The Role of IGF/IGF-IR-Signaling and Extracellular Matrix Effectors in Bone Sarcoma Pathogenesis
Abstract
:Simple Summary
Abstract
1. Introduction
2. IGF/IGFR-IR Signaling in Cancer
2.1. IGF Receptors
2.2. IGF-1 and IGF-2
2.3. IGFBs
2.4. IGF-IR Downstream Signaling
3. IGF-IR/IGF-I Signaling in Sarcoma Pathogenesis
3.1. Osteosarcoma
3.2. Chondrosarcoma
3.3. Ewing’s Sarcoma
4. The Sarcoma Tumor Microenvironment (TME)
The Non-Cellular TME Compartment in Sarcomas
5. Interplay between Matrix Effectors and IGF/IGF-IR Signaling Regulates Sarcoma Functions
5.1. IGF/IGF-IR Regulate Sarcoma Matrix Effectors Synthesis, Structure/Organization, and Downstream Functions
5.1.1. Proteoglycans
5.1.2. Collagens
5.1.3. Adhesion Molecules
5.1.4. Proteases
5.1.5. Summary
5.2. Matrix Effectors Modulate IGF/IGF-IR Pathway Restricted Signaling
6. IGF/IGF-IR Signaling Regulates Tumor Immune Response—Potential Therapeutic Application in Sarcomas
7. IGF Signaling in Tumor Angiogenesis
8. Data from Clinical Trials Focusing on Targeting the IGF-IR
9. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- DeVita, V.T.; Lawrence, T.S.; Rosenberg, S.A. DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology, 10th ed.; Wolters Kluwer Health: Philadelphia, PA, USA, 2015; pp. 1241–1313. [Google Scholar]
- Chibon, F.; Auria, S.A.; Coindre, J.M. Sarcomas Genetics: From Point Mutation to Complex Karyotype, from Diagnosis to Therapies. In Cancer Genomics, 1st ed.; Pfeffer, U., Ed.; Springer: Dordrecht, The Netherlands, 2013; pp. 429–452. [Google Scholar]
- Ferrari, A.; Sultan, I.; Huang, T.T.; Rodriguez-Galindo, C.; Shehadeh, A.; Meazza, C.; Ness, K.K.; Casanova, M.; Spunt, S.L. Soft tissue sarcoma across the age spectrum: A population-based study from the Surveillance Epidemiology and End Results database. Pediatr. Blood Cancer 2011, 57, 943–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amer, K.M.; Munn, M.; Congiusta, D.; Abraham, J.A.; Basu Mallick, A. Survival and Prognosis of Chondrosarcoma Subtypes: SEER Database Analysis. J. Orthop. Res. 2020, 38, 311–319. [Google Scholar] [CrossRef] [PubMed]
- Spjut, H.J.; Ayala, A.G. Skeletal tumors in children and adolescents. Hum. Pathol. 1983, 14, 628–642. [Google Scholar] [CrossRef]
- Hatina, J.; Kripnerova, M.; Houfkova, K.; Pesta, M.; Kuncova, J.; Sana, J.; Slaby, O.; Rodriguez, R. Sarcoma Stem Cell Heterogeneity. Adv. Exp. Med. Biol. 2019, 1123, 95–118. [Google Scholar]
- Yang, J.; Ren, Z.; Du, X.; Hao, M.; Zhou, W. The role of mesenchymal stem/progenitor cells in sarcoma: Update and dispute. Stem. Cell Investig. 2014, 1, 18. [Google Scholar] [PubMed]
- Amin, M.B.; Edge, S.B.; Greene, F.L.; Byrd, D.R.; Brookland, R.K.; Washington, M.K.; Gershenwald, J.E.; Compton, C.C.; Hess, K.R.; Sullivan, D.C.; et al. AJCC Cancer Staging Manual, 8th ed.; Springer International Publishing: Chicago, IL, USA, 2017; pp. 471–548. [Google Scholar]
- Casali, P.G.; Bielack, S.; Abecassis, N.; Aro, H.T.; Bauer, S.; Biagini, R.; Bonvalot, S.; Boukovinas, I.; Bovee, J.; Brennan, B.; et al. Bone sarcomas: ESMO-PaedCan-EURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29 (Suppl. 4), iv79–iv95. [Google Scholar] [CrossRef]
- Stiller, C.A.; Trama, A.; Serraino, D.; Rossi, S.; Navarro, C.; Chirlaque, M.D.; Casali, P.G. Descriptive epidemiology of sarcomas in Europe: Report from the RARECARE project. Eur. J. Cancer 2013, 49, 684–695. [Google Scholar] [CrossRef]
- Fletcher, C.D. The evolving classification of soft tissue tumours—An update based on the new 2013 WHO classification. Histopathology 2014, 64, 2–11. [Google Scholar] [CrossRef]
- Valery, P.C.; Laversanne, M.; Bray, F. Bone cancer incidence by morphological subtype: A global assessment. Cancer Causes Control 2015, 26, 1127–1139. [Google Scholar] [CrossRef]
- Bielack, S.S.; Kempf-Bielack, B.; Delling, G.; Exner, G.U.; Flege, S.; Helmke, K.; Kotz, R.; Salzer-Kuntschik, M.; Werner, M.; Winkelmann, W.; et al. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: An analysis of 1702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J. Clin. Oncol. 2002, 20, 776–790. [Google Scholar] [CrossRef]
- Shin, S.H.; Jeong, H.J.; Han, I.; Cho, H.S.; Kim, H.S. Osteosarcoma and chondrosarcoma of the shoulder: Site-specific comparative analysis. Orthopedics 2013, 36, e179–e185. [Google Scholar] [CrossRef] [Green Version]
- Sampo, M.; Koivikko, M.; Taskinen, M.; Kallio, P.; Kivioja, A.; Tarkkanen, M.; Bohling, T. Incidence, epidemiology and treatment results of osteosarcoma in Finland—A nationwide population-based study. Acta Oncol. 2011, 50, 1206–1214. [Google Scholar] [CrossRef] [Green Version]
- Smeland, S.; Bielack, S.S.; Whelan, J.; Bernstein, M.; Hogendoorn, P.; Krailo, M.D.; Gorlick, R.; Janeway, K.A.; Ingleby, F.C.; Anninga, J.; et al. Survival and prognosis with osteosarcoma: Outcomes in more than 2000 patients in the EURAMOS-1 (European and American Osteosarcoma Study) cohort. Eur. J. Cancer 2019, 109, 36–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Whelan, J.; Seddon, B.; Perisoglou, M. Management of osteosarcoma. Curr. Treat. Options Oncol. 2006, 7, 444–455. [Google Scholar] [CrossRef] [PubMed]
- Smrke, A.; Anderson, P.M.; Gulia, A.; Gennatas, S.; Huang, P.H.; Jones, R.L. Future Directions in the Treatment of Osteosarcoma. Cells 2021, 10, 172. [Google Scholar] [CrossRef] [PubMed]
- Whelan, J.; McTiernan, A.; Cooper, N.; Wong, Y.K.; Francis, M.; Vernon, S.; Strauss, S.J. Incidence and survival of malignant bone sarcomas in England 1979–2007. Int. J. Cancer 2012, 131, E508–E517. [Google Scholar] [CrossRef] [PubMed]
- Shemesh, S.S.; Pretell-Mazzini, J.; Quartin, P.A.J.; Rutenberg, T.F.; Conway, S.A. Surgical treatment of low-grade chondrosarcoma involving the appendicular skeleton: Long-term functional and oncological outcomes. Arch. Orthop. Trauma Surg. 2019, 139, 1659–1666. [Google Scholar] [CrossRef] [PubMed]
- Gelderblom, H.; Hogendoorn, P.C.; Dijkstra, S.D.; van Rijswijk, C.S.; Krol, A.D.; Taminiau, A.H.; Bovee, J.V. The clinical approach towards chondrosarcoma. Oncologist 2008, 13, 320–329. [Google Scholar] [CrossRef]
- Dickey, I.D.; Rose, P.S.; Fuchs, B.; Wold, L.E.; Okuno, S.H.; Sim, F.H.; Scully, S.P. Dedifferentiated chondrosarcoma: The role of chemotherapy with updated outcomes. J. Bone Joint Surg. Am. 2004, 86, 2412–2418. [Google Scholar] [CrossRef]
- Italiano, A.; Mir, O.; Cioffi, A.; Palmerini, E.; Piperno-Neumann, S.; Perrin, C.; Chaigneau, L.; Penel, N.; Duffaud, F.; Kurtz, J.E.; et al. Advanced chondrosarcomas: Role of chemotherapy and survival. Ann. Oncol. 2013, 24, 2916–2922. [Google Scholar] [CrossRef]
- Mitchell, A.D.; Ayoub, K.; Mangham, D.C.; Grimer, R.J.; Carter, S.R.; Tillman, R.M. Experience in the treatment of dedifferentiated chondrosarcoma. J. Bone Joint Surg. Br. 2000, 82, 55–61. [Google Scholar] [CrossRef] [PubMed]
- Noel, G.; Feuvret, L.; Ferrand, R.; Boisserie, G.; Mazeron, J.J.; Habrand, J.L. Radiotherapeutic factors in the management of cervical-basal chordomas and chondrosarcomas. Neurosurgery 2004, 55, 1252–1260, discussion 1260–1262. [Google Scholar] [CrossRef]
- Rizzo, M.; Ghert, M.A.; Harrelson, J.M.; Scully, S.P. Chondrosarcoma of bone: Analysis of 108 cases and evaluation for predictors of outcome. Clin. Orthop. Relat. Res. 2001, 391, 224–233. [Google Scholar] [CrossRef]
- Grunewald, T.G.P.; Cidre-Aranaz, F.; Surdez, D.; Tomazou, E.M.; de Alava, E.; Kovar, H.; Sorensen, P.H.; Delattre, O.; Dirksen, U. Ewing sarcoma. Nat. Rev. Dis Primers 2018, 4, 5. [Google Scholar] [CrossRef] [PubMed]
- Doyle, L.A. Sarcoma classification: An update based on the 2013 World Health Organization Classification of Tumors of Soft Tissue and Bone. Cancer 2014, 120, 1763–1774. [Google Scholar] [CrossRef]
- Watson, S.; Perrin, V.; Guillemot, D.; Reynaud, S.; Coindre, J.M.; Karanian, M.; Guinebretiere, J.M.; Freneaux, P.; Le Loarer, F.; Bouvet, M.; et al. Transcriptomic definition of molecular subgroups of small round cell sarcomas. J. Pathol. 2018, 245, 29–40. [Google Scholar] [CrossRef] [Green Version]
- Grobner, S.N.; Worst, B.C.; Weischenfeldt, J.; Buchhalter, I.; Kleinheinz, K.; Rudneva, V.A.; Johann, P.D.; Balasubramanian, G.P.; Segura-Wang, M.; Brabetz, S.; et al. The landscape of genomic alterations across childhood cancers. Nature 2018, 555, 321–327. [Google Scholar] [CrossRef] [Green Version]
- Tirode, F.; Surdez, D.; Ma, X.; Parker, M.; Le Deley, M.C.; Bahrami, A.; Zhang, Z.; Lapouble, E.; Grossetete-Lalami, S.; Rusch, M.; et al. Genomic landscape of Ewing sarcoma defines an aggressive subtype with co-association of STAG2 and TP53 mutations. Cancer Discov. 2014, 4, 1342–1353. [Google Scholar] [CrossRef] [Green Version]
- Musa, J.; Cidre-Aranaz, F.; Aynaud, M.M.; Orth, M.F.; Knott, M.M.L.; Mirabeau, O.; Mazor, G.; Varon, M.; Holting, T.L.B.; Grossetete, S.; et al. Cooperation of cancer drivers with regulatory germline variants shapes clinical outcomes. Nat. Commun. 2019, 10, 4128. [Google Scholar] [CrossRef] [Green Version]
- Grunewald, T.G.; Bernard, V.; Gilardi-Hebenstreit, P.; Raynal, V.; Surdez, D.; Aynaud, M.M.; Mirabeau, O.; Cidre-Aranaz, F.; Tirode, F.; Zaidi, S.; et al. Chimeric EWSR1-FLI1 regulates the Ewing sarcoma susceptibility gene EGR2 via a GGAA microsatellite. Nat. Genet. 2015, 47, 1073–1078. [Google Scholar] [CrossRef] [Green Version]
- Subbiah, V.; Anderson, P.; Lazar, A.J.; Burdett, E.; Raymond, K.; Ludwig, J.A. Ewing’s sarcoma: Standard and experimental treatment options. Curr. Treat. Options Oncol. 2009, 10, 126–140. [Google Scholar] [CrossRef]
- Balamuth, N.J.; Womer, R.B. Ewing’s sarcoma. Lancet Oncol. 2010, 11, 184–192. [Google Scholar] [CrossRef]
- Li, Y.S.; Liu, Q.; He, H.B.; Luo, W. The possible role of insulin-like growth factor-1 in osteosarcoma. Curr. Probl. Cancer 2019, 43, 228–235. [Google Scholar] [CrossRef] [PubMed]
- Ferreira, A.J.; Boldrini, E.; Lopez, R.V.M.; Scapulatempo Neto, C.; Santos, J.F.C.; Lopes, L.F. Characterization, survival analysis, and expression of IGFR in tumor samples from patients diagnosed with Ewing family tumors treated at the Barretos Cancer Hospital. Rev. Bras. Ortop. 2017, 52, 705–713. [Google Scholar] [CrossRef]
- Beech, D.; Pollock, R.E.; Tsan, R.; Radinsky, R. Epidermal growth factor receptor and insulin-like growth factor-I receptor expression and function in human soft-tissue sarcoma cells. Int. J. Oncol. 1998, 12, 329–336. [Google Scholar] [CrossRef] [PubMed]
- Frysak, Z.; Schovanek, J.; Iacobone, M.; Karasek, D. Insulin-like Growth Factors in a clinical setting: Review of IGF-I. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc. Czech. Repub. 2015, 159, 347–351. [Google Scholar] [CrossRef]
- LeRoith, D.; Roberts, C.T., Jr. The insulin-like growth factor system and cancer. Cancer Lett. 2003, 195, 127–137. [Google Scholar] [CrossRef] [Green Version]
- Brahmkhatri, V.P.; Prasanna, C.; Atreya, H.S. Insulin-like growth factor system in cancer: Novel targeted therapies. Biomed. Res. Int 2015, 2015, 538019. [Google Scholar] [CrossRef] [Green Version]
- Vigneri, P.G.; Tirro, E.; Pennisi, M.S.; Massimino, M.; Stella, S.; Romano, C.; Manzella, L. The Insulin/IGF System in Colorectal Cancer Development and Resistance to Therapy. Front. Oncol. 2015, 5, 230. [Google Scholar] [CrossRef] [Green Version]
- Scheven, B.A.; Hamilton, N.J.; Fakkeldij, T.M.; Duursma, S.A. Effects of recombinant human insulin-like growth factor I and II (IGF-I/-II) and growth hormone (GH) on the growth of normal adult human osteoblast-like cells and human osteogenic sarcoma cells. Growth Regul. 1991, 1, 160–167. [Google Scholar]
- Renehan, A.G.; Zwahlen, M.; Minder, C.; O’Dwyer, S.T.; Shalet, S.M.; Egger, M. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: Systematic review and meta-regression analysis. Lancet 2004, 363, 1346–1353. [Google Scholar] [CrossRef]
- Kasprzak, A.; Adamek, A. Insulin-Like Growth Factor 2 (IGF2) Signaling in Colorectal Cancer-From Basic Research to Potential Clinical Applications. Int. J. Mol. Sci. 2019, 20, 4915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baxter, R.C. Insulin-like growth factor (IGF)-binding proteins: Interactions with IGFs and intrinsic bioactivities. Am. J. Physiol. Endocrinol. Metab. 2000, 278, E967–E976. [Google Scholar] [CrossRef] [PubMed]
- Clemmons, D.R. Role of insulin-like growth factor binding proteins in controlling IGF actions. Mol. Cell Endocrinol. 1998, 140, 19–24. [Google Scholar] [CrossRef]
- Grimberg, A.; Cohen, P. Role of insulin-like growth factors and their binding proteins in growth control and carcinogenesis. J. Cell Physiol. 2000, 183, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Han, J.; Jogie-Brahim, S.; Harada, A.; Oh, Y. Insulin-like growth factor-binding protein-3 suppresses tumor growth via activation of caspase-dependent apoptosis and cross-talk with NF-kappaB signaling. Cancer Lett. 2011, 307, 200–210. [Google Scholar] [CrossRef]
- Miyamoto, S.; Yano, K.; Sugimoto, S.; Ishii, G.; Hasebe, T.; Endoh, Y.; Kodama, K.; Goya, M.; Chiba, T.; Ochiai, A. Matrix metalloproteinase-7 facilitates insulin-like growth factor bioavailability through its proteinase activity on insulin-like growth factor binding protein 3. Cancer Res. 2004, 64, 665–671. [Google Scholar] [CrossRef] [Green Version]
- Favelyukis, S.; Till, J.H.; Hubbard, S.R.; Miller, W.T. Structure and autoregulation of the insulin-like growth factor 1 receptor kinase. Nat. Struct. Biol. 2001, 8, 1058–1063. [Google Scholar] [CrossRef]
- Janssen, J. New Insights from IGF-IR Stimulating Activity Analyses: Pathological Considerations. Cells 2020, 9, 862. [Google Scholar] [CrossRef] [Green Version]
- Girnita, L.; Worrall, C.; Takahashi, S.; Seregard, S.; Girnita, A. Something old, something new and something borrowed: Emerging paradigm of insulin-like growth factor type 1 receptor (IGF-1R) signaling regulation. Cell Mol. Life Sci. 2014, 71, 2403–2427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salisbury, T.B.; Tomblin, J.K. Insulin/Insulin-like growth factors in cancer: New roles for the aryl hydrocarbon receptor, tumor resistance mechanisms, and new blocking strategies. Front. Endocrinol. 2015, 6, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riedemann, J.; Macaulay, V.M. IGF1R signalling and its inhibition. Endocr. Relat. Cancer 2006, 13 (Suppl. 1), S33–S43. [Google Scholar] [CrossRef]
- Klement, R.J.; Fink, M.K. Dietary and pharmacological modification of the insulin/IGF-1 system: Exploiting the full repertoire against cancer. Oncogenesis 2016, 5, e193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chughtai, S. The nuclear translocation of insulin-like growth factor receptor and its significance in cancer cell survival. Cell Biochem. Funct. 2020, 38, 347–351. [Google Scholar] [CrossRef] [PubMed]
- Aleksic, T.; Chitnis, M.M.; Perestenko, O.V.; Gao, S.; Thomas, P.H.; Turner, G.D.; Protheroe, A.S.; Howarth, M.; Macaulay, V.M. Type 1 insulin-like growth factor receptor translocates to the nucleus of human tumor cells. Cancer Res. 2010, 70, 6412–6419. [Google Scholar] [CrossRef] [Green Version]
- Warsito, D.; Sjostrom, S.; Andersson, S.; Larsson, O.; Sehat, B. Nuclear IGF1R is a transcriptional co-activator of LEF1/TCF. EMBO Rep. 2012, 13, 244–250. [Google Scholar] [CrossRef] [Green Version]
- Lin, Y.; Liu, H.; Waraky, A.; Haglund, F.; Agarwal, P.; Jernberg-Wiklund, H.; Warsito, D.; Larsson, O. SUMO-modified insulin-like growth factor 1 receptor (IGF-1R) increases cell cycle progression and cell proliferation. J. Cell Physiol. 2017, 232, 2722–2730. [Google Scholar] [CrossRef] [Green Version]
- Voudouri, K.; Nikitovic, D.; Berdiaki, A.; Kletsas, D.; Karamanos, N.K.; Tzanakakis, G.N. IGF-I/EGF and E2 signaling crosstalk through IGF-IR conduit point affects breast cancer cell adhesion. Matrix Biol. 2016, 56, 95–113. [Google Scholar] [CrossRef]
- Skandalis, S.S.; Afratis, N.; Smirlaki, G.; Nikitovic, D.; Theocharis, A.D.; Tzanakakis, G.N.; Karamanos, N.K. Cross-talk between estradiol receptor and EGFR/IGF-IR signaling pathways in estrogen-responsive breast cancers: Focus on the role and impact of proteoglycans. Matrix Biol. 2014, 35, 182–193. [Google Scholar] [CrossRef]
- De Groot, S.; Rottgering, B.; Gelderblom, H.; Pijl, H.; Szuhai, K.; Kroep, J.R. Unraveling the Resistance of IGF-Pathway Inhibition in Ewing Sarcoma. Cancers 2020, 12, 3568. [Google Scholar] [CrossRef]
- Burrow, S.; Andrulis, I.L.; Pollak, M.; Bell, R.S. Expression of insulin-like growth factor receptor, IGF-1, and IGF-2 in primary and metastatic osteosarcoma. J. Surg. Oncol. 1998, 69, 21–27. [Google Scholar] [CrossRef]
- Abramovitch, S.; Werner, H. Functional and physical interactions between BRCA1 and p53 in transcriptional regulation of the IGF-IR gene. Horm. Metab. Res. 2003, 35, 758–762. [Google Scholar] [PubMed]
- Datta, K.; Nambudripad, R.; Pal, S.; Zhou, M.; Cohen, H.T.; Mukhopadhyay, D. Inhibition of insulin-like growth factor-I-mediated cell signaling by the von Hippel-Lindau gene product in renal cancer. J. Biol. Chem. 2000, 275, 20700–20706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toretsky, J.A.; Kalebic, T.; Blakesley, V.; LeRoith, D.; Helman, L.J. The insulin-like growth factor-I receptor is required for EWS/FLI-1 transformation of fibroblasts. J. Biol. Chem. 1997, 272, 30822–30827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prieur, A.; Tirode, F.; Cohen, P.; Delattre, O. EWS/FLI-1 silencing and gene profiling of Ewing cells reveal downstream oncogenic pathways and a crucial role for repression of insulin-like growth factor binding protein 3. Mol. Cell Biol. 2004, 24, 7275–7283. [Google Scholar] [CrossRef] [Green Version]
- Dynkevich, Y.; Rother, K.I.; Whitford, I.; Qureshi, S.; Galiveeti, S.; Szulc, A.L.; Danoff, A.; Breen, T.L.; Kaviani, N.; Shanik, M.H.; et al. Tumors, IGF-2, and hypoglycemia: Insights from the clinic, the laboratory, and the historical archive. Endocr. Rev. 2013, 34, 798–826. [Google Scholar] [CrossRef] [Green Version]
- Liang, J.; Li, B.; Yuan, L.; Ye, Z. Prognostic value of IGF-1R expression in bone and soft tissue sarcomas: A meta-analysis. Onco Targets Ther. 2015, 8, 1949–1955. [Google Scholar]
- Jentzsch, T.; Robl, B.; Husmann, M.; Bode-Lesniewska, B.; Fuchs, B. Worse prognosis of osteosarcoma patients expressing IGF-1 on a tissue microarray. Anticancer Res. 2014, 34, 3881–3889. [Google Scholar]
- Wang, Y.H.; Han, X.D.; Qiu, Y.; Xiong, J.; Yu, Y.; Wang, B.; Zhu, Z.Z.; Qian, B.P.; Chen, Y.X.; Wang, S.F.; et al. Increased expression of insulin-like growth factor-1 receptor is correlated with tumor metastasis and prognosis in patients with osteosarcoma. J. Surg. Oncol. 2012, 105, 235–243. [Google Scholar] [CrossRef]
- Hong, S.H.; Briggs, J.; Newman, R.; Hoffman, K.; Mendoza, A.; LeRoith, D.; Helman, L.; Yakar, S.; Khanna, C. Murine osteosarcoma primary tumour growth and metastatic progression is maintained after marked suppression of serum insulin-like growth factor I. Int. J. Cancer 2009, 124, 2042–2049. [Google Scholar] [CrossRef]
- Viereck, V.; Siggelkow, H.; Pannem, R.; Braulke, T.; Scharf, J.G.; Kubler, B. Alteration of the insulin-like growth factor axis during in vitro differentiation of the human osteosarcoma cell line HOS 58. J. Cell Biochem. 2007, 102, 28–40. [Google Scholar] [CrossRef] [PubMed]
- Behjati, S.; Tarpey, P.S.; Haase, K.; Ye, H.; Young, M.D.; Alexandrov, L.B.; Farndon, S.J.; Collord, G.; Wedge, D.C.; Martincorena, I.; et al. Recurrent mutation of IGF signalling genes and distinct patterns of genomic rearrangement in osteosarcoma. Nat. Commun. 2017, 8, 15936. [Google Scholar] [CrossRef] [PubMed]
- Luther, G.A.; Lamplot, J.; Chen, X.; Rames, R.; Wagner, E.R.; Liu, X.; Parekh, A.; Huang, E.; Kim, S.H.; Shen, J.; et al. IGFBP5 domains exert distinct inhibitory effects on the tumorigenicity and metastasis of human osteosarcoma. Cancer Lett. 2013, 336, 222–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, C.K.; Oh, S.; Kim, S.J.; Leem, S.H.; Heo, J.; Chung, S.H. Correlation of IGF1R expression with ABCG2 and CD44 expressions in human osteosarcoma. Genes Genom. 2018, 40, 381–388. [Google Scholar] [CrossRef]
- Orlando, B.J.; Liao, M. ABCG2 transports anticancer drugs via a closed-to-open switch. Nat. Commun. 2020, 11, 2264. [Google Scholar] [CrossRef]
- Mao, J.; Zhuang, G.; Chen, Z. Genetic Polymorphisms of Insulin-Like Growth Factor 1 Are Associated with Osteosarcoma Risk and Prognosis. Med. Sci. Monit. 2017, 23, 5892–5898. [Google Scholar] [CrossRef] [Green Version]
- Asmane, I.; Watkin, E.; Alberti, L.; Duc, A.; Marec-Berard, P.; Ray-Coquard, I.; Cassier, P.; Decouvelaere, A.V.; Ranchere, D.; Kurtz, J.E.; et al. Insulin-like growth factor type 1 receptor (IGF-1R) exclusive nuclear staining: A predictive biomarker for IGF-1R monoclonal antibody (Ab) therapy in sarcomas. Eur. J. Cancer 2012, 48, 3027–3035. [Google Scholar] [CrossRef]
- Gianferante, D.M.; Mirabello, L.; Savage, S.A. Germline and somatic genetics of osteosarcoma—Connecting aetiology, biology and therapy. Nat. Rev. Endocrinol. 2017, 13, 480–491. [Google Scholar] [CrossRef]
- Savage, S.A.; Mirabello, L. Bone cancer: Is the osteosarcoma genome targetable? Nat. Rev. Endocrinol. 2017, 13, 506–508. [Google Scholar] [CrossRef]
- Ho, L.; Stojanovski, A.; Whetstone, H.; Wei, Q.X.; Mau, E.; Wunder, J.S.; Alman, B. Gli2 and p53 cooperate to regulate IGFBP-3- mediated chondrocyte apoptosis in the progression from benign to malignant cartilage tumors. Cancer Cell 2009, 16, 126–136. [Google Scholar] [CrossRef] [Green Version]
- Xie, H.; Mao, J.S.; Hu, W.F. Insulin-Like Growth Factor 1 (IGF1) Pathway Member Polymorphisms Are Associated with Risk and Prognosis of Chondrosarcoma. Med. Sci. Monit. 2020, 26, e923853. [Google Scholar] [CrossRef]
- Scotlandi, K.; Benini, S.; Sarti, M.; Serra, M.; Lollini, P.L.; Maurici, D.; Picci, P.; Manara, M.C.; Baldini, N. Insulin-like growth factor I receptor-mediated circuit in Ewing’s sarcoma/peripheral neuroectodermal tumor: A possible therapeutic target. Cancer Res. 1996, 56, 4570–4574. [Google Scholar]
- Herrero-Martin, D.; Osuna, D.; Ordonez, J.L.; Sevillano, V.; Martins, A.S.; Mackintosh, C.; Campos, M.; Madoz-Gurpide, J.; Otero-Motta, A.P.; Caballero, G.; et al. Stable interference of EWS-FLI1 in an Ewing sarcoma cell line impairs IGF-1/IGF-1R signalling and reveals TOPK as a new target. Br. J. Cancer 2009, 101, 80–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moritake, H.; Saito, Y.; Sawa, D.; Sameshima, N.; Yamada, A.; Kinoshita, M.; Kamimura, S.; Konomoto, T.; Nunoi, H. TAE226, a dual inhibitor of focal adhesion kinase and insulin-like growth factor-I receptor, is effective for Ewing sarcoma. Cancer Med. 2019, 8, 7809–7821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakai, S.; Yamada, S.; Outani, H.; Nakai, T.; Yasuda, N.; Mae, H.; Imura, Y.; Wakamatsu, T.; Tamiya, H.; Tanaka, T.; et al. Author Correction: Establishment of a novel human CIC-DUX4 sarcoma cell line, Kitra-SRS, with autocrine IGF-1R activation and metastatic potential to the lungs. Sci. Rep. 2020, 10, 684. [Google Scholar] [CrossRef] [PubMed]
- Tzanakakis, G.; Giatagana, E.M.; Kuskov, A.; Berdiaki, A.; Tsatsakis, A.M.; Neagu, M.; Nikitovic, D. Proteoglycans in the Pathogenesis of Hormone-Dependent Cancers: Mediators and Effectors. Cancers 2020, 12, 2401. [Google Scholar] [CrossRef]
- Joshi, R.S.; Kanugula, S.S.; Sudhir, S.; Pereira, M.P.; Jain, S.; Aghi, M.K. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers 2021, 13, 1399. [Google Scholar] [CrossRef]
- Nikitovic, D.; Berdiaki, A.; Spyridaki, I.; Krasanakis, T.; Tsatsakis, A.; Tzanakakis, G.N. Proteoglycans-Biomarkers and Targets in Cancer Therapy. Front. Endocrinol. 2018, 9, 69. [Google Scholar] [CrossRef] [Green Version]
- Pinto, A.; Dickman, P.; Parham, D. Pathobiologic markers of the ewing sarcoma family of tumors: State of the art and prediction of behaviour. Sarcoma 2011, 2011, 856190. [Google Scholar] [CrossRef] [Green Version]
- Koo, J.; Hayashi, M.; Verneris, M.R.; Lee-Sherick, A.B. Targeting Tumor-Associated Macrophages in the Pediatric Sarcoma Tumor Microenvironment. Front. Oncol. 2020, 10, 581107. [Google Scholar] [CrossRef] [PubMed]
- Hynes, R.O. The extracellular matrix: Not just pretty fibrils. Science 2009, 326, 1216–1219. [Google Scholar] [CrossRef] [Green Version]
- Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [Green Version]
- Berdiaki, A.; Neagu, M.; Giatagana, E.-M.; Kuskov, A.; Tsatsakis, A.M.; Tzanakakis, G.N.; Nikitovic, D. Glycosaminoglycans: Carriers and Targets for Tailored Anti-Cancer Therapy. Biomolecules 2021, 11, 395. [Google Scholar] [CrossRef] [PubMed]
- Nikitovic, D.; Berdiaki, A.; Zafiropoulos, A.; Katonis, P.; Tsatsakis, A.; Karamanos, N.K.; Tzanakakis, G.N. Lumican expression is positively correlated with the differentiation and negatively with the growth of human osteosarcoma cells. FEBS J. 2008, 275, 350–361. [Google Scholar] [CrossRef]
- Cui, J.; Dean, D.; Hornicek, F.J.; Chen, Z.; Duan, Z. The role of extracelluar matrix in osteosarcoma progression and metastasis. J. Exp. Clin. Cancer Res. 2020, 39, 178. [Google Scholar] [CrossRef] [PubMed]
- Nikitovic, D.; Aggelidakis, J.; Young, M.F.; Iozzo, R.V.; Karamanos, N.K.; Tzanakakis, G.N. The biology of small leucine-rich proteoglycans in bone pathophysiology. J. Biol. Chem. 2012, 287, 33926–33933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bianco, P.; Fisher, L.W.; Young, M.F.; Termine, J.D.; Robey, P.G. Expression and localization of the two small proteoglycans biglycan and decorin in developing human skeletal and non-skeletal tissues. J. Histochem. Cytochem. 1990, 38, 1549–1563. [Google Scholar] [CrossRef] [Green Version]
- Nikitovic, D.; Berdiaki, K.; Chalkiadaki, G.; Karamanos, N.; Tzanakakis, G. The role of SLRP-proteoglycans in osteosarcoma pathogenesis. Connect. Tissue Res. 2008, 49, 235–238. [Google Scholar] [CrossRef] [PubMed]
- Nikitovic, D.; Chalkiadaki, G.; Berdiaki, A.; Aggelidakis, J.; Katonis, P.; Karamanos, N.K.; Tzanakakis, G.N. Lumican regulates osteosarcoma cell adhesion by modulating TGFbeta2 activity. Int. J. Biochem. Cell Biol. 2011, 43, 928–935. [Google Scholar] [CrossRef]
- Ho, X.D.; Phung, P.Q.; Le, V.H.; Nguyen, V.; Reimann, E.; Prans, E.; Kõks, G.; Maasalu, K.; Le, N.T.; HTrinh, L.; et al. Whole transcriptome analysis identifies differentially regulated networks between osteosarcoma and normal bone samples. Exp. Biol. Med. 2017, 242, 1802–1811. [Google Scholar] [CrossRef] [PubMed]
- Li, F.; Li, S.; Cheng, T. TGF-beta1 promotes osteosarcoma cell migration and invasion through the miR-143-versican pathway. Cell Physiol. Biochem. 2014, 34, 2169–2179. [Google Scholar] [CrossRef] [PubMed]
- Bergh, P.; Gunterberg, B.; Meis-Kindblom, J.M.; Kindblom, L.G. Prognostic factors and outcome of pelvic, sacral, and spinal chondrosarcomas: A center-based study of 69 cases. Cancer 2001, 91, 1201–1212. [Google Scholar] [CrossRef]
- Pring, M.E.; Weber, K.L.; Unni, K.K.; Sim, F.H. Chondrosarcoma of the pelvis. A review of sixty-four cases. J. Bone Joint Surg. Am. 2001, 83, 1630–1642. [Google Scholar] [CrossRef]
- Skubitz, K.M.; D’Adamo, D.R. Sarcoma. Mayo Clin. Proc. 2007, 82, 1409–1432. [Google Scholar] [CrossRef] [PubMed]
- Riedel, R.F.; Larrier, N.; Dodd, L.; Kirsch, D.; Martinez, S.; Brigman, B.E. The clinical management of chondrosarcoma. Curr. Treat. Options Oncol. 2009, 10, 94–106. [Google Scholar] [CrossRef]
- Totoki, Y.; Yoshida, A.; Hosoda, F.; Nakamura, H.; Hama, N.; Ogura, K.; Fujiwara, T.; Arai, Y.; Toguchida, J.; Tsuda, H.; et al. Unique mutation portraits and frequent COL2A1 gene alteration in chondrosarcoma. Genome Res. 2014, 24, 1411–1420. [Google Scholar] [CrossRef] [Green Version]
- Amary, F.; Perez-Casanova, L.; Ye, H.; Cottone, L.; Strobl, A.C.; Cool, P.; Miranda, E.; Berisha, F.; Aston, W.; Rocha, M.; et al. Synovial chondromatosis and soft tissue chondroma: Extraosseous cartilaginous tumor defined by FN1 gene rearrangement. Mod. Pathol. 2019, 32, 1762–1771. [Google Scholar] [CrossRef]
- Soderstrom, M.; Bohling, T.; Ekfors, T.; Nelimarkka, L.; Aro, H.T.; Vuorio, E. Molecular profiling of human chondrosarcomas for matrix production and cancer markers. Int. J. Cancer 2002, 100, 144–151. [Google Scholar] [CrossRef]
- Aigner, T.; Dertinger, S.; Vornehm, S.I.; Dudhia, J.; von der Mark, K.; Kirchner, T. Phenotypic diversity of neoplastic chondrocytes and extracellular matrix gene expression in cartilaginous neoplasms. Am. J. Pathol. 1997, 150, 2133–2141. [Google Scholar]
- Kawashima, A.; Ueda, Y.; Tsuchiya, H.; Tomita, K.; Nagai, Y.; Nakanishi, I. Immunohistochemical localization of collagenous proteins in cartilaginous tumors: Characteristic distribution of type IX collagen. J. Cancer Res. Clin. Oncol. 1993, 120, 35–40. [Google Scholar] [CrossRef]
- Monga, V.; Mani, H.; Hirbe, A.; Milhem, M. Non-Conventional Treatments for Conventional Chondrosarcoma. Cancers 2020, 12, 1962. [Google Scholar] [CrossRef]
- Zając, A.; Król, S.K.; Rutkowski, P.; Czarnecka, A.M. Biological Heterogeneity of Chondrosarcoma: From (Epi) Genetics through Stemness and Deregulated Signaling to Immunophenotype. Cancers 2021, 13, 1317. [Google Scholar] [CrossRef]
- Peyrode, C.; Weber, V.; David, E.; Vidal, A.; Auzeloux, P.; Communal, Y.; Dauplat, M.M.; Besse, S.; Gouin, F.; Heymann, D.; et al. Quaternary ammonium-melphalan conjugate for anticancer therapy of chondrosarcoma: In vitro and in vivo preclinical studies. Investig. New Drugs 2012, 30, 1782–1790. [Google Scholar] [CrossRef] [PubMed]
- Theocharis, A.D.; Manou, D.; Karamanos, N.K. The extracellular matrix as a multitasking player in disease. FEBS J. 2019, 286, 2830–2869. [Google Scholar] [CrossRef] [Green Version]
- Yin, Y.; Han, Y.; Shi, C.; Xia, Z. IGF-1 regulates the growth of fibroblasts and extracellular matrix deposition in pelvic organ prolapse. Open Med. 2020, 15, 833–840. [Google Scholar] [CrossRef] [PubMed]
- Aggelidakis, J.; Berdiaki, A.; Nikitovic, D.; Papoutsidakis, A.; Papachristou, D.J.; Tsatsakis, A.M.; Tzanakakis, G.N. Biglycan Regulates MG63 Osteosarcoma Cell Growth Through a LPR6/beta-Catenin/IGFR-IR Signaling Axis. Front. Oncol. 2018, 8, 470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Travascio, F.; Elmasry, S.; Asfour, S. Modeling the role of IGF-1 on extracellular matrix biosynthesis and cellularity in intervertebral disc. J. Biomech. 2014, 47, 2269–2276. [Google Scholar] [CrossRef] [PubMed]
- Reiser, K.; Summers, P.; Medrano, J.F.; Rucker, R.; Last, J.; McDonald, R. Effects of elevated circulating IGF-1 on the extracellular matrix in “high-growth” C57BL/6J mice. Am. J. Physiol. 1996, 271, R696–R703. [Google Scholar] [CrossRef]
- Grässel, S.; Aszódi, A. Cartilage. In Physiology and Development; Springer: Chicago, IL, USA, 2017. [Google Scholar]
- Iozzo, R.V.; Schaefer, L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol. 2015, 42, 11–55. [Google Scholar] [CrossRef]
- Takigawa, M.; Okawa, T.; Pan, H.; Aoki, C.; Takahashi, K.; Zue, J.; Suzuki, F.; Kinoshita, A. Insulin-like growth factors I and II are autocrine factors in stimulating proteoglycan synthesis, a marker of differentiated chondrocytes, acting through their respective receptors on a clonal human chondrosarcoma-derived chondrocyte cell line, HCS-2/8. Endocrinology 1997, 138, 4390–4400. [Google Scholar] [CrossRef]
- Iozzo, R.V. Matrix proteoglycans: From molecular design to cellular function. Annu. Rev. Biochem. 1998, 67, 609–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsui, Y.; Araki, N.; Tsuboi, H.; Tsumaki, N.; Nakata, K.; Kawabata, H.; Eyre, D.R.; Yoshikawa, H. Differential expression of aggrecan mRNA isoforms by chondrosarcoma cells. Anticancer Res. 2002, 22, 4169–4172. [Google Scholar] [PubMed]
- Hiraoka, K.; Zenmyo, M.; Komiya, S.; Kawabata, R.; Yokouchi, M.; Suzuki, R.; Hamada, T.; Kato, S.; Nagata, K. Relationship of p21 (waf1/cip1) and differentiation in chondrosarcoma cells. Virchows Arch. 2002, 440, 285–290. [Google Scholar] [CrossRef]
- Piltti, J.; Bygdell, J.; Qu, C.; Lammi, M.J. Effects of long-term low oxygen tension in human chondrosarcoma cells. J. Cell Biochem. 2018, 119, 2320–2332. [Google Scholar] [CrossRef] [PubMed]
- Harvey, A.K.; Yu, X.P.; Frolik, C.A.; Chandrasekhar, S. Parathyroid hormone-(1-34) enhances aggrecan synthesis via an insulin-like growth factor-I pathway. J. Biol. Chem. 1999, 274, 23249–23255. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Askew, E.B.; Knudson, C.B.; Knudson, W. CRISPR/Cas9 knockout of HAS2 in rat chondrosarcoma chondrocytes demonstrates the requirement of hyaluronan for aggrecan retention. Matrix Biol. 2016, 56, 74–94. [Google Scholar] [CrossRef] [Green Version]
- Fede, C.; Stecco, C.; Angelini, A.; Fan, C.; Belluzzi, E.; Pozzuoli, A.; Ruggieri, P.; De Caro, R. Variations in contents of hyaluronan in the peritumoral micro-environment of human chondrosarcoma. J. Orthop. Res. 2019, 37, 503–509. [Google Scholar] [CrossRef] [Green Version]
- Voissiere, A.; Weber, V.; Gerard, Y.; Redini, F.; Raes, F.; Chezal, J.M.; Degoul, F.; Peyrode, C.; Miot-Noirault, E. Proteoglycan-targeting applied to hypoxia-activated prodrug therapy in chondrosarcoma: First proof-of-concept. Oncotarget 2017, 8, 95824–95840. [Google Scholar] [CrossRef]
- Stevens, J.W.; Meyerholz, D.K.; Neighbors, J.D.; Morcuende, J.A. 5’-methylschweinfurthin G reduces chondrosarcoma tumor growth. J. Orthop. Res. 2018, 36, 1283–1293. [Google Scholar] [CrossRef] [Green Version]
- Gotting, C.; Kuhn, J.; Kleesiek, K. Human xylosyltransferases in health and disease. Cell. Mol. Life Sci. 2007, 64, 1498–1517. [Google Scholar] [CrossRef] [PubMed]
- Na, K.Y.; Bacchini, P.; Bertoni, F.; Kim, Y.W.; Park, Y.K. Syndecan-4 and fibronectin in osteosarcoma. Pathology 2012, 44, 325–330. [Google Scholar] [CrossRef]
- Huang, W.T.; Liu, A.G.; Cai, K.T.; He, R.Q.; Li, Z.; Wei, Q.J.; Chen, M.Y.; Huang, J.Y.; Yan, W.Y.; Zhou, H.; et al. Exploration and validation of downregulated microRNA-199a-3p, downstream messenger RNA targets and transcriptional regulation in osteosarcoma. Am. J. Transl. Res. 2019, 11, 7538–7554. [Google Scholar] [PubMed]
- Holmes, D.F.; Lu, Y.; Starborg, T.; Kadler, K.E. Collagen Fibril Assembly and Function. Curr. Top. Dev. Biol. 2018, 130, 107–142. [Google Scholar]
- Boraschi-Diaz, I.; Wang, J.; Mort, J.; Komarova, S. Collagen Type I as a Ligand for Receptor-Mediated Signaling. Front. Phys. 2017, 5. [Google Scholar] [CrossRef]
- Vogel, W.F. Collagen-receptor signaling in health and disease. Eur. J. Dermatol. 2001, 11, 506–514. [Google Scholar]
- Kudo, Y.; Iwashita, M.; Iguchi, T.; Takeda, Y. The regulation of type-I collagen synthesis by insulin-like growth factor-I in human osteoblast-like SaOS-2 cells. Pflugers Arch. 1996, 433, 123–128. [Google Scholar] [CrossRef]
- Damiens, C.; Grimaud, E.; Rousselle, A.V.; Charrier, C.; Fortun, Y.; Heymann, D.; Padrines, M. Cysteine protease production by human osteosarcoma cells (MG63, SAOS2) and its modulation by soluble factors. Cytokine 2000, 12, 539–542. [Google Scholar] [CrossRef]
- Nasu, M.; Sugimoto, T.; Kaji, H.; Chihara, K. Estrogen modulates osteoblast proliferation and function regulated by parathyroid hormone in osteoblastic SaOS-2 cells: Role of insulin-like growth factor (IGF)-I and IGF-binding protein-5. J. Endocrinol. 2000, 167, 305–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, H.; Han, K.L.; Wang, Z.Y.; Chen, Y.; Li, H.T.; Zeng, J.L.; Shen, Z.; Yao, Y. Value of C-telopeptide-cross-linked Type I collagen, osteocalcin, bone-specific alkaline phosphatase and procollagen Type I N-terminal propeptide in the diagnosis and prognosis of bone metastasis in patients with malignant tumors. Med. Sci. Monit. 2011, 17, CR626–CR633. [Google Scholar] [CrossRef] [Green Version]
- Ferreira, A.R.; Alho, I.; Shan, N.; Matias, M.; Faria, M.; Casimiro, S.; Leitzel, K.; Ali, S.; Lipton, A.; Costa, L. N-Telopeptide of Type I Collagen Long-Term Dynamics in Breast Cancer Patients with Bone Metastases: Clinical Outcomes and Influence of Extraskeletal Metastases. Oncologist 2016, 21, 1418–1426. [Google Scholar] [CrossRef] [Green Version]
- Gelse, K.; Poschl, E.; Aigner, T. Collagens--structure, function, and biosynthesis. Adv. Drug Deliv. Rev. 2003, 55, 1531–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, M.; Zhou, Q.; Liang, Q.Q.; Li, C.G.; Holz, J.D.; Tang, D.; Sheu, T.J.; Li, T.F.; Shi, Q.; Wang, Y.J. IGF-1 regulation of type II collagen and MMP-13 expression in rat endplate chondrocytes via distinct signaling pathways. Osteoarthr. Cartil. 2009, 17, 100–106. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Bryan, J.; Franz, C.; Havlioglu, N.; Sandell, L.J. Type IIB procollagen NH-propeptide induces death of tumor cells via interaction with integrins alpha(V)beta and alpha(V)beta. J. Biol. Chem. 2010, 285, 20806–20817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hayashi, S.; Wang, Z.; Bryan, J.; Kobayashi, C.; Faccio, R.; Sandell, L.J. The type II collagen N-propeptide, PIIBNP, inhibits cell survival and bone resorption of osteoclasts via integrin-mediated signaling. Bone 2011, 49, 644–652. [Google Scholar] [CrossRef] [Green Version]
- Tarpey, P.S.; Behjati, S.; Cooke, S.L.; Van Loo, P.; Wedge, D.C.; Pillay, N.; Marshall, J.; O’Meara, S.; Davies, H.; Nik-Zainal, S.; et al. Frequent mutation of the major cartilage collagen gene COL2A1 in chondrosarcoma. Nat. Genet. 2013, 45, 923–926. [Google Scholar] [CrossRef]
- Prokop, I.; Kononczuk, J.; Surazynski, A.; Palka, J. Cross-talk between integrin receptor and insulin-like growth factor receptor in regulation of collagen biosynthesis in cultured fibroblasts. Adv. Med. Sci 2013, 58, 292–297. [Google Scholar] [CrossRef] [Green Version]
- Xu, S.; Xu, H.; Wang, W.; Li, S.; Li, H.; Li, T.; Zhang, W.; Yu, X.; Liu, L. The role of collagen in cancer: From bench to bedside. J. Transl. Med. 2019, 17, 309. [Google Scholar] [CrossRef] [Green Version]
- Chao, C.C.; Lee, W.F.; Yang, W.H.; Lin, C.Y.; Han, C.K.; Huang, Y.L.; Fong, Y.C.; Wu, M.H.; Lee, I.T.; Tsai, Y.H.; et al. IGFBP-3 stimulates human osteosarcoma cell migration by upregulating VCAM-1 expression. Life Sci. 2021, 265, 118758. [Google Scholar] [CrossRef] [PubMed]
- Perks, C.M.; Holly, J.M. Epigenetic regulation of insulin-like growth factor binding protein-3 (IGFBP-3) in cancer. J. Cell Commun. Signal. 2015, 9, 159–166. [Google Scholar] [CrossRef] [Green Version]
- Price, D.; Dorandish, S.; Williams, A.; Iwaniec, B.; Stephens, A.; Marshall, K.; Guthrie, J.; Heyl, D.; Evans, H.G. Humanin Blocks the Aggregation of Amyloid-beta Induced by Acetylcholinesterase, an Effect Abolished in the Presence of IGFBP-3. Biochemistry 2020, 59, 1981–2002. [Google Scholar] [CrossRef]
- Wu, C.M.; Li, T.M.; Hsu, S.F.; Su, Y.C.; Kao, S.T.; Fong, Y.C.; Tang, C.H. IGF-I enhances alpha5beta1 integrin expression and cell motility in human chondrosarcoma cells. J. Cell Physiol. 2011, 226, 3270–3277. [Google Scholar] [CrossRef]
- Benini, S.; Zuntini, M.; Manara, M.C.; Cohen, P.; Nicoletti, G.; Nanni, P.; Oh, Y.; Picci, P.; Scotlandi, K. Insulin-like growth factor binding protein 3 as an anticancer molecule in Ewing’s sarcoma. Int. J. Cancer 2006, 119, 1039–1046. [Google Scholar] [CrossRef] [PubMed]
- Lin, F.; Shen, Z.; Xu, X.; Hu, B.B.; Meerani, S.; Tang, L.N.; Zheng, S.E.; Sun, Y.J.; Min, D.L.; Yao, Y. Evaluation of the expression and role of IGF pathway biomarkers in human sarcomas. Int. J. Immunopathol. Pharmacol. 2013, 26, 169–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molina, E.R.; Chim, L.K.; Salazar, M.C.; Koons, G.L.; Menegaz, B.A.; Ruiz-Velasco, A.; Lamhamedi-Cherradi, S.E.; Vetter, A.M.; Satish, T.; Cuglievan, B.; et al. 3D Tissue-Engineered Tumor Model for Ewing’s Sarcoma That Incorporates Bone-like ECM and Mineralization. ACS Biomater. Sci. Eng. 2020, 6, 539–552. [Google Scholar] [CrossRef] [PubMed]
- Conover, C.A.; Khosla, S. Role of extracellular matrix in insulin-like growth factor (IGF) binding protein-2 regulation of IGF-II action in normal human osteoblasts. Growth Horm. IGF Res. 2003, 13, 328–335. [Google Scholar] [CrossRef]
- Arai, T.; Busby, W., Jr.; Clemmons, D.R. Binding of insulin-like growth factor (IGF) I or II to IGF-binding protein-2 enables it to bind to heparin and extracellular matrix. Endocrinology 1996, 137, 4571–4575. [Google Scholar] [CrossRef] [Green Version]
- Heng, L.; Jia, Z.; Bai, J.; Zhang, K.; Zhu, Y.; Ma, J.; Zhang, J.; Duan, H. Molecular characterization of metastatic osteosarcoma: Differentially expressed genes, transcription factors and microRNAs. Mol. Med. Rep. 2017, 15, 2829–2836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sato, S.; Tang, Y.J.; Wei, Q.; Hirata, M.; Weng, A.; Han, I.; Okawa, A.; Takeda, S.; Whetstone, H.; Nadesan, P.; et al. Mesenchymal Tumors Can Derive from Ng2/Cspg4-Expressing Pericytes with beta-Catenin Modulating the Neoplastic Phenotype. Cell Rep. 2016, 16, 917–927. [Google Scholar] [CrossRef] [Green Version]
- Hsu, S.C.; Nadesan, P.; Puviindran, V.; Stallcup, W.B.; Kirsch, D.G.; Alman, B.A. Effects of chondroitin sulfate proteoglycan 4 (NG2/CSPG4) on soft-tissue sarcoma growth depend on tumor developmental stage. J. Biol. Chem. 2018, 293, 2466–2475. [Google Scholar] [CrossRef] [Green Version]
- Ortiz, M.V.; Roberts, S.S.; Glade Bender, J.; Shukla, N.; Wexler, L.H. Immunotherapeutic Targeting of GPC3 in Pediatric Solid Embryonal Tumors. Front. Oncol. 2019, 9, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, H.H.; Shi, W.; Xiang, Y.Y.; Filmus, J. The loss of glypican-3 induces alterations in Wnt signaling. J. Biol. Chem. 2005, 280, 2116–2125. [Google Scholar] [CrossRef] [Green Version]
- Saikali, Z.; Sinnett, D. Expression of glypican 3 (GPC3) in embryonal tumors. Int. J. Cancer 2000, 89, 418–422. [Google Scholar] [CrossRef]
- Mytilinaiou, M.; Nikitovic, D.; Berdiaki, A.; Papoutsidakis, A.; Papachristou, D.J.; Tsatsakis, A.; Tzanakakis, G.N. IGF-I regulates HT1080 fibrosarcoma cell migration through a syndecan-2/Erk/ezrin signaling axis. Exp. Cell Res. 2017, 361, 9–18. [Google Scholar] [CrossRef]
- Nikitovic, D.; Papoutsidakis, A.; Karamanos, N.K.; Tzanakakis, G.N. Lumican affects tumor cell functions, tumor-ECM interactions, angiogenesis and inflammatory response. Matrix Biol. 2014, 35, 206–214. [Google Scholar] [CrossRef]
- Neill, T.; Schaefer, L.; Iozzo, R.V. Decorin as a multivalent therapeutic agent against cancer. Adv. Drug Deliv. Rev. 2016, 97, 174–185. [Google Scholar] [CrossRef] [Green Version]
- Iozzo, R.V.; Buraschi, S.; Genua, M.; Xu, S.Q.; Solomides, C.C.; Peiper, S.C.; Gomella, L.G.; Owens, R.C.; Morrione, A. Decorin antagonizes IGF receptor I (IGF-IR) function by interfering with IGF-IR activity and attenuating downstream signaling. J. Biol. Chem. 2011, 286, 34712–34721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Belfiore, A.; Frasca, F.; Pandini, G.; Sciacca, L.; Vigneri, R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009, 30, 586–623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, D.; Lu, Y.; Zhu, L.; Liang, T.; Wang, Z.; Ren, J.; He, R.; Wang, K. Anti-osteosarcoma property of decorin-modified titanium surface: A novel strategy to inhibit oncogenic potential of osteosarcoma cells. Biomed. Pharmacother. 2020, 125, 110034. [Google Scholar] [CrossRef]
- Lu, B.J.; Wang, Y.Q.; Wei, X.J.; Rong, L.Q.; Wei, D.; Yan, C.M.; Wang, D.J.; Sun, J.Y. Expression of WNT-5a and ROR2 correlates with disease severity in osteosarcoma. Mol. Med. Rep. 2012, 5, 1033–1036. [Google Scholar] [CrossRef]
- Papoutsidakis, A.; Giatagana, E.M.; Berdiaki, A.; Spyridaki, I.; Spandidos, D.A.; Tsatsakis, A.; Tzanakakis, G.N.; Nikitovic, D. Lumican mediates HTB94 chondrosarcoma cell growth via an IGFIR/Erk1/2 axis. Int J. Oncol 2020, 57, 791–803. [Google Scholar] [CrossRef]
- Conover, C.A.; Oxvig, C. PAPP-A and cancer. J. Mol. Endocrinol. 2018, 61, T1–T10. [Google Scholar] [CrossRef] [Green Version]
- Heitzeneder, S.; Sotillo, E.; Shern, J.F.; Sindiri, S.; Xu, P.; Jones, R.; Pollak, M.; Noer, P.R.; Lorette, J.; Fazli, L.; et al. Pregnancy-Associated Plasma Protein-A (PAPP-A) in Ewing Sarcoma: Role in Tumor Growth and Immune Evasion. J. Natl. Cancer Inst. 2019, 111, 970–982. [Google Scholar] [CrossRef] [PubMed]
- Kirschner, A.; Thiede, M.; Grunewald, T.G.; Alba Rubio, R.; Richter, G.H.; Kirchner, T.; Busch, D.H.; Burdach, S.; Thiel, U. Pappalysin-1 T cell receptor transgenic allo-restricted T cells kill Ewing sarcoma in vitro and in vivo. Oncoimmunology 2017, 6, e1273301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leung, D.W.; Cachianes, G.; Kuang, W.J.; Goeddel, D.V.; Ferrara, N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 1989, 246, 1306–1309. [Google Scholar] [CrossRef]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prelaj, A.; Pircher, C.C.; Massa, G.; Martelli, V.; Corrao, G.; Lo Russo, G.; Proto, C.; Ferrara, R.; Galli, G.; De Toma, A.; et al. Beyond First-Line Immunotherapy: Potential Therapeutic Strategies Based on Different Pattern Progressions: Oligo and Systemic Progression. Cancers 2021, 13, 1300. [Google Scholar] [CrossRef] [PubMed]
- Neagu, M.; Constantin, C.; Caruntu, C.; Dumitru, C.; Surcel, M.; Zurac, S. Inflammation: A key process in skin tumorigenesis. Oncol. Lett. 2019, 17, 4068–4084. [Google Scholar] [CrossRef] [Green Version]
- Neagu, M.; Constantin, C.; Popescu, I.D.; Zipeto, D.; Tzanakakis, G.; Nikitovic, D.; Fenga, C.; Stratakis, C.A.; Spandidos, D.A.; Tsatsakis, A.M. Inflammation and Metabolism in Cancer Cell-Mitochondria Key Player. Front. Oncol. 2019, 9, 348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Solomon, I.; Voiculescu, V.M.; Caruntu, C.; Lupu, M.; Popa, A.; Ilie, M.A.; Albulescu, R.; Caruntu, A.; Tanase, C.; Constantin, C.; et al. Neuroendocrine Factors and Head and Neck Squamous Cell Carcinoma: An Affair to Remember. Dis. Markers 2018, 2018, 9787831. [Google Scholar] [CrossRef] [Green Version]
- Hristova, M.G. Neuroendocrine and immune disequilibrium as a probable link between metabolic syndrome and carcinogenesis. Med. Hypotheses 2018, 118, 1–5. [Google Scholar] [CrossRef] [PubMed]
- Smith, T.J.; Janssen, J. Insulin-like Growth Factor-I Receptor and Thyroid-Associated Ophthalmopathy. Endocr. Rev. 2019, 40, 236–267. [Google Scholar] [CrossRef] [PubMed]
- Thanindratarn, P.; Dean, D.C.; Nelson, S.D.; Hornicek, F.J.; Duan, Z. Chimeric antigen receptor T (CAR-T) cell immunotherapy for sarcomas: From mechanisms to potential clinical applications. Cancer Treat. Rev. 2020, 82, 101934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, X.; Park, H.; Greene, J.; Pao, J.; Mulvey, E.; Zhou, S.X.; Albert, C.M.; Moy, F.; Sachdev, D.; Yee, D.; et al. IGF1R- and ROR1-Specific CAR T Cells as a Potential Therapy for High Risk Sarcomas. PLoS ONE 2015, 10, e0133152. [Google Scholar] [CrossRef] [PubMed]
- Heymann, M.F.; Lezot, F.; Heymann, D. The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma. Cell Immunol. 2019, 343, 103711. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [Green Version]
- Long, A.H.; Highfill, S.L.; Cui, Y.; Smith, J.P.; Walker, A.J.; Ramakrishna, S.; El-Etriby, R.; Galli, S.; Tsokos, M.G.; Orentas, R.J.; et al. Reduction of MDSCs with All-trans Retinoic Acid Improves CAR Therapy Efficacy for Sarcomas. Cancer Immunol. Res. 2016, 4, 869–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bach, L.A.; Fu, P.; Yang, Z. Insulin-like growth factor-binding protein-6 and cancer. Clin. Sci. 2013, 124, 215–229. [Google Scholar] [CrossRef] [Green Version]
- Bach, L.A. IGF-binding proteins. J. Mol. Endocrinol. 2018, 61, T11–T28. [Google Scholar] [CrossRef] [Green Version]
- Jamitzky, S.; Krueger, A.C.; Janneschuetz, S.; Piepke, S.; Kailayangiri, S.; Spurny, C.; Rossig, C.; Altvater, B. Insulin-like growth factor-1 receptor (IGF-1R) inhibition promotes expansion of human NK cells which maintain their potent antitumor activity against Ewing sarcoma cells. Pediatr. Blood Cancer 2015, 62, 1979–1985. [Google Scholar] [CrossRef]
- Folkman, J. Anti-angiogenesis: New concept for therapy of solid tumors. Ann. Surg. 1972, 175, 409–416. [Google Scholar] [CrossRef] [PubMed]
- Culy, C. Bevacizumab: Antiangiogenic cancer therapy. Drugs Today 2005, 41, 23–36. [Google Scholar] [CrossRef] [PubMed]
- Maishi, N.; Hida, K. Tumor endothelial cells accelerate tumor metastasis. Cancer Sci. 2017, 108, 1921–1926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gacche, R.N.; Meshram, R.J. Angiogenic factors as potential drug target: Efficacy and limitations of anti-angiogenic therapy. Biochim. Biophys. Acta 2014, 1846, 161–179. [Google Scholar] [CrossRef] [PubMed]
- Paez-Ribes, M.; Allen, E.; Hudock, J.; Takeda, T.; Okuyama, H.; Vinals, F.; Inoue, M.; Bergers, G.; Hanahan, D.; Casanovas, O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009, 15, 220–231. [Google Scholar] [CrossRef] [Green Version]
- Ohga, N.; Ishikawa, S.; Maishi, N.; Akiyama, K.; Hida, Y.; Kawamoto, T.; Sadamoto, Y.; Osawa, T.; Yamamoto, K.; Kondoh, M.; et al. Heterogeneity of tumor endothelial cells: Comparison between tumor endothelial cells isolated from high- and low-metastatic tumors. Am. J. Pathol. 2012, 180, 1294–1307. [Google Scholar] [CrossRef] [PubMed]
- Maishi, N.; Annan, D.A.; Kikuchi, H.; Hida, Y.; Hida, K. Tumor Endothelial Heterogeneity in Cancer Progression. Cancers 2019, 11, 1511. [Google Scholar] [CrossRef] [Green Version]
- Kikuchi, H.; Maishi, N.; Annan, D.A.; Alam, M.T.; Dawood, R.I.H.; Sato, M.; Morimoto, M.; Takeda, R.; Ishizuka, K.; Matsumoto, R.; et al. Chemotherapy-Induced IL8 Upregulates MDR1/ABCB1 in Tumor Blood Vessels and Results in Unfavorable Outcome. Cancer Res. 2020, 80, 2996–3008. [Google Scholar]
- Annan, D.A.; Kikuchi, H.; Maishi, N.; Hida, Y.; Hida, K. Tumor Endothelial Cell-A Biological Tool for Translational Cancer Research. Int. J. Mol. Sci. 2020, 21, 3238. [Google Scholar] [CrossRef]
- Bergers, G.; Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 2008, 8, 592–603. [Google Scholar] [CrossRef] [Green Version]
- Lin, S.; Zhang, Q.; Shao, X.; Zhang, T.; Xue, C.; Shi, S.; Zhao, D.; Lin, Y. IGF-1 promotes angiogenesis in endothelial cells/adipose-derived stem cells co-culture system with activation of PI3K/Akt signal pathway. Cell Prolif. 2017, 50, e12390. [Google Scholar] [CrossRef] [Green Version]
- Guerreiro, A.S.; Boller, D.; Doepfner, K.T.; Arcaro, A. IGF-IR: Potential role in antitumor agents. Drug News Perspect. 2006, 19, 261–272. [Google Scholar] [CrossRef] [PubMed]
- Kurmasheva, R.T.; Dudkin, L.; Billups, C.; Debelenko, L.V.; Morton, C.L.; Houghton, P.J. The insulin-like growth factor-1 receptor-targeting antibody, CP-751, 871, suppresses tumor-derived VEGF and synergizes with rapamycin in models of childhood sarcoma. Cancer Res. 2009, 69, 7662–7671. [Google Scholar] [CrossRef] [Green Version]
- Marchand, M.; Monnot, C.; Muller, L.; Germain, S. Extracellular matrix scaffolding in angiogenesis and capillary homeostasis. Semin. Cell Dev. Biol. 2019, 89, 147–156. [Google Scholar] [CrossRef]
- Vempati, P.; Popel, A.S.; Mac Gabhann, F. Extracellular regulation of VEGF: Isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev. 2014, 25, 1–19. [Google Scholar] [CrossRef] [Green Version]
- Bartolini, B.; Carava, E.; Caon, I.; Parnigoni, A.; Moretto, P.; Passi, A.; Vigetti, D.; Viola, M.; Karousou, E. Heparan Sulfate in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1245, 147–161. [Google Scholar] [PubMed]
- Goveia, J.; Rohlenova, K.; Taverna, F.; Treps, L.; Conradi, L.C.; Pircher, A.; Geldhof, V.; de Rooij, L.; Kalucka, J.; Sokol, L.; et al. An Integrated Gene Expression Landscape Profiling Approach to Identify Lung Tumor Endothelial Cell Heterogeneity and Angiogenic Candidates. Cancer Cell 2020, 37, 421. [Google Scholar] [CrossRef]
- Maishi, N.; Ohba, Y.; Akiyama, K.; Ohga, N.; Hamada, J.; Nagao-Kitamoto, H.; Alam, M.T.; Yamamoto, K.; Kawamoto, T.; Inoue, N.; et al. Tumour endothelial cells in high metastatic tumours promote metastasis via epigenetic dysregulation of biglycan. Sci. Rep. 2016, 6, 28039. [Google Scholar] [CrossRef] [PubMed]
- Morimoto, H.; Hida, Y.; Maishi, N.; Nishihara, H.; Hatanaka, Y.; Li, C.; Matsuno, Y.; Nakamura, T.; Hirano, S.; Hida, K. Biglycan, tumor endothelial cell secreting proteoglycan, as possible biomarker for lung cancer. Thorac. Cancer 2021. [Google Scholar] [CrossRef]
- Lee, H.; Lim, J.; Oh, J.H.; Cho, S.; Chung, J.H. IGF-1 Upregulates Biglycan and Decorin by Increasing Translation and Reducing ADAMTS5 Expression. Int. J. Mol. Sci. 2021, 22, 1403. [Google Scholar] [CrossRef]
- Andrlova, H.; Mastroianni, J.; Madl, J.; Kern, J.S.; Melchinger, W.; Dierbach, H.; Wernet, F.; Follo, M.; Technau-Hafsi, K.; Has, C.; et al. Biglycan expression in the melanoma microenvironment promotes invasiveness via increased tissue stiffness inducing integrin-beta1 expression. Oncotarget 2017, 8, 42901–42916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haluska, P.; Worden, F.; Olmos, D.; Yin, D.; Schteingart, D.; Batzel, G.N.; Paccagnella, M.L.; de Bono, J.S.; Gualberto, A.; Hammer, G.D. Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother. Pharmacol. 2010, 65, 765–773. [Google Scholar] [CrossRef] [Green Version]
- Langer, C.J.; Novello, S.; Park, K.; Krzakowski, M.; Karp, D.D.; Mok, T.; Benner, R.J.; Scranton, J.R.; Olszanski, A.J.; Jassem, J. Randomized, phase III trial of first-line figitumumab in combination with paclitaxel and carboplatin versus paclitaxel and carboplatin alone in patients with advanced non-small-cell lung cancer. J. Clin. Oncol. 2014, 32, 2059–2066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, H.; Zhang, T.; Shen, H.; Cao, H.; Du, J. The adverse events profile of anti-IGF-1R monoclonal antibodies in cancer therapy. Br. J. Clin. Pharmacol. 2014, 77, 917–928. [Google Scholar] [CrossRef] [PubMed]
- Simpson, A.; Petnga, W.; Macaulay, V.M.; Weyer-Czernilofsky, U.; Bogenrieder, T. Insulin-Like Growth Factor (IGF) Pathway Targeting in Cancer: Role of the IGF Axis and Opportunities for Future Combination Studies. Target. Oncol. 2017, 12, 571–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Maldegem, A.M.; Bovee, J.V.; Peterse, E.F.; Hogendoorn, P.C.; Gelderblom, H. Ewing sarcoma: The clinical relevance of the insulin-like growth factor 1 and the poly-ADP-ribose-polymerase pathway. Eur. J. Cancer 2016, 53, 171–180. [Google Scholar] [CrossRef]
- Malempati, S.; Weigel, B.; Ingle, A.M.; Ahern, C.H.; Carroll, J.M.; Roberts, C.T.; Reid, J.M.; Schmechel, S.; Voss, S.D.; Cho, S.Y.; et al. Phase I/II trial and pharmacokinetic study of cixutumumab in pediatric patients with refractory solid tumors and Ewing sarcoma: A report from the Children’s Oncology Group. J. Clin. Oncol. 2012, 30, 256–262. [Google Scholar] [CrossRef]
- Weigel, B.; Malempati, S.; Reid, J.M.; Voss, S.D.; Cho, S.Y.; Chen, H.X.; Krailo, M.; Villaluna, D.; Adamson, P.C.; Blaney, S.M. Phase 2 trial of cixutumumab in children, adolescents, and young adults with refractory solid tumors: A report from the Children’s Oncology Group. Pediatr. Blood Cancer 2014, 61, 452–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olmos, D.; Postel-Vinay, S.; Molife, L.R.; Okuno, S.H.; Schuetze, S.M.; Paccagnella, M.L.; Batzel, G.N.; Yin, D.; Pritchard-Jones, K.; Judson, I.; et al. Safety, pharmacokinetics, and preliminary activity of the anti-IGF-1R antibody figitumumab (CP-751, 871) in patients with sarcoma and Ewing’s sarcoma: A phase 1 expansion cohort study. Lancet Oncol. 2010, 11, 129–135. [Google Scholar] [CrossRef] [Green Version]
- Juergens, H.; Daw, N.C.; Geoerger, B.; Ferrari, S.; Villarroel, M.; Aerts, I.; Whelan, J.; Dirksen, U.; Hixon, M.L.; Yin, D.; et al. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. J. Clin. Oncol. 2011, 29, 4534–4540. [Google Scholar] [CrossRef] [Green Version]
- Pappo, A.S.; Patel, S.R.; Crowley, J.; Reinke, D.K.; Kuenkele, K.P.; Chawla, S.P.; Toner, G.C.; Maki, R.G.; Meyers, P.A.; Chugh, R.; et al. R1507, a monoclonal antibody to the insulin-like growth factor 1 receptor, in patients with recurrent or refractory Ewing sarcoma family of tumors: Results of a phase II Sarcoma Alliance for Research through Collaboration study. J. Clin. Oncol. 2011, 29, 4541–4547. [Google Scholar] [CrossRef]
- Tap, W.D.; Demetri, G.; Barnette, P.; Desai, J.; Kavan, P.; Tozer, R.; Benedetto, P.W.; Friberg, G.; Deng, H.; McCaffery, I.; et al. Phase II study of ganitumab, a fully human anti-type-1 insulin-like growth factor receptor antibody, in patients with metastatic Ewing family tumors or desmoplastic small round cell tumors. J. Clin. Oncol. 2012, 30, 1849–1856. [Google Scholar] [CrossRef] [PubMed]
- Anderson, P.M.; Bielack, S.S.; Gorlick, R.G.; Skubitz, K.; Daw, N.C.; Herzog, C.E.; Monge, O.R.; Lassaletta, A.; Boldrini, E.; Papai, Z.; et al. A phase II study of clinical activity of SCH 717454 (robatumumab) in patients with relapsed osteosarcoma and Ewing sarcoma. Pediatr. Blood Cancer 2016, 63, 1761–1770. [Google Scholar] [CrossRef] [PubMed]
- Pappo, A.S.; Vassal, G.; Crowley, J.J.; Bolejack, V.; Hogendoorn, P.C.; Chugh, R.; Ladanyi, M.; Grippo, J.F.; Dall, G.; Staddon, A.P.; et al. A phase 2 trial of R1507, a monoclonal antibody to the insulin-like growth factor-1 receptor (IGF-1R), in patients with recurrent or refractory rhabdomyosarcoma, osteosarcoma, synovial sarcoma, and other soft tissue sarcomas: Results of a Sarcoma Alliance for Research Through Collaboration study. Cancer 2014, 120, 2448–2456. [Google Scholar]
- Scotlandi, K.; Manara, M.C.; Serra, M.; Marino, M.T.; Ventura, S.; Garofalo, C.; Alberghini, M.; Magagnoli, G.; Ferrari, S.; Lopez-Guerrero, J.A.; et al. Expression of insulin-like growth factor system components in Ewing’s sarcoma and their association with survival. Eur. J. Cancer 2011, 47, 1258–1266. [Google Scholar] [CrossRef]
- Zhao, X.; Li, J.; Yu, D. MicroRNA-939-5p directly targets IGF-1R to inhibit the aggressive phenotypes of osteosarcoma through deactivating the PI3K/Akt pathway. Int. J. Mol. Med. 2019, 44, 1833–1843. [Google Scholar] [CrossRef] [Green Version]
- Tian, Z.; Niu, X.; Yao, W. Receptor Tyrosine Kinases in Osteosarcoma Treatment: Which Is the Key Target? Front. Oncol. 2020, 10, 1642. [Google Scholar] [CrossRef]
- Ameline, B.; Kovac, M.; Nathrath, M.; Barenboim, M.; Witt, O.; Krieg, A.H.; Baumhoer, D. Overactivation of the IGF signalling pathway in osteosarcoma: A potential therapeutic target? J. Pathol. Clin. Res. 2021, 7, 165–172. [Google Scholar] [CrossRef]
- Ghedira, D.; Voissiere, A.; Peyrode, C.; Kraiem, J.; Gerard, Y.; Maubert, E.; Vivier, M.; Miot-Noirault, E.; Chezal, J.M.; Farhat, F.; et al. Structure-activity relationship study of hypoxia-activated prodrugs for proteoglycan-targeted chemotherapy in chondrosarcoma. Eur. J. Med. Chem. 2018, 158, 51–67. [Google Scholar] [CrossRef]
- Vlahovic, G.; Meadows, K.L.; Hatch, A.J.; Jia, J.; Nixon, A.B.; Uronis, H.E.; Morse, M.A.; Selim, M.A.; Crawford, J.; Riedel, R.F.; et al. A Phase I Trial of the IGF-1R Antibody Ganitumab (AMG 479) in Combination with Everolimus (RAD001) and Panitumumab in Patients with Advanced Cancer. Oncologist 2018, 23, 782–790. [Google Scholar] [CrossRef] [Green Version]
- Yap, T.A.; Olmos, D.; Molife, L.R.; de Bono, J.S. Targeting the insulin-like growth factor signaling pathway: Figitumumab and other novel anticancer strategies. Expert Opin. Investig. Drugs 2011, 20, 1293–1304. [Google Scholar] [CrossRef]
- Radic-Sarikas, B.; Tsafou, K.P.; Emdal, K.B.; Papamarkou, T.; Huber, K.V.; Mutz, C.; Toretsky, J.A.; Bennett, K.L.; Olsen, J.V.; Brunak, S.; et al. Combinatorial Drug Screening Identifies Ewing Sarcoma-specific Sensitivities. Mol. Cancer Ther. 2017, 16, 88–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levy, J.M.M.; Towers, C.G.; Thorburn, A. Targeting autophagy in cancer. Nat. Rev. Cancer 2017, 17, 528–542. [Google Scholar] [CrossRef]
- Bustos, S.O.; Antunes, F.; Rangel, M.C.; Chammas, R. Emerging Autophagy Functions Shape the Tumor Microenvironment and Play a Role in Cancer Progression—Implications for Cancer Therapy. Front. Oncol. 2020, 10, 606436. [Google Scholar] [CrossRef]
Regulator | ECM Target | Effect | Cells | Ref. |
---|---|---|---|---|
IGF-1 and IGF-2 | Aggrecan | Maintaining high expression | Chondrocytes/Chondrosarcomas (In vitro; cell lines) | [125] |
IGF-1 | PGs and p21 | Increased expression | Chondrosarcomas (In vitro; cell lines) | [128] |
IGF-1 | Xylosyltransferase I and alkaline phosphatase | Increased expression | Osteosarcomas (In vitro; cell lines) | [135] |
IGF-1 | Collagen I | Increased expression | Osteosarcomas (In vitro; cell lines) | [141] |
IGFBP-4 | Collagen I | Decreased expression | Osteosarcomas (In vitro; cell lines | [141] |
IGF-I | Cysteine protease | Decreased activity | Osteosarcomas (In vitro; cell lines) | [142] |
IGF-1 and/or IGFBP-5 | Collagen I | Enhance Estrogen-mediated PTH-dependent expression | Osteosarcomas (In vitro; cell lines) | [143] |
IGF-1 | Collagen II | Increased expression | Chondrocytes (In vitro; rat primary cell cultures) | [147] |
IGFBP-1 | VCAM-1 | Increased expression | Osteosarcoma (In vitro; primary cell cultures; tissue biopsies) | [153] |
IGF-1 | α5β1-integrin | Increased expression | Chondrosarcoma (In vitro; primary cell cultures) | [156] |
IGFBP-3 | MMP-9 | Decreased activity | Ewing sarcoma (In vitro; primary cell and cell line cultures) | [157] |
IGF-IR | MMP-2 and MMP-9 | Increased expression | Osteosarcomas (In vitro; cell lines; tissue biopsies) | [158] |
Regulator | Activity | Effect | Tumor | Ref. |
---|---|---|---|---|
Heparin/HSPGs | Regulate IGF-1/IGF-2- binding to IGFBP-2 | Attenuation of IGF signaling/Inhibition of apoptosis | Osteosarcoma cells/Osteoblasts | [160,161] |
NG2/CSPG4 | Decreases IGFBP3 expression and facilitates IGF-signaling | Decreased tumor size | Murine and human sarcoma models | [164] |
Syndecan 2 | Co-receptor for IGF-1 and linker to ezrin | Facilitates IGF-I-dependent fibrosarcoma cell migration | Fibrosarcoma | [168] |
PAPP-A | Cleavage of inhibitory IGF-binding proteins | Increased free IGF-I, cell growth and downstream IGF signaling | Ewing sarcoma | [179] |
Tumor Type | Drug | Phase | Clinical Results | Safety Results | Ref. |
---|---|---|---|---|---|
EWS and other solid tumors | Cixutumumab | I/II (only pediatric patients) | Limited activity in EWS | Well tolerated | [221] |
EWS and desmoplastic small round cell tumors | Ganitumab | II | Limited activity in EWS | Generally well tolerated | [226] |
EWS and other sarcomas | figitumumab | I | EWS objective responses: complete response, partial response, and stable disease in EWS, synovial sarcoma, and fibrosarcoma, lasting over 4months | Well tolerated; mild-to-moderate adverse effects | [223] |
EWS | figitumumab | I/II | Modest activity as single agent in advanced E | Good tolerability | [224] |
EWS | R1507 | II | Partial response | Well tolerated | [225] |
Bone and soft-tissue sarcomas | R1507 | II | Limited efficacy; overall response rate 2.5%) | Well tolerated | [228] |
Resectable osteosarcoma metastases (Group 1), unresectable osteosarcoma metastases (Group 2), and Ewing sarcoma metastases(Group 3) | Robatumumab | II | Limited efficacy in osteosarcoma patients, 6 of EWS patientshave remained healthy after receiving 25–115 doses of robatumumab with remissions of >4 years duration | Well tolerated | [227] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Tzanakakis, G.N.; Giatagana, E.-M.; Berdiaki, A.; Spyridaki, I.; Hida, K.; Neagu, M.; Tsatsakis, A.M.; Nikitovic, D. The Role of IGF/IGF-IR-Signaling and Extracellular Matrix Effectors in Bone Sarcoma Pathogenesis. Cancers 2021, 13, 2478. https://doi.org/10.3390/cancers13102478
Tzanakakis GN, Giatagana E-M, Berdiaki A, Spyridaki I, Hida K, Neagu M, Tsatsakis AM, Nikitovic D. The Role of IGF/IGF-IR-Signaling and Extracellular Matrix Effectors in Bone Sarcoma Pathogenesis. Cancers. 2021; 13(10):2478. https://doi.org/10.3390/cancers13102478
Chicago/Turabian StyleTzanakakis, George N., Eirini-Maria Giatagana, Aikaterini Berdiaki, Ioanna Spyridaki, Kyoko Hida, Monica Neagu, Aristidis M. Tsatsakis, and Dragana Nikitovic. 2021. "The Role of IGF/IGF-IR-Signaling and Extracellular Matrix Effectors in Bone Sarcoma Pathogenesis" Cancers 13, no. 10: 2478. https://doi.org/10.3390/cancers13102478
APA StyleTzanakakis, G. N., Giatagana, E. -M., Berdiaki, A., Spyridaki, I., Hida, K., Neagu, M., Tsatsakis, A. M., & Nikitovic, D. (2021). The Role of IGF/IGF-IR-Signaling and Extracellular Matrix Effectors in Bone Sarcoma Pathogenesis. Cancers, 13(10), 2478. https://doi.org/10.3390/cancers13102478