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

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

ESMO Factsheets on Biomarkers

KIT

The KIT proto-oncogene maps to 4q12 and encodes the 145-kDA transmembrane receptor tyrosine kinase, KIT (CD117), the cellular homolog of the Hardy–Zuckerman 4 feline sarcoma viral oncogene, v-kit[1, 2]. KIT is a member of the type III tyrosine kinase receptor family, which also includes the platelet derived growth factor receptor-alpha (PDGFRA)[2]. KIT is activated through binding of its ligand, stem cell factor, to its extracellular domain, which initialises downstream signalling cascades, including the JAK–STAT3, phosphatidylinositide-3- kinase (PI3K)–AKT–mTOR, and RAS–MAPK pathways, important in regulating cellular functions such as proliferation and apoptosis, chemotaxis and adhesion[1, 3]. Normal expression of KIT is critical for the development and maintenance of different cell types, including interstitial cells of Cajal involved in gastrointestinal pacemaker activity[1, 3].

In 1998 a critical discovery showed that gain-of-function mutations in KIT are a key oncogenic driver in most gastrointestinal stromal tumours (GISTs)[4]. These mutations are present in over 80% of GISTs and lead to constitutive, ligand-independent activation of the KIT receptor and its downstream pathways, ultimately increasing cell proliferation and inhibiting apoptosis[2].

KIT mutations in GIST

Mutations in KIT mainly affect those exons that encode the functional domains of the tyrosine kinase receptor, namely exons 9, 11, 13 and 17[3].

Exon 11, which encodes the juxtamembrane domain of KIT, is the most frequently mutated region, affecting 70–75% of GIST[5, 6]. The conformational changes in KIT due to exon 11 mutations disrupt the autoinhibitory domain of the receptor and permit continuous kinase activation[7]. Deletions affecting codons 557-558 of exon 11 are the most common KIT mutation, detected in 23-28% of GISTs[2]. Point mutations, duplications and homo/hemizygous exon 11 mutation status have also been identified[2]. The vast majority (>80%) of exon 11-mutated GISTs are located in the stomach, although they may arise essentially anywhere in the GI tract[1].

Mutations in exon 9, which encodes the extracellular domain of KIT, occur in 7–15% of GIST cases[2]. These mutations are believed to mimic the conformational change that the extracellular KIT receptor undergoes when ligand is bound, thus leading to dimerisation and constitutive activation[1]. The majority of exon 9 mutations are duplications of codons 502 and 503 and are almost exclusively associated with GISTS of small intestinal origin[3, 8]

Mutations in exons 13 and 17 are not common, occurring in 1-2% of GISTs[2, 9, 10]. Mutations in exon 13 encoding the ATP-binding region of KIT are speculated to interfere with the physiological auto-inhibitory function of the juxtamembrane domain, and usually arise in the stomach[1, 10]. Mutations in exon 17 encoding the activation loop of the kinase seem to stabilize the active conformation[10], and appear to arise twice as frequently in the small intestine as in the stomach[1, 2]. Mutations in KIT can also appear after treatment, termed secondary mutations, and are usually in exons 13, 14, and 17 accompanied by a primary KIT mutation[11]

KIT 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)[11]. KIT detection by immunohistochemistry is unrelated to the existence of underlying KIT mutations, which have been identified in a proportion of GISTs that stain negative for CD117[11, 12]. Inclusion of mutational analysis for known mutations in genes such as KIT in the diagnostic work-up of GIST is paramount for the selection of appropriate therapy (see below) but may also help confirm the diagnosis of suspect GIST that do not exhibit positive immunoreactivity for CD117/DOG1[13]

KIT mutations as a prognostic biomarker in GIST

Exon 11 deletions affecting codons 557/558 have been associated with an aggressive, metastasising phenotype and indicate an overall poorer prognosis in both high-risk and non­high-risk gastric GIST[2, 14-16]. In contrast, gastric GISTs harbouring exon 11 duplications or point mutations have been linked with a more favourable clinical course[2, 15, 16]

KIT exon 9 duplications have been associated with a poor clinical prognosis as compared with other mutations[16]. However, the worse prognosis of KIT exon 9 mutants seems to be related to the almost exclusive high-risk extra-gastric tumour location of these GISTs rather than to an intrinsic aggressive biologic behaviour[2]

KIT mutations as a predictive biomarker in GIST

As a key oncogenic driver expressed in the majority of GISTs, KIT is a key therapeutic target. KIT mutational status holds predictive value for GIST sensitivity to targeted treatments and routine genotyping has become an integral part of management of GISTs undergoing tyrosine kinase inhibitor therapy[2].

Imatinib (Glivec®, Novartis Pharmaceuticals), a tyrosine kinase inhibitor that competitively binds the ATP-binding domain of KIT has demonstrated pronounced clinical efficacy in GISTs [17-19].  Imatinib 400 mg/day was licensed over 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[19].

In the adjuvant setting, studies demonstrate that patients with KIT exon 11 mutations benefit most from imatinib[20, 21].  In the ACOSOG Z 9001 trial of 645 patients receiving one year adjuvant imatinib or placebo after resection of primary GIST, imatinib therapy was associated with longer recurrence-free survival in patients with a exon 11 deletion of any type[20]. In another study, patients with exon 11 mutations benefited from longer 3-year adjuvant treatment, whereas no significant improvement over 12 months of imatinib was found in the subsets of patients whose GIST harboured an exon 9 or PDGFRA mutation, or patients who had no mutation in either gene, although patient numbers were small in these latter groups[21].

In the advanced GIST setting, improved treatment outcomes with imatinib have been observed for patients with KIT exon 11 mutations compared with patients with exon 9 mutations or wild-type KIT[22, 23]. In the North American Phase III SWOG S0033 study, the presence of exon 11 mutation (n = 283) correlated with improved treatment outcome when compared with exon 9 mutation (n = 32) and wild-type (n = 67) genotypes for objective response (71.7% v 44.4% [P=0.007]; and 44.6% [P=0.0002], respectively); time to tumour progression (median 24.7 months v 16.7 and 12.8 months, respectively); and overall survival (OS) (median 60.0 months v 38.4 and 49.0 months, respectively)[23]. In the joint analysis of the S0033 and European Organisation for Research and Treatment of Cancer 62005 trials comparing standard 400 mg once daily imatinib dose versus 400 mg twice daily, the sole predictive factor of response was the presence of a KIT exon 9 mutation[24]. The estimated risk of progression or death was reduced by 42% in the high-dose arm (compared with the standard-dose arm) in patients with KIT exon 9 mutated tumours. The study concluded that for most patients, the recommended imatinib dose is 400 mg daily, with exception of KIT exon 9 mutated tumours for which the 400 mg twice daily dose can be considered[24].

Although 80–85% patients with advanced GIST benefit from imatinib treatment, all will subsequently develop secondary resistance, which is frequently associated with secondary KIT mutations[2]. Imatinib can only bind to the inactive conformation of KIT, and both primary and secondary resistance to imatinib can be partially explained by a conformational shift in the kinase domain of KIT that favours the activated state[2, 25]. Activation loop mutations (i.e. those affecting exon 17) indirectly induce imatinib resistance by stabilising the active receptor conformation[7]. In a Phase I/II study, half of the patients progressing on imatinib had a secondary KIT mutation detected in either exon 13, 14 or 17[26]. Secondary mutations were more likely to be found in patients who initially harboured KIT exon 11 mutations (73%) as compared with KIT exon 9 mutations (19%), likely due to the longer exposure to imatinib of the primary exon 11 mutant subgroup[2, 26].

In 2006 a second tyrosine kinase inhibitor, Sunitinib (Sutent®, Pfizer), was approved for use in imatinib-resistant GIST[27]. Sunitinib (50 mg/day over 4 weeks followed by 2-week rest period) is licensed for the treatment of unresectable and/or metastatic malignant GIST in adults after failure of imatinib treatment due to resistance or intolerance [27, 28].

Response to sunitinib appears to be influenced by both primary and secondary KIT mutations.  Patients harbouring primary exon 9 mutations appear to achieve better outcomes than those with exon 11 mutation or wild-type KIT/PDGFRA[26, 29]. In a worldwide, open-label treatment-use study of patients resistant or intolerant to imatinib a median progression-free survival (PFS) of 7.1 months was reported for patients who received sunitinib[29]. The PFS in patients with a primary exon 9 mutation was significantly better as compared with those with a primary exon 11 mutation (12.3 vs. 7.0 months, respectively), which also translated to longer overall survival (OS) and higher objective response rate ORR. Regarding secondary mutations, in a study including primarily KIT-mutated GIST patients, the median PFS and OS with sunitinib was significantly longer for secondary KIT exon 13 or 14 mutations (which involve the KIT-ATP binding pocket) than for those with secondary exon 17 or 18 mutations (which involve the KIT activation loop) (PFS 7.8 vs. 2.3 months, respectively; OS 13.0 vs. 4.0 months, respectively[26].

A third tyrosine kinase inhibitor, Regorafenib (Stivarga®, Bayer), is indicated for patients with unresectable or metastatic GIST who progressed on or are intolerant to prior treatment with imatinib and sunitinib[30, 31]. There is little evidence available of the predictive power of mutational status in third-line regorafenib treatment of GISTs, but and similar benefits have been observed for patients with primary KIT exon 11 or exon 9 mutations[30].

KIT 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[11].  

The most widely used method for detecting KIT 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[11]. Next-generation sequencing can provide greater sensitivity with several GIST-specific gene panels now commercially available[11].

Patient selection

Given its diagnostic, prognostic and predictive value, mutational analysis of KIT should be included in the diagnostic work-up of all GISTs as standard practice for optimal management[13].

References

  1. Mei L, Smith SC, Faber AC, et al. Gastrointestinal Stromal Tumors: The GIST of Precision Medicine. Trends Cancer 2018;4(1):74-91.
  2. 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.
  3. 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.
  4. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 1998;279(5350):577-580.
  5. Corless CL, Heinrich MC. Molecular pathobiology of gastrointestinal stromal sarcomas. Annu Rev Pathol 2008;3:557-586.
  6. Lasota J, Miettinen M. Clinical significance of oncogenic KIT and PDGFRA mutations in gastrointestinal stromal tumours. Histopathology 2008;53(3):245-266.
  7. Gajiwala KS, Wu JC, Christensen J, et al. KIT kinase mutants show unique mechanisms of drug resistance to imatinib and sunitinib in gastrointestinal stromal tumor patients. Proc Natl Acad Sci U S A 2009;106(5):1542-1547.
  8. Antonescu CR, Sommer G, Sarran L, et al. Association of KIT exon 9 mutations with nongastric primary site and aggressive behavior: KIT mutation analysis and clinical correlates of 120 gastrointestinal stromal tumors. Clin Cancer Res 2003;9(9):3329-3337.
  9. Joensuu H, Rutkowski P, Nishida T, et al. KIT and PDGFRA mutations and the risk of GI stromal tumor recurrence. J Clin Oncol 2015;33(6):634-642.
  10. Lasota J, Corless CL, Heinrich MC, et al. Clinicopathologic profile of gastrointestinal stromal tumors (GISTs) with primary KIT exon 13 or exon 17 mutations: a multicenter study on 54 cases. Mod Pathol 2008;21(4):476-484.
  11. 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.
  12. 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.
  13. 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:iii21-26.
  14. Martin J, Poveda A, Llombart-Bosch A, et al. Deletions affecting codons 557-558 of the c-KIT gene indicate a poor prognosis in patients with completely resected gastrointestinal stromal tumors: a study by the Spanish Group for Sarcoma Research (GEIS). J Clin Oncol 2005;23(25):6190-6198.
  15. 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.
  16. 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.
  17. 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.
  18. 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.
  19. Novartis Pharmaceuticals UK Ltd. Glivec 400 mg film-coated tablets - Summary of Product Characteristics. 2017. Accessed May 2018.
  20. 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.
  21. Joensuu H, Wardelmann E, Sihto H, et al. Effect of KIT and PDGFRA mutations on survival in patients with gastrointestinal stromal tumors treated with adjuvant imatinib: an exploratory analysis of a randomized clinical trial. JAMA Oncol 2017;3(5):602-609.
  22. 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.
  23. 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.
  24. Gastrointestinal Stromal Tumor Meta-Analysis Group (Meta-GIST). Comparison of two doses of imatinib for the treatment of unresectable or metastatic gastrointestinal stromal tumors: a meta-analysis of 1,640 patients. J Clin Oncol 2010;28(7):1247-1253.
  25. 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.
  26. 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.
  27. Pfizer Ltd. SUTENT 50 mg hard capsules - Summary of Product Characteristics. 2018. Accessed May 2018.
  28. 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.
  29. 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.
  30. 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.
  31. Bayer plc. Stivarga 40 mg film-coated tablets - Summary of Product Characteristics. 2017.
Last update: 05 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.