake. In another study, in 2008, Parente et al. reported results obtained from the analysis of 44 skin lesions, consisting of 12 benign nevi, 12 Spitz nevi and 20 primitive melanomas. The authors conducted an immunohistochemical study for markers such as GLUT1 and GLUT3 and reported that, unlike GLUT3 (expressed in all the lesions analyzed), GLUT1 was downregulated in 55% of primary melanomas, suggesting that glucose transport occurred across the plasma membrane by other transporters [16]. In 2009, Strobel et al. reported their experience with liver metastases resection samples from uveal and skin melanoma. Studying the reliability of FDG-PET/CT and the serum S-100B assay, the authors found that, while these determinations were reliable (sensitive) for skin-priming melanoma metastases, they were not as reliable in cases of metastasis from uveal melanoma [17]. In 2012, Park et al. conducted a detailed study to investigate the correlative association between glucose uptake in FDG-PET/CT, GLUT1/3 expression, exokinase-2 (HK-2) expression and Ki-67 expression in malignant melanoma. Enrolling 19 patients with a proven histological diagnosis of MM and pre-subjected to FDG-PET/CT, the authors found that the metabolic uptake increased with increasing GLUT1/3 expression, while HK-2 and Ki67 did not appear to be correlated [18,19,20]. Another important aspect was addressed by two other groups [21,24] regarding the role that hypoxia plays in the translocation and, therefore, expression of GLUT in cancer cells: from these works it emerged that the hypoxic condition is able to activate a pathway signal transduction mediated by HIF-1-alpha. In addition to other actions, this is able to promote the expression of glucose transporters, allowing the neoplastic cell to continue to survive through airborne glycolysis (Warburg effect) from oxidative phosphorylation. This has also been partially demonstrated in non-melanoma skin cancer (NMSC) [24].
In 2019, Dura et al. conducted a study of 400 cases (225 melanomas and 175 benign nevi) to evaluate the expression of GLUT1 in immunohistochemistry. They found that 69/225 melanomas were positive for this marker and, moreover, demonstrated an increasing expression (evaluated according to a semiquantitative score, Immunoscore 15) with increasing Breslow thickness. Intriguingly, the authors demonstrated that GLUT1 expression was correlated with shorter 10-year disease-free survival, relapse-free survival and shorter metastasis-free survival (MFS) [25]. Various other authors [22,26,27,28,29] have examined these aspects, sometimes with confirmatory but sometimes contradictory results. A very interesting recent paper by Reckzeh et al. described the discovery of a powerful inhibitor of glucose transporters 1 and 3, Glutor, that is capable of blocking the uptake of neoplastic cells, blocking the “metabolic plasticity” of cancer cells. Used together with a CB-839 glutaminase inhibitor, the combination can block cell proliferation and survival [30].
Importantly, a certain variability in the results obtained from immunohistochemistry for GLUT1-3 emerges from this revision of the literature. It is necessary to bear in mind that in immunoassays the binding between antibody and protein (GLUT) could be hindered by the phenomenon of the glycosylation of these receptors, following which they become more similar to glucose [31]. Finally, future research directions should take into account the different subcellular localization of GLUTs depending on the various isoforms and the various functional moments [32].

5. Conclusions

A better knowledge and closer study of the tumor phenotype have been for many years one of the most important targets in cancer research. The study of how the cancer cell is able to evade control mechanisms and apoptosis and proliferate has aroused great interest and attention. Against this background, various efforts have been made to better understand the metabolism of neoplastic cells, paying particular attention to the concept of “metabolic plasticity” and the Warburg effect. The results discussed regarding the role and expression of GLUT are still far from definitive, but further steps toward understanding and stopping this mechanism have, at least in part, been taken. New studies and new discoveries should lead to the further clarification of some aspects since the various mechanisms of glucose uptake by neoplastic cells are not limited to the transporters of the GLUT family alone.

Author Contributions

Conceptualization, G.C. (Gerardo Cazzato), A.C. (Anna Colagrande) and L.L.; methodology, G.C. (Gerardo Cazzato), A.C. (Antonietta Cimmino), E.B. and C.A.; software, G.C. (Gerardo Cazzato) and T.L.; validation, G.C. (Gerardo Cazzato), P.R., C.A., C.F. and G.I.; formal analysis, G.C. (Gerardo Cazzato) and F.A.; investigation, G.C. (Gerardo Cazzato); resources, G.C. (Gerardo Cazzato), V.L. and G.C. (Gennaro Cormio); data curation, G.C. (Gerardo Cazzato) and L.L.; writing—original draft preparation, G.C. (Gerardo Cazzato), A.C. (Anna Colagrande and Antonietta Cimmino), G.I., L.R. and R.R.; writing—review and editing, C.F. and L.L.; visualization, S.S.; supervision, G.C. (Gerardo Cazzato). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ali, Z.; Yousaf, N.; Larkin, J. Melanoma epidemiology, biology and prognosis. EJC Suppl. 2013, 11, 81–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Davis, L.E.; Shalin, S.C.; Tackett, A.J. Current state of melanoma diagnosis and treatment. Cancer Biol. Ther. 2019, 20, 1366–1379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Abbas, O.; Miller, D.D.; Bhawan, J. Cutaneous malignant melanoma: Update on diagnostic and prognostic biomarkers. Am. J. Dermatopathol. 2014, 36, 363–379. [Google Scholar] [CrossRef] [PubMed]
  4. Macapinlac, H.A. FDG PET and PET/CT imaging in lymphoma and melanoma. Cancer J. 2004, 10, 262–270. [Google Scholar] [CrossRef]
  5. Ždralević, M.; Brand, A.; Di Ianni, L.; Dettmer, K.; Reinders, J.; Singer, K.; Peter, K.; Schnell, A.; Bruss, C.; Decking, S.M.; et al. Double genetic disruption of lactate dehydrogenases A and B is required to ablate the “Warburg effect” restricting tumor growth to oxidative metabolism. J. Biol. Chem. 2018, 293, 15947–15961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Oxford Centre for Evidence-Based Medicine 2011 Levels of Evidence. Available online: http://www.cebm.net/wp-content/uploads/2014/06/CEBM-Levels-of-Evidence-2.1.pdf (accessed on 27 September 2021).
  7. Cazzato, G.; Arezzo, F.; Colagrande, A.; Cimmino, A.; Lettini, T.; Sablone, S.; Resta, L.; Ingravallo, G. “Animal-Type Melanoma/Pigmented Epithelioid Melanocytoma”: History and Features of a Controversial Entity. Dermatopathology 2021, 8, 271–276. [Google Scholar] [CrossRef]
  8. Cazzato, G.; Colagrande, A.; Cimmino, A.; Caporusso, C.; Candance, P.M.V.; Trabucco, S.M.R.; Zingarelli, M.; Lorusso, A.; Marrone, M.; Stellacci, A.; et al. Urological Melanoma: A Comprehensive Review of a Rare Subclass of Mucosal Melanoma with Emphasis on Differential Diagnosis and Therapeutic Approaches. Cancers 2021, 13, 4424. [Google Scholar] [CrossRef]
  9. Ito, S.; Fukusato, T.; Nemoto, T.; Sekihara, H.; Seyama, Y.; Kubota, S. Coexpression of glucose transporter 1 and matrix metal-loproteinase-2 in human cancers. J. Natl. Cancer Inst. 2002, 94, 1080–1091. [Google Scholar] [CrossRef]
  10. Chang, S.; Lee, S.; Lee, C.; Kim, J.I.; Kim, Y. Expression of the human erythrocyte glucose transporter in transitional cell carcinoma of the bladder. Urology 2000, 55, 448–452. [Google Scholar] [CrossRef]
  11. Grover-McKay, M.; Walsh, S.A.; Seftor, E.A.; Thomas, P.A.; Hendrix, M.J. Role for glucose transporter 1 protein in human breast cancer. Pathol. Oncol. Res. 1998, 4, 115–120. [Google Scholar] [CrossRef]
  12. Kunkel, M.; Moergel, M.; Stockinger, M.; Jeong, J.H.; Fritz, G.; Lehr, H.A.; Whiteside, T.L. Overexpression of GLUT-1 is associated with resistance to radiotherapy and adverse prognosis in squamous cell carcinoma of the oral cavity. Oral Oncol. 2007, 43, 796–803. [Google Scholar] [CrossRef] [PubMed]
  13. Ito, T.; Noguchi, Y.; Satoh, S.; Hayashi, H.; Inayama, Y.; Kitamura, H. Expression of facilitative glucose transporter isoforms in lung carcinomas: Its relation to histologic type, differentiation grade, and tumor stage. Mod. Pathol. 1998, 11, 437–443. [Google Scholar]
  14. Wachsberger, P.R.; Gressen, E.L.; Bhala, A.; Bobyock, S.B.; Storck, C.; Coss, R.A.; Berd, D.; Leeper, D.B. Variability in glucose transporter-1 levels and hexokinase activity in human melanoma. Melanoma Res. 2002, 12, 35–43. [Google Scholar] [CrossRef] [PubMed]
  15. Yamada, K.; Brink, I.; Bissé, E.; Epting, T.; Engelhardt, R. Factors influencing [F-18] 2-fluoro-2-deoxy-D-glucose (F-18 FDG) uptake in melanoma cells: The role of proliferation rate, viability, glucose transporter expression and hexokinase activity. J. Dermatol. 2005, 32, 316–334. [Google Scholar] [CrossRef]
  16. Parente, P.; Coli, A.; Massi, G.; Mangoni, A.; Fabrizi, M.M.; Bigotti, G. Immunohistochemical expression of the glucose transporters Glut-1 and Glut-3 in human malignant melanomas and benign melanocytic lesions. J. Exp. Clin. Cancer Res. 2008, 27, 34. [Google Scholar] [CrossRef] [Green Version]
  17. Strobel, K.; Bode, B.; Dummer, R.; Veit-Haibach, P.; Fischer, D.R.; Imhof, L.; Goldinger, S.; Steinert, H.C.; von Schulthess, G.K. Limited value of 18F-FDG PET/CT and S-100B tumour marker in the detection of liver metastases from uveal melanoma compared to liver metastases from cutaneous melanoma. Eur. J. Nucl. Med. Mol. Imaging 2009, 36, 1774–1782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Park, S.G.; Lee, J.H.; Lee, W.A.; Han, K.M. Biologic correlation between glucose transporters, hexokinase-II, Ki-67 and FDG uptake in malignant melanoma. Nucl. Med. Biol. 2012, 39, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
  19. Airley, R.; Evans, A.; Mobasheri, A.; Hewitt, S.M. Glucose transporter Glut-1 is detectable in peri-necrotic regions in many human tumor types but not normal tissues: Study using tissue microarrays. Ann. Anat. 2010, 192, 133–138. [Google Scholar] [CrossRef]
  20. Singer, K.; Kastenberger, M.; Gottfried, E.; Hammerschmied, C.G.; Büttner, M.; Aigner, M.; Seliger, B.; Walter, B.; Schlösser, H.; Hartmann, A.; et al. Warburg phenotype in renal cell carcinoma: High expression of glucose-transporter 1 (GLUT-1) correlates with low CD8+ T-cell infiltration in the tumor. Int. J. Cancer 2011, 128, 2085–2095. [Google Scholar] [CrossRef]
  21. Slominski, A.; Kim, T.K.; Brożyna, A.A.; Janjetovic, Z.; Brooks, D.L.; Schwab, L.P.; Skobowiat, C.; Jóźwicki, W.; Seagroves, T.N. The role of melanogenesis in regulation of melanoma behavior: Melanogenesis leads to stimulation of HIF-1α expression and HIF-dependent attendant pathways. Arch. Biochem. Biophys. 2014, 563, 79–93. [Google Scholar] [CrossRef] [Green Version]
  22. Su, J.; Gao, T.; Jiang, M.; Wu, L.; Zeng, W.; Zhao, S.; Peng, C.; Chen, X. CD147 silencing inhibits tumor growth by suppressing glucose transport in melanoma. Oncotarget 2016, 7, 64778–64784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Matthews, N.H.; Li, W.Q.; Qureshi, A.A.; Weinstock, M.A.; Cho, E. Epidemiology of Melanoma. In Cutaneous Melanoma: Etiology and Therapy [Internet]; Ward, W.H., Farma, J.M., Eds.; Codon Publications: Brisbane, Australia, 2017; Chapter 1. Available online: https://www.ncbi.nlm.nih.gov/books/NBK481862/ (accessed on 11 October 2021).
  24. Seleit, I.; Bakry, O.A.; Al-Sharaky, D.R.; Ragab, R.A.A.; Al-Shiemy, S.A. Evaluation of Hypoxia Inducible Factor-1α and Glucose Transporter-1 Expression in Non Melanoma Skin Cancer: An Immunohistochemical Study. J. Clin. Diagn. Res. 2017, 11, EC09–EC16. [Google Scholar] [CrossRef] [PubMed]
  25. Důra, M.; Němejcová, K.; Jakša, R.; Bártů, M.; Kodet, O.; Tichá, I.; Michálková, R.; Dundr, P. Expression of Glut-1 in Malignant Melanoma and Melanocytic Nevi: An Immunohistochemical Study of 400 Cases. Pathol. Oncol. Res. 2019, 25, 361–368. [Google Scholar] [CrossRef]
  26. Ruby, K.N.; Liu, C.L.; Li, Z.; Felty, C.C.; Wells, W.A.; Yan, S. Diagnostic and prognostic value of glucose transporters in mela-nocytic lesions. Melanoma Res. 2019, 29, 603–611. [Google Scholar] [CrossRef] [PubMed]
  27. Na, K.J.; Choi, H.; Oh, H.R.; Kim, Y.H.; Lee, S.B.; Jung, Y.J.; Koh, J.; Park, S.; Lee, H.J.; Jeon, Y.K.; et al. Reciprocal change in Glucose metabolism of Cancer and Immune Cells mediated by different Glucose Transporters predicts Immunotherapy response. Theranostics 2020, 10, 9579–9590. [Google Scholar] [CrossRef] [PubMed]
  28. Meyer, H.J.; Wienke, A.; Surov, A. Associations between GLUT expression and SUV values derived from FDG-PET in different tumors-A systematic review and meta analysis. PLoS ONE 2019, 14, e0217781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Chen, X.; Lu, P.; Zhou, S.; Zhang, L.; Zhao, J.H.; Tang, J.H. Predictive value of glucose transporter-1 and glucose transporter-3 for survival of cancer patients: A meta-analysis. Oncotarget 2017, 8, 13206–13213. [Google Scholar] [CrossRef]
  30. Reckzeh, E.S.; Karageorgis, G.; Schwalfenberg, M.; Ceballos, J.; Nowacki, J.; Stroet, M.C.M.; Binici, A.; Knauer, L.; Brand, S.; Choidas, A.; et al. Inhibition of Glucose Transporters and Glutaminase Synergistically Impairs Tumor Cell Growth. Cell Chem. Biol. 2019, 26, 1214–1228.e25. [Google Scholar] [CrossRef]
  31. Asano, T.; Katagiri, H.; Takata, K.; Lin, J.L.; Ishihara, H.; Inukai, K.; Tsukuda, K.; Kikuchi, M.; Hirano, H.; Yazaki, Y.; et al. The role of N-glycosylation of GLUT1 for glucose transport activity. J. Biol. Chem. 1991, 266, 24632–24636. [Google Scholar] [CrossRef]
  32. Maaßen, T.; Vardanyan, S.; Brosig, A.; Merz, H.; Ranjbar, M.; Kakkassery, V.; Grisanti, S.; Tura, A. Monosomy-3 Alters the Expression Profile of the Glucose Transporters GLUT1-3 in Uveal Melanoma. Int. J. Mol. Sci. 2020, 21, 9345. [Google Scholar] [CrossRef]
Figure 1. Research of the literature and selection of articles according to PRISMA guidelines.
Figure 1. Research of the literature and selection of articles according to PRISMA guidelines.
Cells 10 03090 g001
Table 1. Summary of the papers examined in this review.
Table 1. Summary of the papers examined in this review.
Author(s)Type of PaperNeoplasmProcedureResults
Park et al. [7]Research article19 cases of malignant melanomaFDG-PET/CT and immunohistochemistry for GLUT1/3GLUT1/3 mediate glucose uptake in FDG-PET/CT
HK-2 and Ki-67 play no role
Dura et al. [8]Original article225 cases of MM/175
benign nevus
Immunohistochemistry
for GLUT1
GLUT1 was expressed in
69/225 MM
GLUT1 was negative in
benign nevus
Strobel et al. [9]Research article27 liver metastases of 13 patients with uveal melanoma
43 liver metastases of 14 patients with CMM
FDG-PET/CT
S100B serum marker
Cytology
Histology
Immunohistochemistry for GLUT1 and S-100
46% of liver metastases by UM were negative in FDG- PET/CT
All liver metastases by CMM were positive
S-100B values were significantly higher in the CM patient group compared with the UM patients
No obvious difference for GLUT1 and S-100 protein
Ruby et al. [10]Research article91 MM
18 metastases of MM
56 benign nevus
Immunohistochemistry for GLUT2/3GLUT2 was negative in all melanomas and benign nevi 85.3% expressed either GLUT1 or GLUT3 or both, 39.4% of melanoma cases coexpressed GLUT1 and GLUT3, 17.4% of melanoma cases only expressed GLUT1, 28.4% of melanoma cases only expressed GLUT3 and 14.7% of melanoma cases were negative for both markers
Seleit et al. [11]Case–control and retrospective study20BCC
20 SCC
Immunohistochemistry for HIF-1-alpha and GLUT1HIF-1-alpha has a role in NMSC pathogenesis, GLUT1 is downregulated
Wachsberger et al. [12]Original article31 MMWestern immunoblot analysis for GLUT1 and HK-230 MM exhibited a 22-fold variation in levels of GLUT1 and 29 exhibited a nine-fold variation in total cellular
Hexokinase-II activity
Slominski et al. [13]Original article2 cellular lines of MM:
amelanotic/melanotic
Cultured cellsMelanogenesis upregulates HIF-1-alpha expression in MM
Na et al. [14]Original article63 pulmonary squamous cell carcinomaFDG-PET/CT
RNA-seq Immunohistochemistry
GLUT1/3 may be responsible for a different response to immunotherapy
Yamada et al. [15]Comparative study4 cellular lines of human MM: SK-MEL 23, SK-MEL 24 and G361
and 1 murine: B16
FDG-PET/CT
Immunohistochemistry for HK-2 and GLUT1
Proliferation rate, cell viability and HK expression were important to mediate
FDG uptake
Parente et al. [16]Original articleMalignant melanoma/benign nevusImmunohistochemistry for GLUT1-3GLUT1: positive in >90% benign nevus
GLUT1: absent in 55% MM GLUT3: present in both (MM and benign nevus)
Singer et al. [17]Research article249 RCCTissue microarray analysisIn RCC an increased expression of GLUT1 correlates with a decreased presence of
CD8 + T lymphocytes
Airley et al. [18]Research articleDifferent and various tumorsTissue microarray analysis and immunohistochemistry for GLUT1Glut-1 expression in peri- necrotic regions
Meyer et al. [19]Meta-analysisDifferent and various tumorsFDG-PET/TCOverall, only a moderate association was found between GLUT 1/3 expression and SUV values
derived from FDG-PET
Chen et al. [20]Review12 studies about GLUT1
2 studies about GLUT3
Review with meta-analysisA combination of GLUTs 1 and 3 might help predict malignancy of cancers and direct effective cancer
therapy
Reckzeh et al. [21]Original articleVarious cellular linesVarious proceduresGlutor, very potent glucose
uptake inhibitor
Su et al. [22]Research articleCellular line of MM:
A375
Immunoprecipitation
and siRNA
CD147 could downregulate
GLUT1 expression by reducing the proliferative
rate of MM cells
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cazzato, G.; Colagrande, A.; Cimmino, A.; Abbatepaolo, C.; Bellitti, E.; Romita, P.; Lospalluti, L.; Foti, C.; Arezzo, F.; Loizzi, V.; et al. GLUT1, GLUT3 Expression and 18FDG-PET/CT in Human Malignant Melanoma: What Relationship Exists? New Insights and Perspectives. Cells 2021, 10, 3090. https://doi.org/10.3390/cells10113090

AMA Style

Cazzato G, Colagrande A, Cimmino A, Abbatepaolo C, Bellitti E, Romita P, Lospalluti L, Foti C, Arezzo F, Loizzi V, et al. GLUT1, GLUT3 Expression and 18FDG-PET/CT in Human Malignant Melanoma: What Relationship Exists? New Insights and Perspectives. Cells. 2021; 10(11):3090. https://doi.org/10.3390/cells10113090

Chicago/Turabian Style

Cazzato, Gerardo, Anna Colagrande, Antonietta Cimmino, Caterina Abbatepaolo, Emilio Bellitti, Paolo Romita, Lucia Lospalluti, Caterina Foti, Francesca Arezzo, Vera Loizzi, and et al. 2021. "GLUT1, GLUT3 Expression and 18FDG-PET/CT in Human Malignant Melanoma: What Relationship Exists? New Insights and Perspectives" Cells 10, no. 11: 3090. https://doi.org/10.3390/cells10113090

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