Loading...

INTERNATIONAL JOURNAL OF CLINICAL AND MEDICAL CASES (ISSN:2517-7346)

Heterogenous Intertumoral KIT Mutations in Long-term Stage 4 Gastrointestinal Stromal Tumor Survivor

Seiya Liu1*, Irene Goo1, Ty K. Subhawong2, Andrew E. Rosenberg3, Jonathan C Trent4,6, Emily Jonczak4, Elizabeth Montgomery3, Alan Livingstone5, Gina D’Amato4,6

1University of Miami Miller School of Medicine, Miami, United States
2 Department of Radiology, University of Miami Miller School of Medicine, FL, United States
3 Department of Pathology,  University of Miami Miller School of Medicine, Miami, FL, United States
4 Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL, United States
5 Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
6 Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, United States

CitationCitation COPIED

Liu S, Goo I, Subhawong T, Rosenberg AE, Trent JC, et al. HeterogenousIntertumoral KIT Mutations in Long-term Stage 4 Gastrointestinal Stromal Tumor Survivor, Case Report. Int J Clin Med Cases. 2022 Feb;5(1):174.

Abstract

Gastrointestinal stromal tumors (GISTs) are mesenchymal neoplasms of the gastrointestinal tract, with a prevalence of 5% out of all sarcomas. We herein report the case of a 54-year-old man with metastatic GIST who initially presented to the emergency department with severe “anemia” CT examination revealed a 22 cm mass adherent to his small bowel and associated pelvic ascites and metastases. Surgical excision of the abdominal wall mass and partial omentectomy demonstrated extensive necrosis and a tumor mitotic rate of 17 per 5 mm2 . Molecular profiling of the primary mass showed a KIT exon 9 mutation. Despite eight years of sunitinib treatment, followup PET-CT evaluation showed three enlarging omental/peritoneal masses. Surgical excision and molecular profiling of each mass, opposed to the standard practice of profiling only one, demonstrated different KIT exon mutations between each mass, each with resistances to different tyrosine kinase inhibitor treatments. 

Article Highlights

• Metastatic gastrointestinal stromal tumor can attain novel mutations

• Performance of therapies may decline in the setting of novel KITmutations

• Molecular profiling of multiple metastatic nodules may aid in treating specific mutations

Keywords

Gastrointestinal stromal tumor, Tyrosine kinase inhibitor, Sarcoma, Nextgeneration sequencing, KIT exon mutation 

Introduction

Gastrointestinal stromal tumors (GISTs) are the most common type of mesenchymal neoplasm of the gastrointestinal tract [1]. KIT and PDGFRA mutations that characterize these tumors produce proteins that can be detected by immunohistochemistry in 85% of reported GISTs [2-5]. As such, CD117 (KIT) immunohistochemistry is a reliable diagnostic tool [6,7]. The development of next-generation sequencing (NGS) technology has also improved the detection of gene alterations in tumor-specific gene sequences, identifying new gene variations in KIT-expressing GISTs [8,9]. These identifiable and targetable mutations have led to the successful deployment of tyrosine kinase inhibitors (TKIs) such as imatinib, sunitinib, regorafenib, ripretinib and avapritinib for treatment of KIT-positive, KIT-mutant, or PDGRFA-positive GISTs [10-13]. In terms of clinical management, it is important to assess patient-specific and local tumor-specific GIST mutations for risk of resistance to existing therapies due to variations in molecular subsets [7,14,15]. Herein we report a case of recurrent, metastatic GIST with heterogenous intertumoral KIT mutations. To better understand this tumor and the associated treatments, we also reviewed relevant literature.

Case Study

A 54-year-old male with no past medical history presented to the emergency room with complaints of chronic fatigue. Bloodwork showed a hemoglobin of 4.9 and he received further work-up with computed tomography (CT). CT evaluation demonstrated a large 22cm mass adherent to his small bowel with associated pelvic ascites and numerous tumor deposits. The patient underwent surgical resection of the abdominal wall mass and partial omentectomy. The primary site was determined to be small bowel. Pathological analysis of the samples revealed a highgrade GIST with extensive necrosis and a mitotic rate of 17 per 5mm2 . Subsequent molecular analysis showed that the primary tumor harbored a KIT exon 9 mutation.

Given his metastatic disease, the patient initially received imatinib 400 mg daily, which was increased to 800 mg once a KIT exon 9 mutation was identified. Due to progression of disease after 6 months, he was transitioned to sunitinib, initially with a 50 mg regimen daily for four weeks on and 2 weeks off, then to a 37.5 mg continuous regimen due to adverse effects. With sunitinib he developed a chronic hand-foot skin reaction and diarrhea, for which he took a three-day break per month to alleviate symptoms. Additionally, he used urea cream for his hands and feet, as well as loperamide and diphenoxylate-atropine for diarrhea relief. He was followed for elevated transaminases, elevated ferritin, and iron saturation, but with no established diagnosis of hemochromatosis due to his history of hepatic steatosis. His iron levels improved after ceasing alcohol intake. He received phlebotomies for symptom relief.

After eight years of sunitinib use with stable disease, a followup PET-CT scan showed three enlarging omental/peritoneal implants on the right side of his abdomen, with the largest measuring 3.4 x 2.3 cm compared to 1.4 x 1.2 cm on previous scan five months prior, with a SUV uptake value of 11.7, increased from 5.0 (Figure 1a). The patient underwent metastasectomy, with excision of residual metastatic GIST in the retropubic space (space of Retzius), omentum, as well as removal of a left upper quadrant implant and Trucut biopsy of the left liver lobe. Of note, molecular profiling (Caris Life Sciences TM) was conducted for each excised tumor rather than the standardized practice of only assessing one site. Two omental implants demonstrated different KIT mutation variants on exon 17, exon 9, and exon 1, shown to be sensitive to regorafenib but to have resistance to imatinib and sunitinib. One pelvic peritoneum implant demonstrated KIT exon 9 and 13 mutation variants, while another retroperitoneal implant showed KIT exon 9 mutation variants. Both peritoneal implants were thus interpreted as susceptible to imatinib, to sunitinib if imatinib-refractory, or to regorafenib if imatinib/ sunitinib-refractory. Guardant 360 ®CDx liquid biopsy testing for comprehensive tumor mutation profiling revealed 0.2% cfDNA of TP53 R248W alteration but detected no KIT mutations. Four months after surgery, the patient developed ulcerative skin lesions on the heels of both feet, as well as elevated transaminases and testicular swelling. As such, sunitinib administration was withheld and other treatment modalities were considered.

Due to disease progression and worsening symptoms, the patient then received third-line treatment, namely regorafenib, at 160mg daily. During the first week of treatment, patient had poor tolerance of this regimen and therefore dose was decreased to 120 mg for one week. In the 18 days of regorafenib treatment, the patient reported complete loss of appetite, associated weight loss of five pounds, severe fatigue, one episode of unusual hemorrhage, bilateral upper extremity skin sensitivity and erythema, pain and swelling in the left knee, bilateral skin bullae in his heels, chills and shivering, and a fever of 104.8 Fahrenheit. Symptoms resolved after withholding treatment. Currently, the patient awaits initiation of a fourth-line therapy, namely ripretinib, for progressive disease despite regorafenib treatment (Figure 2).  

Figure 1: PET, CT, histology, and gross samples of patient tumor implants after eight years of sunitinib use with stable disease.

Discussion

Gastrointestinal stromal tumors (GISTs) are common gastrointestinal mesenchymal tumors that present with variable immunophenotypes [7,13,15]. The discovery of a host of KIT mutations in various exons has led to the development of effective targeted therapies using tyrosine kinase inhibitors (TKIs). While imatinib has been established as an effective first-line therapy for GIST, about 15% of GIST patients have tumors that are resistant to imatinib due to variant KIT mutations, notably in exons 9, 13, and 17 [5,7,14,16]. The emergence of sunitinib and other TKIs as treatment options in imatinib-resistant patients has provided other viable treatment options in the face of singular resistance to imatinib [5,10,17].

This 54-year-old male patient with metastatic GIST originating from the small bowel demonstrates the importance of conducting next-generation sequencing (NGS) on multiple metastatic sites. Given the heterogenous mutations at each tumor location, clinical management would be dramatically different without the relevant knowledge of exon-specific resistances. The sudden development of disease progression and metastatic implants despite multiple years of stable disease with sunitinib treatment demonstrates the importance of follow-up, imaging, and next generation sequencing of resected tumors. Previous studies have also described the development of multiple TKI-resistant KIT mutations after initial imatinib and subsequent sunitinib treatment [17]. If metastatic implants have heterogenous KIT mutations, as is the case for this patient, the clinical approach must change appropriately to effectively control further disease progression.

Of particular interest, the omental implants with KIT exon 17, exon 9, and exon 1 mutations, respectively, grew the most aggressively compared to the other pelvic, peritoneal, and retroperitoneal implants with KIT exon 9 and 13 mutations. Exon 17 mutations are most common in small intestinal GIST with no discernible difference in clinical prognosis compared to nonKIT-mutated, wild-type GISTs [18]. Given the patient’s clinical presentation, one of the omental implants with an exon 17 mutation likely resulted from tumor implantation of the primary site. However, the largest omental implant with KIT exon 17 and 9 mutations is consistent with previous studies that outlined the aggressiveness of non-gastric exon 9 mutated tumors [5,18]. KIT exon 13 mutations are more common in secondary imatinib resistance, and associated with aggressive behavior for gastric GISTs, but not for small intestinal GISTs, which are intrinsically more aggressive than gastric GISTs [19]. The KIT exon mutations present in the implants are consistent with expected growth patterns of non-gastric GISTs. The omental GISTs harboring KITexon 9 mutations demonstrate the most explosive growth and the retroperitoneal and pelvic peritoneum implants with KIT exon 9 and 13 mutations demonstrated lower levels of growth and smaller size compared to the omental implants (Figure 2).

Localized GISTs are commonly treated with surgical excision. Metastatic disease in the context of GIST is less likely to be resolved with surgical excision, but rather require more targeted approaches [8]. The development of NGS has opened the door to identifying targetable mutations. While standard practice of use for NGS has been limited to assessing only one site of disease for mutations, as opposed to multiple tumor locations, this case of intertumoral heterogenous mutations demonstrates the need to analyze multiple sites in the setting of metastatic disease [5]. Although GIST has a relatively high 5-year survival rate across all stages, long-term survival (>10+ years) dramatically decreases despite standard TKI treatment, potentially due to the rise of novel TKI-resistant mutations [20]. 

Figure 2: Exon variants of each excised tumor with potential benefits and lack of benefits to treatments.

Conclusion

Improved understanding of KIT mutations will lead to improved outcomes. Assessing patient-specific and local tumorspecific GIST mutations for variations in resistance is crucial for clinical management of treatment-resistant GISTs. Discovering agents that will target the current TKI resistant mutations and implementing multi-tumor analysis is therefore of the utmost importance.

References

1. Zhao X, Yue C. Gastrointestinal stromal tumor. JGastrointestOncol. 2012 Sep;3(3):189-208.
2. Kindblom LG, Remotti HE, Aldenborg F, Meis-KindblomJM. Gastrointestinal pacemaker cell tumor (GIPACT):gastrointestinal stromal tumors show phenotypiccharacteristics of the interstitial cells of Cajal. Am J Pathol.1998 May;152(5):1259-1269.
3. Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998 Jan;279(5350):577-580. 
4. Heinrich MC, Corless CL, Duensing A, McGreevey L, Chen CJ, etal. PDGFRA activating mutations in gastrointestinal stromaltumors. Science. 2003 Jan;299(5607):708-710.
5. Oppelt PJ, Hirbe AC, Van Tine BA. Gastrointestinal stromaltumors (GISTs): point mutations matter in management, areview. J GastrointestOncol. 2017 Jun;8(3):466-473.
6. Hornick JL, Fletcher CD. Immunohistochemical stainingfor KIT (CD117) in soft tissue sarcoma is very limited indistribution. Am J Pathol. 2002 Feb;117(2):188-193.
7. Chirieac LR, Trent JC, Steinert DM, Choi H, Yang Y, et al.Correlation of immunophenotype with progressionfree survival in patients with gastrointestinal stromaltumors treated with imatinibmesylate. Cancer. 2006Nov;107(9):2237-2244.
8. Jour G, Scarborough JD, Jones RL, Loggers E, Pollack SM, etal. Molecular profiling of soft tissue sarcomas using nextgeneration sequencing: a pilot study toward precisiontherapeutics. Hum Pathol. 2014 Aug;45(8):1563-1571.
9. Fletcher JA, Rubin BP. KIT mutations in GIST. CurrOpin GenetDev. 2007 Feb;17(1):3-7.
10. Joensuu H, Roberts PJ, Sarlomo-Rikala M, Andersson LC,Tervahartiala P, et al. Effect of the tyrosine kinase inhibitorSTI571 in a patient with a metastatic gastrointestinal stromaltumor. N Engl J Med. 2001 Apr;344(14):1052-1056.
11. George S, Wang Q, Heinrich MC, Corless CL, Zhu M, et al.Efficacy and safety of regorafenib in patients with metastaticand/or unresectable GI stromal tumor after failure of imatiniband sunitinib: a multicenter phase II trial. J ClinOncol. 2012Jul;30(19):2401-2407.
12. Wu CP, Lusvarghi S, Wang JC, Hsiao SH, Huang YH, et al.Avapritinib: A Selective Inhibitor of KIT and PDGFRalpha thatReverses ABCB1 and ABCG2-Mediated Multidrug Resistancein Cancer Cell Lines. Mol Pharm. 2019;16(7):3040-3052.
13. Blay JY, Serrano C, Heinrich MC, Zalcberg J, Bauer S, et al.Ripretinib in patients with advanced gastrointestinal stromaltumours (INVICTUS): a double-blind, randomised, placebocontrolled, phase 3 trial. Lancet Oncol. 2020 Jul;21(7):923-934.
14. Blay JY, Le Cesne A, Cassier PA, Ray-Coquard IL.Gastrointestinal stromal tumors (GIST): a rare entity, atumor model for personalized therapy, and yet ten differentmolecular subtypes. Discov Med. 2012 May;13(72):357-367.
15. Grunewald S, Klug LR, Muhlenberg T, Lategahn J, FalkenhorstJ, et al. Resistance to Avapritinib in PDGFRA-Driven GISTIs Caused by Secondary Mutations in the PDGFRA KinaseDomain. Cancer Discov. 2021 Jan;11(1):108-125.
16. Boyar MS, Taub RN. New strategies for treating GIST whenimatinib fails. Cancer Invest. 2007 Aug;25(5):328-335.
17. Liegl B, Kepten I, Le C, Zhu M, Demetri GD, Heinrich MC, et al.Heterogeneity of kinase inhibitor resistance mechanisms inGIST. J Pathol. 2008 Sep;216(1):64-74
18. Zhang H, Liu Q. Prognostic Indicators for Gastrointestinal Stromal Tumors: A Review. TranslOncol. 2020Oct;13(10):100812.
19. Lasota J, Corless CL, Heinrich MC, Debiec-Rychter M, SciotR, et al. Clinicopathologic profile of gastrointestinal stromaltumors (GISTs) with primary KIT exon 13 or exon 17mutations: a multicenter study on 54 cases. Mod Pathol.2008;21(4):476-484.
20. Heinrich MC, Rankin C, Blanke CD, Demetri GD, Borden EC, etal. Correlation of Long-term Results of Imatinib in Advanced Gastrointestinal Stromal Tumors With Next-GenerationSequencing Results: Analysis of Phase 3 SWOG Intergroup Trial S0033. JAMA Oncol. 2017;3(7):944-952.