Oops, you're using an old version of your browser so some of the features on this page may not be displaying properly.

MINIMAL Requirements: Google Chrome 24+Mozilla Firefox 20+Internet Explorer 11Opera 15–18Apple Safari 7SeaMonkey 2.15-2.23

PDGFRA in Gastrointestinal Stromal Tumours (GIST): ESMO Biomarker Factsheet

ESMO Factsheets on Biomarkers

PDGFRA

The PDGFRA gene encodes the platelet-derived growth factor receptor alpha, a member of the type III tyrosine kinase receptor family, which also includes the stem cell factor receptor, KIT[1]PDGFRA and KIT map to the same genomic loci, 4q12, and their protein structures are highly homologous, composed of several domains that each have a specific role in the process of tyrosine kinase activation [1, 2]. Several platelet-derived growth factor (PDGF) isoforms can bind and activate PDGFRA[2, 3]. Activated PDGFRA initialises similar downstream pathways as activated KIT, including the JAK–STAT3, phosphatidylinositide-3- kinase (PI3K)–AKT–mTOR, and RAS–MAPK pathways, important in regulating critical cellular functions such as proliferation and apoptosis[1, 2].

Gastrointestinal stromal tumours (GISTs) are the most common type of mesenchymal tumours of the digestive tract, the majority of which (82-87%) are characterised by the presence of mutually exclusive gain-of-function mutations in PDGFRA and KIT [1, 4-6]. After KITPDGFRA is the second most commonly mutated oncogene in GISTs[2]. Gain-of-function mutations lead to constitutive, ligand independent activation of PDGFRA and its downstream pathways, ultimately increasing cell proliferation and inhibiting apoptosis[1]. The discovery of activating mutations in the KIT and PDGFRA tyrosine kinase receptors and their role in the pathogenesis of GIST has revolutionised understanding of the biology and therapy of this disease[1].

PDGFRA mutations in GIST

PDGFRA mutation frequency is variable, ranging from <2% to 14% of GISTs, likely reflecting the low representation of PDGFRA-mutated GISTs in clinical trials due to their largely benign clinical behaviour[1, 7-14]PDGFRA-mutated GISTs are mostly of gastric origin and display epithelioid morphology or mixed epithelioid and spindle histology[10, 13-16]. Mutations (deletions, point mutations, duplications, insertions, and complex mutations) are found in exons coding for the functional domains of PDGFRA[17].

Primary PDGFRA mutations are found mainly in exons 12 and 18 and more rarely in exon 14[18]. PDGFRA mutations affecting exon 18 (the second kinase domain, corresponding to exon 17 of KIT) have been identified in approximately 6% of GISTs and are believed to aberrantly stabilise the kinase activation loop[2, 19]. The most frequent mutation results in an exon 18 D842V substitution, detected in up to 75% of all PDGFRA-mutated tumours[1, 7-11, 13-15]. Mutations affecting exon 12 (the juxtamembrane domain) are the second most common form of PDGFRA mutation, identified in approximately 1–2% of GISTs[2, 3, 20]. The PDGFRA juxtamembrane domain, is believed to mediate an autoinhibitory function and mutation in this inhibitory domain induces hyperactivation[2, 19]. Mutations in exon 14 are rare (<0.1% of GISTs), and frequently cluster at codon 659[2, 18, 21]. The function of exon 14 is less well studied but being close to exon 12 it may contribute to the autoinhibitory function of the juxtamembrane domain[2]. Secondary PDGFRA mutations have been identified in exon 18, generally accompanied by a primary mutation in same gene[17].

PDGFRA mutations as a diagnostic biomarker in GIST

KIT (CD117) expression is detected immunohistochemically in >95% of GISTS, making it a key diagnostic marker, together with anoctamin1 (DOG1)[18]. Approximately 5% of patients with GIST do not express KIT but may harbour KIT or PDGFRA mutations[1, 15, 18, 22].  Mutational analysis of GIST-mutated genes such as PDGFRA is paramount for selection of appropriate therapy for GIST (see below) but may also help confirm the diagnosis of suspect GIST that do not exhibit positive immunoreactivity for CD117/DOG1[23].

PDGFRA mutations as a prognostic biomarker in GIST

PDGFRA-mutated GISTs tend to follow a more indolent clinical course and are almost exclusively (90–93%) of prognostically favourable gastric origin[1, 10-12, 16]. A multivariate analysis of the Polish Clinical GIST registry that included 427 tumours found PDGFRA mutations in 12.9% of the cases and that mutations were associated with a 70% lower risk of 5-year relapse (hazard ratio = 0.298, 95% confidence interval 0.147– 0.602; P = 0.001) than patients with KIT deletions involving codons 557/558[10]. In the ConticaGIST study of 1,056 treatment-naïve patients with localised GIST who underwent surgery with curative intention, PDGFRA exon 18 mutation status correlated with a favourable disease outcome (median disease-free survival [DFS] not reached; 5-year DFS, 75%) in comparison with other mutations. There was no significant difference in DFS of PDGFRA D842V versus other exon 18 mutations; however, among gastric PDGFRA mutations, the vast majority that progressed (11 of 14) carried the exon 18 D842V substitution[11]

PDGFRA mutations as a predictive biomarker in GIST

Tyrosine kinase inhibitor therapy has improved survival in patients with GIST, increasing the life expectancy from about 1 year to 5 years in the advanced setting[24] . However, GISTs are composed of many different molecular subtypes, and response to tyrosine kinase therapy correlates with the underlying kinase genotype of the tumour[1, 4]. Routine genotyping has therefore become an integral part of management of GISTs undergoing tyrosine kinase inhibitor therapy[1]

The tyrosine kinase inhibitor, imatinib (Glivec®, Novartis Pharmaceuticals), was licensed >15 years ago for the treatment of adult patients with KIT (CD 117) positive unresectable and/or metastatic malignant GIST, and for the adjuvant treatment of adult patients who are at significant risk of relapse following resection of KIT (CD117)-positive GIST[25-27].  Imatinib has demonstrated pronounced clinical efficacy in GISTs, however, approximately 10–15% of patients show primary resistance with early progression within 3–6 months of initiating therapy and 40-50% of patients with advanced GIST subsequently develop secondary resistance with a mean time to progression of about 24 months[1, 28]. Imatinib can only bind to the inactive conformation of tyrosine kinase receptors and both primary and secondary resistance to imatinib can be partially explained by mutations that cause a conformational shift in the kinase domain that favours the activated state[1, 28, 29]. The most common mutation in PDGFRA, the exon 18 D842V substitution, results in a distortion of the kinase activation loop, thus strongly tilting the protein conformation in favour of the activated structure and is generally believed to lead to primary imatinib resistance[1, 4-6, 9, 14]. Patients with PDGFRA D842V‑mutant GIST have low response rates and short progression-free and overall survival during imatinib treatment[4, 30-33]. In vitro and clinical studies suggest that PDGFRA exon 18 mutations not involving D842 and exon 12 mutations are generally sensitive to imatinib treatment[5, 6, 14, 33]. In patients with the D842V mutation, adjuvant therapy with imatinib is not recommended and mutational testing is highly relevant to avoid overtreatment of GISTs with imatinib in this setting[1]

A second tyrosine kinase inhibitor, sunitinib (Sutent®, Pfizer), is licensed for use in imatinib-resistant GIST[34, 35], however preclinical studies of imatinib-resistant GIST cell lines evaluating sunitinib sensitivity PDGFRA D842V (activation loop) mutants remained resistant[1, 36]. Clinical data are as yet too limited to investigate the effects of PDGFRA mutations on efficacy outcomes following sunitinib treatment[1, 33, 37].

Regorafenib (Stivarga®, Bayer), is indicated as a third-line tyrosine kinase inhibitor for patients with unresectable or metastatic GIST who progressed on or are intolerant to prior treatment with imatinib and sunitinib[38, 39]. While the predictive power of mutational status for this agent is not yet known, it has demonstrated efficacy in one case of GIST with a PDGFRA D842A mutation[40].

An ongoing phase I study (NCT2508532) in advanced GIST is assessing the safety and clinical activity of a novel agent, BLU-285, a potent, highly selective oral inhibitor that targets PDGFRA D842V and KIT exon 17 mutants[41]. Adult patients with unresectable GISTs who had received two or more kinase inhibitors previously were given BLU-285 once daily[2]. The treatment was well tolerated and among 17 patients with tumours harbouring PDGFRA D842V mutations, seven had a PR while ten had stable disease, suggesting that precision-targeted therapy with BLU-285 is associated with significant activity against tumours previously resistant to other GIST therapies[2].

PDGFRA mutation analysis in GIST

Mutation analysis should be performed using an appropriate, validated, technique, and performed by specifically trained personnel and results should always specify the type of analysis performed, the region or mutations evaluated, and the sensitivity of the detection method used[18].  

The most widely used method for detecting PDGFRA mutations is amplification of the exons of interest by polymerase chain reaction (PCR) followed by direct sequencing (Sanger method) of amplification products. Due to limitations in the sensitivity of this technique it is essential that the methodology is appropriately optimised and controlled and performed only on samples containing ≥50% tumour cells[18]. Next-generation sequencing can provide greater sensitivity with several GIST-specific gene panels are now commercially available[18].

Patient selection

Given its diagnostic, prognostic and predictive value, mutational analysis of PDGFRA should be included in the diagnostic work-up of all GISTs as standard practice for optimal management[23].

References 

  1. Szucs Z, Thway K, Fisher C, et al. Molecular subtypes of gastrointestinal stromal tumors and their prognostic and therapeutic implications. Future Oncol 2017;13(1):93-107.
  2. Mei L, Smith SC, Faber AC, et al. Gastrointestinal Stromal Tumors: The GIST of Precision Medicine. Trends Cancer 2018;4(1):74-91.
  3. Joensuu H, DeMatteo RP. The management of gastrointestinal stromal tumors: a model for targeted and multidisciplinary therapy of malignancy. Annu Rev Med 2012;63:247-258.
  4. Corless CL, Barnett CM, Heinrich MC. Gastrointestinal stromal tumours: origin and molecular oncology. Nat Rev Cancer 2011;11(12):865-878.
  5. Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003;299(5607):708-710.
  6. Hirota S, Ohashi A, Nishida T, et al. Gain-of-function mutations of platelet-derived growth factor receptor alpha gene in gastrointestinal stromal tumors. Gastroenterology 2003;125(3):660-667.
  7. Corless CL, Ballman KV, Antonescu CR, et al. Pathologic and molecular features correlate with long-term outcome after adjuvant therapy of resected primary GI stromal tumor: the ACOSOG Z9001 trial. J Clin Oncol 2014;32(15):1563-1570.
  8. Debiec-Rychter M, Sciot R, Le Cesne A, et al. KIT mutations and dose selection for imatinib in patients with advanced gastrointestinal stromal tumours. Eur J Cancer 2006;42(8):1093-1103.
  9. Heinrich MC, Owzar K, Corless CL, et al. Correlation of kinase genotype and clinical outcome in the North American Intergroup Phase III Trial of imatinib mesylate for treatment of advanced gastrointestinal stromal tumor: CALGB 150105 Study by Cancer and Leukemia Group B and Southwest Oncology Group. J Clin Oncol 2008;26(33):5360-5367.
  10. Wozniak A, Rutkowski P, Piskorz A, et al. Prognostic value of KIT/PDGFRA mutations in gastrointestinal stromal tumours (GIST): Polish Clinical GIST Registry experience. Ann Oncol 2012;23(2):353-360.
  11. Wozniak A, Rutkowski P, Schoffski P, et al. Tumor genotype is an independent prognostic factor in primary gastrointestinal stromal tumors of gastric origin: a european multicenter analysis based on ConticaGIST. Clin Cancer Res 2014;20(23):6105-6116.
  12. Lasota J, Dansonka-Mieszkowska A, Sobin LH, et al. A great majority of GISTs with PDGFRA mutations represent gastric tumors of low or no malignant potential. Lab Invest 2004;84(7):874-883.
  13. Lasota J, Miettinen M. Clinical significance of oncogenic KIT and PDGFRA mutations in gastrointestinal stromal tumours. Histopathology 2008;53(3):245-266.
  14. Corless CL, Schroeder A, Griffith D, et al. PDGFRA mutations in gastrointestinal stromal tumors: frequency, spectrum and in vitro sensitivity to imatinib. J Clin Oncol 2005;23(23):5357-5364.
  15. Corless CL, Heinrich MC. Molecular pathobiology of gastrointestinal stromal sarcomas. Annu Rev Pathol 2008;3:557-586.
  16. Lasota J, Miettinen M. KIT and PDGFRA mutations in gastrointestinal stromal tumors (GISTs). Semin Diagn Pathol 2006;23(2):91-102.
  17. Martin-Broto J, Rubio L, Alemany R, et al. Clinical implications of KIT and PDGFRA genotyping in GIST. Clin Transl Oncol 2010;12(10):670-676.
  18. Martin-Broto J, Martinez-Marin V, Serrano C, et al. Gastrointestinal stromal tumors (GISTs): SEAP-SEOM consensus on pathologic and molecular diagnosis. Clin Transl Oncol 2017;19(5):536-545.
  19. Dibb NJ, Dilworth SM, Mol CD. Switching on kinases: oncogenic activation of BRAF and the PDGFR family. Nat Rev Cancer 2004;4(9):718-727.
  20. Barnett CM, Corless CL, Heinrich MC. Gastrointestinal stromal tumors: molecular markers and genetic subtypes. Hematol Oncol Clin North Am 2013;27(5):871-888.
  21. Chetty R, Serra S. Molecular and morphological correlation in gastrointestinal stromal tumours (GISTs): an update and primer. J Clin Pathol 2016;69(9):754-760.
  22. Medeiros F, Corless CL, Duensing A, et al. KIT-negative gastrointestinal stromal tumors: proof of concept and therapeutic implications. Am J Surg Pathol 2004;28(7):889-894.
  23. ESMO/European Sarcoma Network Working Group. Gastrointestinal stromal tumours: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014;25 Suppl 3:iii21-6.
  24. von Mehren M, Joensuu H. Gastrointestinal Stromal Tumors. J Clin Oncol 2018;36(2):136-143.
  25. Novartis Pharmaceuticals UK Ltd. Glivec 400 mg film-coated tablets - Summary of Product Characteristics. 2017. Accessed May 2018.
  26. Blanke CD, Rankin C, Demetri GD, et al. Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033. J Clin Oncol 2008;26(4):626-632.
  27. Verweij J, Casali PG, Zalcberg J, et al. Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: randomised trial. Lancet 2004;364(9440):1127-1134.
  28. Gramza AW, Corless CL, Heinrich MC. Resistance to Tyrosine Kinase Inhibitors in Gastrointestinal Stromal Tumors. Clin Cancer Res 2009;15(24):7510-7518.
  29. Roskoski R, Jr. Structure and regulation of Kit protein-tyrosine kinase--the stem cell factor receptor. Biochem Biophys Res Commun 2005;338(3):1307-1315.
  30. Biron P, Cassier PA, Fumagalli E, et al. Outcome of patients (pts) with PDGFRAD842V mutant gastrointestinal stromal tumor (GIST) treated with imatinib (IM) for advanced disease. J Clin Oncol 2010;28(15_suppl):10051.
  31. Cassier PA, Fumagalli E, Rutkowski P, et al. Outcome of patients with platelet-derived growth factor receptor alpha-mutated gastrointestinal stromal tumors in the tyrosine kinase inhibitor era. Clin Cancer Res 2012;18(16):4458-4464.
  32. Farag S, Somaiah N, Choi H, et al. Clinical characteristics and treatment outcome in a large multicentre observational cohort of PDGFRA exon 18 mutated gastrointestinal stromal tumour patients. Eur J Cancer 2017;76:76-83.
  33. Heinrich MC, Maki RG, Corless CL, et al. Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J Clin Oncol 2008;26(33):5352-5359.
  34. Pfizer Ltd. SUTENT 50 mg hard capsules - Summary of Product Characteristics. 2018. Accessed May 2018.
  35. Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 2006;368(9544):1329-1338.
  36. Prenen H, Cools J, Mentens N, et al. Efficacy of the kinase inhibitor SU11248 against gastrointestinal stromal tumor mutants refractory to imatinib mesylate. Clin Cancer Res 2006;12(8):2622-2627.
  37. Reichardt P, Demetri GD, Gelderblom H, et al. Correlation of KIT and PDGFRA mutational status with clinical benefit in patients with gastrointestinal stromal tumor treated with sunitinib in a worldwide treatment-use trial. BMC Cancer 2016;16:22.
  38. Bayer plc. Stivarga 40 mg film-coated tablets - Summary of Product Characteristics. 2017. Accessed May 2018.
  39. Demetri GD, Reichardt P, Kang YK, et al. Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013;381(9863):295-302.
  40. Kollar A, Maruzzo M, Messiou C, et al. Regorafenib treatment for advanced, refractory gastrointestinal stromal tumor: a report of the UK managed access program. Clin Sarcoma Res 2014;4:17.
  41. BLU-285, DCC-2618 show activity against GIST. Cancer Discov 2017;7:121-122.
Last update: 06 Jul 2018

This site uses cookies. Some of these cookies are essential, while others help us improve your experience by providing insights into how the site is being used.

For more detailed information on the cookies we use, please check our Privacy Policy.

Customise settings
  • Necessary cookies enable core functionality. The website cannot function properly without these cookies, and you can only disable them by changing your browser preferences.