Editorial Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Jul 26, 2018; 10(7): 78-81
Published online Jul 26, 2018. doi: 10.4252/wjsc.v10.i7.78
Ectopic expression of the osteogenic master gene RUNX2 in melanoma Maria Teresa Valenti, Luca Dalle Carbonare, Monica Mottes
Maria Teresa Valenti, Luca Dalle Carbonare, Department of Medicine, University of Verona, Verona 37100, Italy
Monica Mottes, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona 37100, Italy
ORCID number: Maria Teresa Valenti (0000-0003-1166-8033); Luca Dalle Carbonare (0000-0003-3263-6671); Monica Mottes (0000-0002-7390-2246).
Author contributions: Valenti MT and Mottes M conceived the study; Valenti MT, Dalle Carbonare L and Mottes M drafted the manuscript; all authors approved the final version of the paper.
Conflict-of-interest statement: The authors have no conflict of interest to declare.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Maria Teresa Valenti, BSc, PhD, Chief Technician, Research Fellow, Department of Medicine, University of Verona, Ple Scuro 10, Verona 37100, Italy. mariateresa.valenti@univr.it
Telephone: +39-45-8128450
Received: May 23, 2018
Peer-review started: May 25, 2018
First decision: June 5, 2018
Revised: June 11, 2018
Accepted: June 28, 2018
Article in press: June 28, 2018
Published online: July 26, 2018

Abstract

The transcription factor RUNX2 is the osteogenic master gene expressed in mesenchymal stem cells during osteogenic commitment as well as in pre-osteoblasts and early osteoblasts. However, RUNX2 is also ectopically expressed in melanoma and other cancers. Malignant melanoma (MM) is a highly metastatic skin cancer. The incidence of MM has increased considerably in the past half-century. The expression levels and mutation rates of genes such as BRAF, KIT, NRAS, PTEN, P53, TERT and MITF are higher in melanoma than in other solid malignancies. Additionally, transcription factors can affect cellular processes and induce cellular transformation since they control gene expression. Recently, several studies have identified alterations in RUNX2 expression. In particular, the regulation of KIT by RUNX2 and the increased expression of RUNX2 in melanoma specimens have been shown. Melanocytes, whose transformation results in melanoma, arise from the neural crest and therefore show “stemness” features. RUNX2 plays an important role in the re-activation of the MAPK and PI3K/AKT pathways, thus endowing melanoma cells with a high metastatic potential. In melanoma, the most frequent metastatic sites are the lung, liver, brain and lymph nodes. In addition, bone metastatic melanoma has been described. Notably, studies focusing on RUNX2 may contribute to the identification of an appropriate oncotarget in melanoma.

Key Words: RUNX2, Mesenchymal stem cells, Epitelial mesenchymal transition, Bone, Melanoma

Core tip: In addition to its physiological expression in osteogenic cells, RUNX2 ectopic expression is also reported in several cancers. Melanoma cells, which arise from the neural crest, are not typical epithelial cells and exhibit “stemness” features. For this reason the epithelial mesenchymal transition process may be enhanced in melanoma cells. RUNX2 expression in such context increases the migration and invasiveness of melanoma cells; it can therefore be considered a new oncotarget in melanoma.



INTRODUCTION

In recent times, the incidence of malignant melanoma (MM) has increased. The increased number of MM patients may be ascribed to lifestyle and environmental changes. Unfortunately, the mortality rate of MM is very high, as MM is highly metastatic and genetically resistant to apoptosis. The surgical treatment of skin lesions before cell spreading is considered a successful therapeutic approach. However, after metastasis and dissemination of cells has occurred, the therapeutic tools are limited. Despite an initial positive response to treatment, melanoma cells become resistant to chemotherapy[1]. Many studies performed in animal models and in cell culture have identified the complex mechanism involved in the metastatic progression of melanoma. Mutations in master transcription regulators such as BRAF, KIT, NRAS, PTEN, P53, TERT, and MITF have been found frequently[2]. In particular, the microphthalmia-associated transcription factor (MITF) is an important oncogene, and it plays a critical role in melanoma transformation due to its role in melanocyte proliferation and differentiation[3]. However, transcription factors in general may affect cellular processes and thereby induce cellular transformation. Among them, RUNX2, the master gene of osteogenic differentiation, has been shown to be involved in the transformation and metastatic progression of melanoma[4-6]. RUNX2, together with RUNX1 and RUNX3, belongs to the RUNT-related gene family[7]. These heterodimeric transcription factors have a DNA-binding “A” subunit and a non-DNA binding “B” subunit. The genomic and cDNA structures of the RUNT family members are evolutionarily conserved[8]. RUNX2 is located on human chromosome 6; its coding sequence is organized in eight exons and controlled by two promoters. Isoforms may originate by the alternative use of promoters or by the alternative splicing of exons; however, the DNA-binding domain remains invariant[7]. RUNX2 induces the commitment of mesenchymal stem cells to the osteogenic lineage by acting either as a monomer or as a heterodimeric complex, which shows a higher DNA binding activity[7]. Interestingly, differences in RUNX2 expression are associated with the skeletal differences between modern humans and Neanderthals[9]. Otto et al[10] using an in vivo model, showed that the ossification process is lacking in RUNX2 KO mice. RUNX2 is expressed not only in mesenchymal stem cells to commit these cells to osteogenic differentiation but also in pre-osteoblasts and early osteoblasts. It is then downregulated, as its continuous expression would prevent osteoblast maturation. This aspect is important for bone remodelling. In acromegaly patients, who are characterized by an excessive production of GH and IGF-1, a high fracture risk occurs; we have demonstrated that RUNX2 gene overexpression induces bone loss and impairs bone remodelling in these patients[11].

RUNX2 OVEREXPRESSION IN CANCER

The RUNX family members are frequently deregulated in human cancer. Runx proteins are involved in critical cellular processes such DNA repair, apoptosis, hypoxia, the inflammatory response, EMT, stem cell functions, and oncogene-induced senescence, and they interact with different signalling pathways such as the RAS-ERK, WNT, TGFβ, BMP and Notch pathways[12]. In addition, RUNX2 is involved in osteolytic diseases, neoangiogenic processes, and invasion and metastasis of solid tumours[4]. RUNX2 can be associated with HDA6 and can thereby prevent the pro-apoptotic activity of P53 through its deacetylation[13]. RUNX2 is ectopically expressed in several solid tumours such as breast cancer, pancreatic cancer, prostate cancer, lung cancer, and ovarian epithelial cancer; therefore, we have proposed RUNX2 as a mesenchymal stem marker for cancer[14,15]. Interestingly, we observed higher levels of RUNX2 expression in cancer patients with bone metastases compared to patients without bone metastases[15]. In thyroid cancer patients, we observed higher expression of RUNX2 in cancer tissue as well as in circulating mRNA compared to those levels of RUNX2 expression in controls[16]. In addition, RUNX2 expression was higher in cancer patients with microcalcifications compared to patients without microcalcifications[16]. Finally, it has been reported that RUNX2 induces its target genes SDF-1, CXCR7 and BSP, promoting bone homing[17].

RUNX2 IN MELANOMA

Ectopic expression of RUNX2 in melanoma tissue and in melanoma cell lines has been shown to be associated with bone sialoprotein (BSP)[18]. BSP has been associated with tumour invasiveness[18]. RUNX2 also regulates several matrix metalloproteinases that are involved in melanoma progression; therefore, RUNX2 may be regarded as a common regulator of both BSP and metalloproteinases[19]. This finding could also explain the expression of other bone proteins, such as osteopontin and osteocalcin, in melanoma[20,21]. TGFβ, which induces metastasis formation in advanced melanoma stages[22], can upregulate the expression of RUNX2 in melanoma cell lines[23]. In contrast, the tumour suppressor p14ARF reduces RUNX2 expression[24]. Loss-of-function p14ARF mutations lead to increased expression of RUNX2 and melanoma progression[24]. Ectopic expression of RUNX2 in melanoma has been associated with invasiveness; it has been demonstrated that RUNX2 overexpression mediates the migration ability of melanoma cells[6]. Notably RUNX2 stimulates VEGF gene expression[25] and the upregulation of angiogenesis, consequent to RUNX2 overexpression, may represent a critical step for tumor metastasis.

Furthermore, RUNX2 overexpression upregulates SOX9, SNAI2 and SMAD3. These transcription factors are involved in EMT as well as in cytoskeletal remodelling and thus enhance the motility and invasive potential of cancer cells[17]. In melanoma, the overexpression of EGF-R, PDGFRβ, AXL and IGF-1R induces the reactivation of the MAPK and PI3K/AKT pathways, which are involved in migration and invasion processes[26]. The reactivation of these pathways is due to tyrosine kinase receptor-based autocrine loops, in which RUNX2 plays a pivotal role[5]. The MAPK and AKT pathways regulate most physiological processes, such as proliferation, differentiation and cell survival. It has been suggested that the MAPK and AKT pathways are strongly associated with each other in melanoma; the link between the PI3K/AKT (AKT pathway) and MAPK/ERK1/2 (MAPK pathway) cascades is well known[27]. AKT enhances RUNX2 expression either directly or by inactivating RUNX2 regulators. Similarly, RUNX2 activates the PI3K/AKT pathway by regulating different components[4]. This reciprocal activation induces tumour progression and aggressiveness. In addition, RUNX2 promotes the crosstalk between MAPK and AKT through EGFR, as has been shown in human mammary epithelial cells[28] through the reduction of ERK-mediated inhibition of EGFR and the AKT pathway. The lung, liver, brain and lymph nodes are the most frequent metastatic sites for melanoma cells[29]. Since melanocytes arise from the neural crest, they are not typical epithelial cells; they show “stemness” features. This last finding could partially explain the high metastatic potential of melanoma[29]. Solid malignancies such as breast, prostate and lung cancers form metastases in bone. However, metastatic bone diseases from MM are under-investigated. The data in the literature report an incidence of skeletal metastases in MM patients that ranges from 0.85% to 18.6%[30]. Recently, Zekri et al[31], managing MM patients at two referral cancer centres in England, showed that bone metastases occur in 4.1% of patients at all stages of MM. In addition, another study showed that 17.2% of metastatic melanoma patients had metastases in bone tissue.

CONCLUSION

MM accounts for less than 5% of cutaneous malignancies. However, its incidence has been increasing considerably in the past half-century. Therefore, extensive research focusing on the genes involved in melanoma transformation and on the modulation of these genes is needed. Several studies have highlighted the involvement of RUNX2, the master gene of osteogenic differentiation, in melanoma. The invasiveness and metastatic features conferred by RUNX2 seem to be related to its ability to promote EMT. In addition, melanocytes are not typical epithelial cells, and their stemness features could explain melanoma invasiveness. Since metastatic melanoma has a poor prognosis, new and more effective therapeutic tools should be developed in order to implement current therapies. In this context, the exploitation of molecules actually involved in melanoma, such as RUNX2, represents an important frontier for the identification of new oncotargets. Transcription factors have been ignored thus far by the pharmaceutical industry; nonetheless, many studies have identified their central role in cellular transformation. We believe that future efforts directed towards unravelling the complex roles of RUNX2 may contribute to the identification of new therapeutic tools that improve the quality of life of melanoma patients.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Cell and tissue engineering

Country of origin: Italy

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Kiselev SL, Politi LE S- Editor: Ji FF L- Editor: A E- Editor: Tan WW

References
1.  Whiteman DC, Pavan WJ, Bastian BC. The melanomas: a synthesis of epidemiological, clinical, histopathological, genetic, and biological aspects, supporting distinct subtypes, causal pathways, and cells of origin. Pigment Cell Melanoma Res. 2011;24:879-897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 176]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
2.  Damsky WE, Bosenberg M. Melanocytic nevi and melanoma: unraveling a complex relationship. Oncogene. 2017;36:5771-5792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 113]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
3.  Levy C, Khaled M, Fisher DE. MITF: master regulator of melanocyte development and melanoma oncogene. Trends Mol Med. 2006;12:406-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 804]  [Cited by in F6Publishing: 782]  [Article Influence: 43.4]  [Reference Citation Analysis (0)]
4.  Cohen-Solal KA, Boregowda RK, Lasfar A. RUNX2 and the PI3K/AKT axis reciprocal activation as a driving force for tumor progression. Mol Cancer. 2015;14:137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 107]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
5.  Boregowda RK, Medina DJ, Markert E, Bryan MA, Chen W, Chen S, Rabkin A, Vido MJ, Gunderson SI, Chekmareva M. The transcription factor RUNX2 regulates receptor tyrosine kinase expression in melanoma. Oncotarget. 2016;7:29689-29707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
6.  Boregowda RK, Olabisi OO, Abushahba W, Jeong BS, Haenssen KK, Chen W, Chekmareva M, Lasfar A, Foran DJ, Goydos JS. RUNX2 is overexpressed in melanoma cells and mediates their migration and invasion. Cancer Lett. 2014;348:61-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
7.  Dalle Carbonare L, Innamorati G, Valenti MT. Transcription factor Runx2 and its application to bone tissue engineering. Stem Cell Rev. 2012;8:891-897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 96]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
8.  Otto F, Lübbert M, Stock M. Upstream and downstream targets of RUNX proteins. J Cell Biochem. 2003;89:9-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 176]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
9.  Benítez-Burraco A, Boeckx C. Possible functional links among brain- and skull-related genes selected in modern humans. Front Psychol. 2015;6:794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
10.  Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR, Stamp GW, Beddington RS, Mundlos S, Olsen BR. Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell. 1997;89:765-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2205]  [Cited by in F6Publishing: 2140]  [Article Influence: 79.3]  [Reference Citation Analysis (0)]
11.  Valenti MT, Mottes M, Cheri S, Deiana M, Micheletti V, Cosaro E, Davì MV, Francia G, Dalle Carbonare L. Runx2 overexpression compromises bone quality in acromegalic patients. Endocr Relat Cancer. 2018;25:269-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
12.  Ito Y. RUNX genes in development and cancer: regulation of viral gene expression and the discovery of RUNX family genes. Adv Cancer Res. 2008;99:33-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 167]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
13.  Ozaki T, Nakamura M, Shimozato O. Novel Implications of DNA Damage Response in Drug Resistance of Malignant Cancers Obtained from the Functional Interaction between p53 Family and RUNX2. Biomolecules. 2015;5:2854-2876.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
14.  Valenti MT, Serafini P, Innamorati G, Gili A, Cheri S, Bassi C, Dalle Carbonare L. Runx2 expression: A mesenchymal stem marker for cancer. Oncol Lett. 2016;12:4167-4172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
15.  Valenti MT, Mori A, Malerba G, Dalle Carbonare L. Mesenchymal stem cells: A new diagnostic tool? World J Stem Cells. 2015;7:789-792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
16.  Dalle Carbonare L, Frigo A, Francia G, Davì MV, Donatelli L, Stranieri C, Brazzarola P, Zatelli MC, Menestrina F, Valenti MT. Runx2 mRNA expression in the tissue, serum, and circulating non-hematopoietic cells of patients with thyroid cancer. J Clin Endocrinol Metab. 2012;97:E1249-E1256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
17.  Baniwal SK, Khalid O, Gabet Y, Shah RR, Purcell DJ, Mav D, Kohn-Gabet AE, Shi Y, Coetzee GA, Frenkel B. Runx2 transcriptome of prostate cancer cells: insights into invasiveness and bone metastasis. Mol Cancer. 2010;9:258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 125]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
18.  Riminucci M, Corsi A, Peris K, Fisher LW, Chimenti S, Bianco P. Coexpression of bone sialoprotein (BSP) and the pivotal transcriptional regulator of osteogenesis, Cbfa1/Runx2, in malignant melanoma. Calcif Tissue Int. 2003;73:281-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
19.  Hofmann UB, Westphal JR, Zendman AJ, Becker JC, Ruiter DJ, van Muijen GN. Expression and activation of matrix metalloproteinase-2 (MMP-2) and its co-localization with membrane-type 1 matrix metalloproteinase (MT1-MMP) correlate with melanoma progression. J Pathol. 2000;191:245-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
20.  Kiss T, Ecsedi S, Vizkeleti L, Koroknai V, Emri G, Kovács N, Adany R, Balazs M. The role of osteopontin expression in melanoma progression. Tumour Biol. 2015;36:7841-7847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
21.  Salvatierra E, Alvarez MJ, Leishman CC, Rivas Baquero E, Lutzky VP, Chuluyan HE, Podhajcer OL. SPARC Controls Melanoma Cell Plasticity through Rac1. PLoS One. 2015;10:e0134714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
22.  Lasfar A, Cohen-Solal KA. Resistance to transforming growth factor β-mediated tumor suppression in melanoma: are multiple mechanisms in place? Carcinogenesis. 2010;31:1710-1717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
23.  Mohammad KS, Javelaud D, Fournier PG, Niewolna M, McKenna CR, Peng XH, Duong V, Dunn LK, Mauviel A, Guise TA. TGF-beta-RI kinase inhibitor SD-208 reduces the development and progression of melanoma bone metastases. Cancer Res. 2011;71:175-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 172]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
24.  Packer LM, Pavey SJ, Boyle GM, Stark MS, Ayub AL, Rizos H, Hayward NK. Gene expression profiling in melanoma identifies novel downstream effectors of p14ARF. Int J Cancer. 2007;121:784-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
25.  Zelzer E, Glotzer DJ, Hartmann C, Thomas D, Fukai N, Soker S, Olsen BR. Tissue specific regulation of VEGF expression during bone development requires Cbfa1/Runx2. Mech Dev. 2001;106:97-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 249]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
26.  Yadav V, Denning MF. Fyn is induced by Ras/PI3K/Akt signaling and is required for enhanced invasion/migration. Mol Carcinog. 2011;50:346-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
27.  Pappalardo F, Russo G, Candido S, Pennisi M, Cavalieri S, Motta S, McCubrey JA, Nicoletti F, Libra M. Computational Modeling of PI3K/AKT and MAPK Signaling Pathways in Melanoma Cancer. PLoS One. 2016;11:e0152104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
28.  Tandon M, Chen Z, Pratap J. Role of Runx2 in crosstalk between Mek/Erk and PI3K/Akt signaling in MCF-10A cells. J Cell Biochem. 2014;115:2208-2217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
29.  Braeuer RR, Watson IR, Wu CJ, Mobley AK, Kamiya T, Shoshan E, Bar-Eli M. Why is melanoma so metastatic? Pigment Cell Melanoma Res. 2014;27:19-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 92]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
30.  Fon GT, Wong WS, Gold RH, Kaiser LR. Skeletal metastases of melanoma: radiographic, scintigraphic, and clinical review. AJR Am J Roentgenol. 1981;137:103-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
31.  Zekri J, Marples M, Taylor D, Kandukurti K, McParland L, Brown JE. Complications of bone metastases from malignant melanoma. J Bone Oncol. 2017;8:13-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]