#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Novel Aspects of Genetics, Molecular Biology and Clinical Oncology of Sarcomas


Authors: K. Houfková;  J. Hatina
Authors‘ workplace: Ústav biologie, LF UK v Plzni
Published in: Klin Onkol 2020; 33(1): 66-78
Category: Short Communication
doi: https://doi.org/10.14735/amko202066

Overview

The Connective Tissue Oncology Group Annual Meeting 2018 (CTOS 2018) took place in Rome from 4 to 17 November 2018, and the 39th Plenary Meeting of the Scandinavian Sarcoma Group (SSGM 2019) was held in Bergen from 8 to 10 May 2019. These two large international conferences brought together an overwhelming majority of molecular and clinical specialists in the sarcoma field, especially those working on soft tissue sarcoma. Topics discussed on the conferences included, among others, sarcoma genetics, clinical and molecular subclassification, targeted therapy, clinical prognostication, and new experimental sarcoma models. A large ongoing international study on germinal sarcoma genetics was presented, the interim results of which revealed the extremely complex nature of genetic disposition to sarcoma, and, surprisingly, a rather prominent place among predisposing genes for those coding for structural telomere constituents. Fusion oncogenes dominate somatic sarcoma genetics, especially because of their origin and impact on sarcoma clinical behaviour, and are especially relevant for karyotypically simple paediatric sarcomas. A crucial issue in karyotypically complex sarcomas are the efforts being made to obtain a subclassification of sarcoma, other than those based on pathology, using either the clinical characteristics of sarcomas (uterine leiomyosarcoma vs. soft tissue leiomyosarcoma) or specific gene expression profiles (molecular subtypes in undifferentiated pleiomorphic sarcoma), which showed that molecular characterization can open the way for subtype specific therapies. Other examples of where this type of strategy can be applied include gastrointestinal stromal tumours, infantile fibrosarcoma, and inflammatory myofibroblastic tumours, where targeted therapy could be conceived based on the actionable mutations identified. Attempts in this direction have been made also for clear cell sarcoma and dedifferentiated liposarcoma, albeit the effectiveness of molecular-targeted treatments for these sarcomas is still poor, and progress in the treatment of osteosarcoma is still rather slow. Actually, the platelet-derived growth factor signalling system holds a prominent position in searches for targeted therapies, not only against rare sarcoma types, where are activated by mutations (some gastrointestinal stromal tumours, infantile hereditary myofibromatosis, and dermatofibrosarcoma protuberans), but also against other more usual sarcoma types, where the blocking anti-PDGFRα-antibody olaratumab has been successfully integrated into combinatorial chemotherapeutic regimens. In the field of clinical prognostication, remarkable progress in sarcoma nomograms was reported. Interesting results were also presented in the area of new experimental sarcoma models.

Participation on both scientifi c conferences and all the experimental work leading to the presented sarcoma models were supported by the Czech Science Foundation project No. 17-17636S.

The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study.

The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.

Submitted: 20. 9. 2019

Accepted: 6. 11. 2019

Keywords:

osteosarcoma – soft tissue sarcomas – chondrosarcoma – genetic predisposition – molecular subtypes – targeted therapy – prognostic nomograms – experimental sarcoma models


Sources

1. Fletcher CD, Unni KK, Mertens F (eds). WHO classification of tumours of soft tissue and bone. 4th ed. Lyon: IARC Press 2013.

2. Calvete O, Martinez P, Garcia-Pavia P et al. A mutation in the POT1 gene is responsible for cardiac angiosarcoma in TP53-negative Li–Fraumeni-like families. Nat Commun 2015; 6: 8383. doi: 10.1038/ncomms9383.

3. Koelsche C, Renner M, Hartmann W et al. TERT promoter hotspot mutations are recurrent in myxoid liposarcomas but rare in other soft tissue sarcoma entities. J Exp Clin Cancer Res 2014; 33 (1): 33. doi: 10.1186/1756-9966-33-33.

4. Delespaul L, Lesluyes T, Pérot G et al. Recurrent TRIO fusion in nontranslocation–related sarcomas. Clin Cancer Res 2017; 23 (3): 857–867. doi: 10.1158/1078-0432.CCR-16-0290.

5. Ballinger ML, Goode DL, Ray-Coquard I et al. Monogenic and polygenic determinants of sarcoma risk: an international genetic study. Lancet Oncol 2016; 17 (9): 1261–1271. doi: 10.1016/S1470-2045 (16) 30147-4.

6. Anderson ND, de Borja R, Young MD et al. Rearrangement bursts generate canonical gene fusions in bone and soft tissue tumors. Science 2018; 361 (6405): 8419. doi: 10.1126/science.aam8419.

7. Mertens F, Antonescu CR, Mitelman F. Gene fusions in soft tissue tumors: recurrent and overlapping pathogenetic themes. Genes Chromosomes Cancer 2016; 55 (4): 291–310. doi: 10.1002/gcc.22335.

8. Specht K, Hartmann W. Ewing-Sarkome und Ewing-artige Sarkome. Pathologe 2018; 39 (2): 154–163. doi: 10.1007/s00292-018-0421-2.

9. Cancer Genome Atlas Research Network. Comprehensive and integrated genomic characterization of adult soft tissue sarcomas. Cell 2017; 171 (4): 950–965. doi: 10.1016/j.cell.2017.10.014.

10. George S, Serrano C, Hensley ML et al. Soft tissue and uterine leiomyosarcoma. J Clin Oncol 2017; 36 (2): 144–150. doi: 10.1200/JCO.2017.75.9845.

11. Synnott NC, Bauer MR, Madden S et al. Mutant p53 as a therapeutic target for the treatment of triple-negative breast cancer: preclinical investigation with the anti-p53 drug, PK11007. Cancer Lett 2018; 414: 99–106. doi: 10.1016/j.canlet.2017.09.053.

12. Darvin P, Toor SM, Sasidharan Nair V et al. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med 2018; 50 (12): 165. doi: 10.1038/s12276-018-0191-1.

13. Broto JM, Hindi N, Redondo A et al. IMMUNOSARC: A collaborative Spanish (GEIS) and Italian (ISG) Sarcoma Groups phase I/II trial of sunitinib plus nivolumab in selected bone and soft tissue sarcoma subtypes – results of the phase I part. Ann Oncol 2019; 30 (Suppl 5): v683-v709. doi: 10.1093/annonc/mdz283.

14. Dufresne A, Brahmi M, Karanian M et al. Using biology to guide the treatment of sarcomas and aggressive connective-tissue tumours. Nat Rev Clin Oncol 2018; 15 (7): 443–458. doi: 10.1038/s41571-018-0012-4.

15. Knezevich SR, McFadden DE, Tao W et al. A novel ETV6-NTRK3 gene fusion in congenital fibrosarcoma. Nat Genet 1998; 18 (2): 184–187. doi: 10.1038/ng0298-184.

16. Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol 2018; 15 (12): 731–747. doi: 10.1038/s41571-018-0113-0.

17. DuBois SG, Laetsch TW, Federman N et al. The use of neoadjuvant larotrectinib in the management of children with locally advanced TRK fusion sarcomas. Cancer 2018; 124 (21): 4241–4247. doi: 10.1002/cncr.31701.

18. Davis JL, Lockwood CM, Albert CM et al. Infantile NTRK-associated mesenchymal tumors. Pediatr Dev Pathol 2018; 21 (1): 68–78. doi: 10.1177/1093526617712639.

19. Papadopoulos N, Lennartsson J. The PDGF/PDGFR pathway as a drug target. Mol Aspects Med 2018; 62: 75–88. doi: 10.1016/j.mam.2017.11.007.

20. Cheung YH, Gayden T, Campeau PM et al. A recurrent PDGFRB mutation causes familial infantile myofibromatosis. Am J Hum Genet 2013; 92 (6): 996–1000. doi: 10.1016/ j.ajhg.2013.04.026.

21. Martignetti JA, Tian L, Li D et al. Mutations in PDGFRB cause autosomal-dominant infantile myofibromatosis. Am J Hum Genet 2013; 92 (6): 1001–1007. doi: 10.1016/ j.ajhg.2013.04.024.

22. Mudry P, Slaby O, Neradil J et al. Case report: rapid and durable response to PDGFR targeted therapy in a child with refractory multiple infantile myofibromatosis and a heterozygous germline mutation of the PDGFRB gene. BMC Cancer 2017; 17 (1): 119. doi: 10.1186/s12885-017-3115-x.

23. Noujaim J, Thway K, Fisher C et al. Dermatofibrosarcoma protuberans: from translocation to targeted therapy. Cancer Biol Med 2015; 12 (4): 375–384. doi: 10.7497/ j.issn.2095-3941.2015.0067.

24. Lowery CD, Blosser W, Dowless M et al. Olaratumab exerts antitumor activity in preclinical models of pediatric bone and soft tissue tumors through inhibition of platelet-derived growth factor receptor α. Clin Cancer Res 2018; 24 (4): 847–857. doi: 10.1158/1078-0432.CCR-17-1258.

25. Antoniou G, Lee AT, Huang PH et al. Olaratumab in soft tissue sarcoma – current status and future perspectives. Eur J Cancer 2018; 92: 33–39. doi: 10.1016/ j.ejca.2017.12.026.

26. Meyers PA, Chou AJ. Muramyl tripeptide-phosphatidyl ethanolamine encapsulated in liposomes (L-MTP-PE) in the treatment of osteosarcoma. Adv Exp Med Biol 2014; 804: 307–321. doi: 10.1007/978-3-319-04843-7_17.

27. Baumhoer D. Die klonale Evolution des Osteosarkoms. Pathologe 2016; 37 (Suppl 2): 163–168. doi: 10.1007/s00292-016-0200-x.

28. Kovac M, Blattmann C, Ribi S et al. Exome sequencing of osteosarcoma reveals mutation signatures reminiscent of BRCA deficiency. Nat Commun 2015; 6: 8940. doi: 10.1038/ncomms9940.

29. Loh AH, Stewart E, Bradley CL. Combinatorial screening using orthotopic patient derived xenograft-expanded early phase cultures of osteosarcoma identify novel therapeutic drug combinations. Cancer Letters 2019; 442: 262–270. doi: 10.1016/j.canlet.2018.10.033.

30. Balachandran VP, Gonen M, Smith JJ et al. Nomograms in oncology: more than meets the eye. Lancet Oncol 2015; 16 (4): 173–180. doi: 10.1016/S1470-2045 (14) 71116-7.

31. Eilber FC, Kattan MW. Sarcoma nomogram: validation and a model to evaluate impact of therapy. J Am Coll Surg 2007; 205 (Suppl 4): S90–S95. doi: 10.1016/j.jamcollsurg.2007.06.335.

32. Tattersall HL, Callegaro D, Ford SJ et al. Staging, nomograms and other predictive tools in retroperitoneal soft tissue sarcoma. Chin Clin Oncol 2018; 7 (4): 36. doi: 10.21037/cco.2018.08.01.

33. Anaya DA, Lahat G, Wang X et al. Postoperative nomogram for survival of patients with retroperitoneal sarcoma treated with curative intent. Ann Oncol 2009; 21 (2): 397–402. doi: 10.1093/annonc/mdp298.

34. Kattan MW, Leung DH, Brennan MF. Postoperative nomogram for 12-year sarcoma-specific death. J Clin Oncol 2002; 20 (3): 791–796. doi: 10.1200/JCO.2002.20.3.791.

35. Canter RJ, Qin LX, Maki RG et al. A synovial sarcoma-specific preoperative nomogram supports a survival benefit to ifosfamide-based chemotherapy and improves risk stratification for patients. Clin Cancer Res 2008; 14 (24): 8191–8197. doi: 10.1158/1078-0432.CCR-08-0843.

36. Zivanovic O, Jacks LM, Iasonos A. et al. A nomogram to predict postresection 5-year overall survival for patients with uterine leiomyosarcoma. Cancer 2012; 118 (3): 660–669. doi: 10.1002/cncr.26333.

37. Dalal KM, Kattan MW, Antonescu CR et al. Subtype specific prognostic nomogram for patients with primary liposarcoma of the retroperitoneum, extremity, or trunk. Ann Surg 2006; 244 (3): 381–391. doi: 10.1097/01.sla.0000234795.98607.00.

38. Gronchi A, Miceli R, Shurell E et al. Outcome prediction in primary resected retroperitoneal soft tissue sarcoma: histology-specific overall survival and disease-free survival nomograms built on major sarcoma center data sets. J Clin Oncol 2013; 31 (13): 1649–1655. doi: 10.1200/JCO.2012.44.3747.

39. Callegaro D, Miceli R, Bonvalot S et al. Development and external validation of two nomograms to predict overall survival and occurrence of distant metastases in adults after surgical resection of localised soft-tissue sarcomas of the extremities: a retrospective analysis. Lancet Oncol 2016; 17 (5): 671–680. doi: 10.1016/S1470-2045 (16) 00010-3.

40. Raut CP, Miceli R, Strauss DC et al. External validation of a multi-institutional retroperitoneal sarcoma nomogram. Cancer 2016; 122 (9): 1417–1424. doi: 10.1002/cncr.29931.

41. Woll PJ, Reichardt P, Le Cesne A et al. Adjuvant chemotherapy with doxorubicin, ifosfamide, and lenograstim for resected soft-tissue sarcoma (EORTC 62931): a multicentre randomised controlled trial. Lancet Oncol 2012; 13 (10): 1045–1054. doi: 10.1016/S1470-2045 (12) 70346-7.

42. Hatina J, Hájková L, Peychl J et al. Establishment and characterization of clonal cell lines derived from a fibrosarcoma of the H2-K/v-jun transgenic mouse. Tumour Biol 2003; 24 (4): 176–184. doi: 10.1159/000074427.

43. Peychl J, Hatina J, Reischig J et al. Vztah motility a invazivity transformovaných buněk-model H2-K/V-JUN fibrosarkomových buněčných linií. Klin Onkol 2003; 16 (5): 223–226.

44. Mariani O, Brennetot C, Coindre JM et al. JUN oncogene amplification and overexpression block adipocytic differentiation in highly aggressive sarcomas. Cancer Cell 2007; 11 (4): 361–374. doi: 10.1016/j.ccr.2007. 02.007.

45. Endo M, Nishita M, Fujii M et al. Insight into the role of Wnt5a-induced signaling in normal and cancer cells. Int Rev Cell Mol Biol 2015; 314: 117–148. doi: 10.1016/ bs.ircmb.2014.10.003.

46. Qiang YW, Hu B, Chen Y et al. Bortezomib induces osteoblast differentiation via Wnt-independent activation of beta-catenin/TCF signaling. Blood 2009; 113 (18): 4319–4330. doi: 10.1182/blood-2008-08-174300.

47. Witz IP. Tumor–microenvironment interactions: dangerous liaisons. Adv Cancer Res 2008; 100: 203–229. doi: 10.1016/S0065-230X (08) 00007-9.

48. Brodin BA, Wennerberg K, Lidbrink E et al. Drug sensitivity testing on patient-derived sarcoma cells predicts patient response to treatment and identifies c-Sarc inhibitors as active drugs for translocation sarcomas. Br J Cancer 2019; 120 (4): 435–443. doi: 10.1038/s41416-018-0359-4.

Labels
Paediatric clinical oncology Surgery Clinical oncology
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#