Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Nov 7, 2021; 27(41): 7125-7133
Published online Nov 7, 2021. doi: 10.3748/wjg.v27.i41.7125
Current update on molecular cytogenetics, diagnosis and management of gastrointestinal stromal tumors
Mindy X Wang, Catherine Devine, Dhakshinamoorthy Ganeshan, Department of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
Nicole Segaran, Department of Radiology, Mayo Clinic Arizona, Phoenix, AZ 85259, United States
ORCID number: Mindy X Wang (0000-0002-3457-9327); Catherine Devine (0000-0003-0353-2574); Nicole Segaran (0000-0002-6787-3004); Dhakshinamoorthy Ganeshan (0000-0001-5027-3347).
Author contributions: Wang MX, Devine C and Segaran N contributed to the manuscript; all authors contributed to the design of the study; Ganeshan D designed the structure of the overall manuscript, made critical revisions related to important intellectual content of the manuscript; and all authors approved the final version of the manuscript.
Conflict-of-interest statement: The authors declare no conflict of interests for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Dhakshinamoorthy Ganeshan, MD, Associate Professor, Department of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States. dganeshan@mdanderson.org
Received: June 7, 2021
Peer-review started: June 7, 2021
First decision: June 20, 2021
Revised: July 28, 2021
Accepted: September 15, 2021
Article in press: September 15, 2021
Published online: November 7, 2021
Processing time: 152 Days and 2.7 Hours

Abstract

Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal (GI) tract and are thought to arise from precursors of the interstitial cells of Cajal. GISTs can arise anywhere in the GI tract, but most commonly originate from the stomach and small intestine. The majority of GISTs occur as a result of activating mutations in two receptor protein tyrosine kinases: KIT and/or platelet-derived growth factor receptor-α. Mutational analyses allow for predicting patient prognosis and treatment response. Clinical presentations can vary from no symptoms, typical in the case of small incidentally found tumors, to GI bleeding, abdominal discomfort, and ulcer-related symptoms when the tumor is enlarged. Imaging plays a critical role in the diagnosis and management of these tumors with multiphasic computed tomography serving as the imaging modality of choice. Magnetic resonance imaging and positron emission tomography-computed tomography can serve as imaging adjuncts in lesion characterization, especially with liver metastases, and subsequent staging and assessment for treatment response or recurrence. Surgical resection is the preferred management for small GISTs, while tyrosine kinase inhibitors − imatinib mesylate and sunitinib malate − serve as crucial molecular-targeted therapies for locally advanced and metastatic GISTs. This review article highlights the clinical presentation, pathology and molecular cytogenetics, imaging features, and current management of GISTs.

Key Words: Gastrointestinal stromal tumors, Cytogenetics, Diagnostic imaging, Computed tomography, Magnetic resonance imaging, Imatinib mesylate

Core Tip: Gastrointestinal stromal tumors (GISTs) often occur as a result of activating mutations in two receptor protein tyrosine kinases: KIT and/or platelet-derived growth factor receptor-α, allowing for effective molecular targeted therapies for these patients. Mutational analyses help predict patient prognosis and treatment response. Imaging plays a critical role in the diagnosis and management of GISTs. Multiphasic computed tomography serves as the imaging modality of choice in their diagnosis and follow-up. It is crucial to understand and identify the key imaging features of GISTs and their expected appearance with treatment response and disease recurrence.



INTRODUCTION

Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal (GI) tract and are thought to arise from the precursors of the interstitial cells of Cajal. GISTs can arise anywhere in the GI tract, most commonly from the stomach and small intestine[1,2]. The majority of GISTs occur as a result of activating mutations in two receptor protein tyrosine kinases: KIT and/or platelet-derived growth factor receptor-α (PDGFRA)[1]. Clinical presentations can vary from no symptoms, typical in the case of small incidentally found tumors, to GI bleeding, abdominal discomfort, and ulcer-related symptoms when the tumor is enlarged. Imaging plays a critical role in the diagnosis and management of these tumors with multiphasic computed tomography (CT) serving as the imaging modality of choice. Magnetic resonance imaging (MRI) and positron emission tomography-computed tomography (PET-CT) can serve as imaging adjuncts in lesion characterization, especially with liver metastases, and subsequent staging and assessment for treatment response or recurrence. Surgical resection is the preferred management for small GISTs, while tyrosine kinase inhibitors − imatinib mesylate and sunitinib malate − serve as crucial targeted therapies for locally advanced and metastatic GISTs[1].

This review article highlights the clinical presentation, pathology and molecular cytogenetics, imaging features, and current management of GISTs.

EPIDEMIOLOGY

The annual incidence of GISTs is estimated to be at least 3000 per year in the United States[1]. GISTs are often diagnosed in older adults ages 50-70 years with a median age at diagnosis ranging from 59 to 66 years[3-5]. GISTs can occur in all geographic and ethnic groups, and men and women are equally affected[6]. There are no known risk factors for developing GIST. A small subset of patients may present with the non-inherited Carney’s triad, which is comprised of GIST, often with loss of function of succinate dehydrogenase (SDH), paragangliomas, and pulmonary chondromas[1]. While most GISTs occur sporadically, rare hereditary GISTs have been reported[5]. Familial GISTs are related to inherited germline mutations in either KIT or PDGFRA and also manifest with cutaneous hyperpigmentation, irritable bowel syndrome, dysphagia, and diverticular disease[1]. In Caryney-Stratakis syndrome, patients present with GIST and paragangliomas related to loss of function mutations within SDH genes. Small intestinal GISTs can also be associated with neurofibromatosis type 1, an autosomal dominant disorder in which patients more often present with café au lait spots, gliomas, and neurofibromas[1,5]. A very small group of GISTs (1%-2%) occur in the pediatric population.

CLINICAL FEATURES

Clinical presentations vary depending on the size and location of tumors. GISTs can arise anywhere along the GI tract. They most often arise from the stomach (60%), followed by the jejunum and ileum (30%), duodenum (5%), colorectum (4%), and esophagus or appendix (< 1%)[2]. Rarely, GISTs can develop outside the GI tract in the mesentery, omentum, or retroperitoneum. The majority of GISTs are benign (70%-80%). Patients are often asymptomatic, especially when the tumor size is small (less than 2 cm)[1]. When the tumor is enlarged, symptoms may include abdominal pain, bleeding, abdominal distension, early satiety, fatigue, and palpable mass[7]. Unfortunately, these nonspecific symptoms may result in delayed diagnosis and management of the disease. Infrequently, patients with advanced GISTs may present with severe hypoglycemia and hypothyroidism[8,9]. Laboratory work-up may reveal anemia, which may be related to bleeding or intratumoral hemorrhage. Metastases are uncommon (10%-20% of cases); however, when they do occur, they can occur via local or hematogenous spread. The most common metastatic sites include the liver, omentum, and peritoneal cavity[1,2]. Lymph node and extra-abdominal metastases are extremely rare[5]. In severe cases, patients may present with acute abdomen, melena, or hematemesis secondary to frank hemorrhage due to tumor invasion or rupture. Such emergent clinical presentation is more often seen in small intestine GISTs compared to gastric GISTs[5].

Pediatric GISTs occur in approximately 1%-2% of cases and are predominantly seen in girls presenting with multiple nodules in the stomach. These patients typically present with anemia, weakness, and syncope due to GI bleeding[1]. In addition to liver and peritoneal metastases, lymph node metastases uniquely occur in this group of patients.

PATHOLOGY AND MOLECULAR CYTOGENETICS

In gross pathology, GISTs can widely vary in size, ranging from 1-2 cm to more than 20 cm in diameter. The median size at presentation is approximately 5 cm[1]. They are well-circumscribed gray-white to red-brown masses in the bowel wall that can be submucosal, intramural, or subserosal in location[10]. They are generally unencapsulated but may have pseudocapsules. GISTs typically arise from the muscularis propria and exhibit an exophytic growth pattern. Intraluminal or mixed growth patterns may also be seen. There are three main histologic subtypes: (1) Spindle cell (60%-70%); (2) Epithelioid (30%-40%); and (3) Combination of both spindle cell and epithelioid in various proportions[10]. On microscopy, spindle cell subtypes demonstrate highly cellular, fascicular, whorled, storiform, or palisading architecture, while epithelioid tumors appear more fascicular or nested[10]. The mitotic rates can vary widely from virtually absent to high. Other findings may include areas of hemorrhage or necrosis.

The majority of GISTs (80%-90%) occur as a result of activating mutations in two receptor protein tyrosine kinases: KIT and/or PDGFRA. In 1998, Hirota et al[11] published the revolutionary finding that the majority of GISTs (94%) expressed activating mutations in KIT (CD117), now a key diagnostic immunohistochemical marker for GIST that distinguishes it from leiomyomas, leiomyosarcomas, or other mesenchymal tumors. KIT belongs to the type II transmembrane receptor tyrosine kinase family that includes PDGFRA and PDGFRB. The c-kit proto-oncogene encodes KIT; in combination with the stem cell factor extracellular ligand, this c-kit product normally plays an essential role in cellular survival, proliferation, and differentiation[10]. Activating mutations in KIT result in altered cell growth. In addition, Hirota et al[11] demonstrated that the interstitial cells of Cajal, which are the pacemaker cells involved in regulating the peristalsis located primarily in the muscularis propria, were the only cells that were double-positive for KIT and CD34 in the GI wall. Therefore, GISTs, which share morphological, structural, and immunohistochemical features with interstitial cells of Cajal, are thought to arise from them or their stem cell precursors[10-12]. Germline or sporadic gain of function mutations in c-kit result in both benign and malignant GIST tumorigenesis[10]. Aside from KIT mutation, PDFGFRA mutation can be an alternative cytogenetic change that can also result in similar downstream effects of tumor progression[13]. Hence, imatinib mesylate, a selective adenosine triphosphate-competitive inhibitor of KIT, PDGFRA and PDGFRB, serves as a groundbreaking therapy for GISTs[14]. Furthermore, DOG1, a calcium-dependent, receptor-activated chloride channel protein, has been found to be expressed in GISTs regardless of mutation type; this marker can aid in the diagnosis of KIT-negative tumors, such as those with PDGFRA mutations that are KIT-negative[15].

Other markers for GISTs include CD34 antigen (70%), smooth muscle actin (30%-40%), desmin (< 5%), and S100 protein (approximately 5%)[16]. The expression of these markers varies depending on the location of the tumor. CD34 is often found in esophageal, gastric, and rectal tumors, while smooth muscle actin is seen in small intestine tumors. Prognostic predictors vary considerably in the literature. It has been suggested that mitotic activity and tumor size are potential prognostic predictors: A mitotic index of at least 5 per 50 high power fields (HPF) and a size greater than 5 cm are suggestive of malignant behavior, while a mitotic index of 5 or less per 50 HPF and a size less than 2 cm are suggestive of benign GIST[1,6,17]. Ki-67 can also be used to predict malignant potential[18]. Tumor location is another prognostic factor; intestinal GISTs demonstrate worse outcomes compared to gastric GISTs with regard to tumor size and mitotic rates[19]. GISTs that carry KIT exon 11 point mutations and insertions have a favorable prognosis, while those with KIT exon 9 mutations or KIT exon 11 deletions have a worse prognosis[19,20]. A small number of patients with GISTs may harbor concomitant BRAF gene mutations, which may portend poorer prognosis due to their primary resistance to imatinib mesylate therapy[21]; in such cases, patients may benefit from selective BRAF inhibitors. Further genotyping is advised for patients with KIT-negative GIST for management planning.

In the pediatric population, GISTs typically do not have KIT or PDGFRA mutations, and generally demonstrate the epithelioid subtype and express CD117[22]. Compared to adults, these pediatric GISTs uniquely overexpress fibroblast growth factor 4 (FGF4), brain and acute leukemia, cytoplasmic, insulin-like growth factor I receptor, NEL-like 1, cytokine receptor-like factor 1, pleomorphic adenoma gene 1, and FGF3)[23]. With KIT activation, these GISTs are similar to adult GISTs that carry KIT mutations. Although there is limited literature on the clinical benefits of tyrosine kinase inhibitors, sunitinib malate is suspected to be superior to imatinib mesylate in these pediatric cases[23].

IMAGING FEATURES
Fluoroscopic examination

Fluoroscopic examination is not routinely used for identifying GISTs. However, patients who undergo double-contrast barium studies may demonstrate a submucosal or well-circumscribed mass with smooth mucosal surface and obtuse angles at the margins[24]. With necrosis or ulceration, they may demonstrate irregular contours. Evaluation of extraluminal structures is limited with this approach. Further evaluation of the lesion and presence of metastatic disease with cross-sectional imaging is ultimately required.

Ultrasonography

Ultrasonography is not routinely used for imaging GISTs, especially since the tumor origin cannot be well-identified. When small, GIST may be homogeneously hypoechoic. When large, GIST may present as a heterogenous mass, which may reflect internal necrosis or hemorrhage; these findings suggest high malignant potential[25]. Hepatic metastases can be identified, although their sonographic appearance is nonspecific.

CT

CT serves as the imaging modality of choice in the diagnosis and follow-up of GISTs. Multiphasic protocol with noncontrast, arterial, and portal venous phases should be obtained. The noncontrast images help identify hemorrhage and provide a baseline for evaluating tumor enhancement. Adequate gastric distension is essential to help distinguish intramural mass; therefore, negative oral contrast agents can aid in the visualization of the enhancing mucosa[26]. The imaging appearance of GISTs depends on their size and aggressiveness. Classic CT features of GISTs include large, hypervascular, enhancing masses that may demonstrate heterogeneity due to hemorrhage, cystic degeneration, or necrosis[27]. These tumors typically displace adjacent structures and vessels, although they may exhibit direct invasion of adjacent structures resulting in ulceration and fistulization in the GI tract in advanced stages. When small, GISTs appear homogeneous and may be incidentally found on CT or endoscopy. Metastases are present in approximately 50% of patients, and metastases often involve the liver and mesentery; they demonstrate similar imaging features as primary GISTs[24,27]. Lymph node metastases are extremely rare[27]. Features of high-grade GISTs include liver metastasis, GI wall infiltration, irregular surface, ill-defined margins, inhomogeneous enhancement, and peritoneal spread[28].

Regardless of tumor size, a change from a heterogeneously hyperattenuating mass to a homogeneously hypoattenuating mass with decreased enhancing tumor nodules and intratumoral vessels suggest response to imatinib mesylate[24,27]. The attenuation of treated lesions reaches approximately 20-25 Hounsfield units, which is close to simple density[29]. Although the tumors may enlarge during treatment as a result of intratumoral hemorrhage or myxoid degeneration, this does not suggest disease progression in the setting of decreased tumor enhancement[27]. The Response Evaluation Criteria in Solid Tumors has been found to be insensitive in evaluating treatment response as it does not account for tumor density, intratumoral vessels, or tumor metabolism[30]. Therefore, Choi et al[31] proposed a modified CT response evaluation criteria to account for such features on CT as tumor response to tyrosine kinase inhibitor therapy cannot be determined based on size alone (Table 1). Disease recurrence is signified by the development of enhancing tumor nodules within the treated hypoattenuating tumor[27]. A summary of key imaging features is highlighted in Table 2.

Table 1 Modified computed tomography response evaluation criteria for gastrointestinal stromal tumors.
Response
Definition
Complete responseDisappearance of all lesions; No new lesions
Partial responseA decrease in size1 of ≥ 10% or decrease in tumor density (HU) ≥ 15% on CT; No new lesions; No obvious progression of nonmeasurable disease
Stable diseaseDose not meet criteria for complete response, partial response, or progressive disease; No symptomatic deterioration attributed to tumor progression
Progressive diseaseAn increase in tumor size of ≥ 10% and does not meet criteria of partial response by tumor density (HU) on CT; New lesions; New intratumoral nodules or increase in size of existing intratumoral nodules
Table 2 Imaging features of gastrointestinal stromal tumors.

CT
MRI
Primary and metastatic GISTsSmall: Homogeneous mass; Large: Hypervascular, enhancing masses with heterogeneity due to hemorrhage, cystic degeneration, or necrosisDepend on the amount of hemorrhage, necrosis and cystic degeneration; solid tumor components with low T1 signal, intermediate-to-high T2 signal, and enhancement; low mean ADC values may predict high malignancy potential
Treatment responseHomogeneously hypoattenuating mass with decreased enhancing tumor nodules and intratumoral vesselsIncreased T2 signal, increased cystic degeneration of solid tumoral components, increased ADC values
Disease recurrenceDevelopment of enhancing tumor nodules within the treated hypoattenuating tumorNew peripheral thickening and enhancement of cystic tumor
MRI

MRI serves as an imaging adjunct, especially for young patients for whom repeated ionizing radiation exposure should be minimized, for evaluating liver metastases, and for evaluating rectal tumors. MRI has been found to be superior in characterizing treated liver metastases compared to CT, especially with identifying foci of hypervascularity[32]. Conversely, MRI is less helpful in identifying mesenteric lesions due to the lack of oral contrast and respiratory gating[32]. Generally, the recommended MRI sequences include T1-weighted in and out of phase axial, T2-weighted coronal turbo spin echo, T2-weighted fat-saturated axial respiratory triggered turbo spin echo, and T1-weighted fat-saturated 3D volumetric acquisition in noncontrast, early arterial, portal venous, and hepatic venous phases[32].

MRI features of GISTs vary depending on the amount of hemorrhage, necrosis and cystic degeneration. Solid tumor components demonstrate low T1 signal, intermediate-to-high T2 signal, and enhancement with contrast[24]. The presence of intratumoral cystic change with low apparent diffusion coefficient (ADC) values are predictors of high malignant potential[33]. A negative correlation between mean ADC values and malignancy risk of GISTs has been demonstrated[33]. Upon treatment response, GIST metastases demonstrate increased T2 signal with increased cystic degeneration of solid tumoral components and increased ADC values[34]. With disease recurrence, new peripheral thickening and enhancement of cystic metastases can be seen[24].

PET-CT

18F-fluorodeoxyglucose (18F-FDG) PET-CT can aid in staging, detecting early response to treatment, and detecting early recurrence of GIST[34]. PET-CT can be helpful in distinguishing tumors from benign tissue given the expected increased glucose metabolism of viable tumor cells. PET-CT is more sensitive than CT in detecting treatment response due to detecting decreased 18F-FDG uptake, which is typically observed before a change in tumor size[35]. Such changes can be detected 24 h to 1 mo after therapy initiation[36]. For patients on imatinib mesylate, increased 18F-FDG uptake may signify treatment resistance or lack of medication compliance.

MANAGEMENT AND SURVEILLANCE

Surgical resection is the mainstay of treatment, especially for small-to-medium sized GISTs without metastasis. Obtaining preoperative biopsy is controversial due to the risk of tumoral hemorrhage and seeding; therefore, postoperative pathology is required for diagnosis[1]. During resection, the tumor should be handled carefully to avoid bleeding, rupture, and peritoneal seeding. Ideally, the tumor resection should include an intact pseudocapsule and negative microscopic margins. Follow-up imaging intervals depend on the GIST’s risk group categorization: A very low-risk GIST is likely cured by surgery and does not require follow-up; a low-risk GIST may need annual CT or MRI follow-up for 5 years; an intermediate-risk GIST needs annual CT or MRI follow-up for 5 years with the first scan completed 6-8 mo after surgery; and a high-risk GIST should be followed every 6 mo for the first 5 years, then annually for the next 5 years[35].

Prior to 2000, cytotoxic chemotherapy had not been found to be clinically effective in the management of GISTs[1]. However, following the Food and Drug Administration approval of imatinib mesylate for treating metastatic and locally advanced KIT-positive GISTs in 2002, the management of GISTs has rapidly expanded. As previously stated, imatinib mesylate is a potent tyrosine kinase inhibitor that acts on enzymes including KIT, leukemia-specific BCR-ABL chimera, and PDGFRA. Imatinib mesylate can be utilized preoperatively to downsize the tumor and/or as adjuvant therapy to prevent recurrence. Preoperative imatinib mesylate can be utilized for large and poorly positioned GISTs that may be marginally resectable; imatinib mesylate has been shown to induce tumor cell apoptosis and decrease tumor glucose metabolism on PET-CT[37]. Postoperatively, 1-year of adjuvant imatinib mesylate has been shown to prolong overall survival, although the optimal duration of postoperative treatment is unclear[38]. For unresectable or metastatic GISTs, a phase II trial of imatinib mesylate therapy demonstrated 68% objective response rate regardless of imatinib dosage, and the median time to at least partial response was 2.7 mo[39]. The median survival of patients with metastatic GISTs improved significantly from 19 mo as reported by DeMatteo et al[40] in the pre-imatinib era to 73 mo with imatinib mesylate as reported by Menge et al[5]. If there is imaging evidence of disease progression despite using standard-dose imatinib mesylate, dose escalation of imatinib mesylate or utilization of sunitinib malate, a second line tyrosine kinase inhibitor, may be considered[1]. Sunitinib malate acts as a less specific tyrosine kinase inhibitor on KIT, PDGFR, vascular endothelial growth factor receptors, Fms-related tyrosine kinase 3, colony-stimulating factor-1R, and RET; as a result, sunitinib malate demonstrates activity against angiogenesis in addition to tumor activity related to receptor tyrosine kinase inhibition[1]. For imatinib and sunitinib-resistant GISTs, investigational therapeutic options include second generation tyrosine kinase inhibitors, such as sorafenib, dasatinib, and nilotinib[1]. Follow-up CT should be obtained within 3 mo of initiating imatinib mesylate with surveillance scans completed every 3 to 6 mo for unresectable or metastatic GISTs; the follow-up interval can be less frequent for low-risk GISTs[1].

With the advancement of these molecular-targeted therapies, multiple associated adverse effects have been demonstrated, and some of these may be identified on follow-up imaging. Fluid retention is commonly seen with imatinib mesylate and can manifest with pleural effusions, pericardial effusion, ascites, or extensive subcutaneous edema[27]. Imatinib mesylate can be associated with intratumoral hemorrhage, especially in patients with large bulky tumors[1,27]. Tyrosine kinase inhibitors are associated with pancreatic findings. For instance, imatinib mesylate is associated with asymptomatic pancreatic swelling; a ≥ 22% increase in pancreatic volume has been shown to be a poor prognostic indicator[41]. Conversely, sunitinib malate is associated with pancreatic atrophy, and this finding is associated with poor prognosis[42]. Moreover, there are several case reports of pancreatitis associated with sunitinib malate and sorafenib therapy[43]. It is important to identify these adverse effects on imaging, which would allow for dose reduction, dose interruption, or drug discontinuation in the appropriate setting.

CONCLUSION

GISTs are the most common mesenchymal tumors of the GI tract and often arise from the stomach or small intestine. The majority of GISTs occur as a result of activating mutations in two receptor protein tyrosine kinases, KIT and/or PDGFRA, leading to tumorigenesis. Mutational analyses allow for predicting patient prognosis and treatment response. Clinical presentations can vary from no symptoms to GI bleeding, abdominal discomfort, and ulcer-related symptoms. While most GISTs are benign, some cases can be aggressive with metastases. Imaging plays a key role in the diagnosis and follow-up of these tumors. It is crucial to understand and identify the key imaging features of GISTs and their expected appearance upon treatment response and disease recurrence. Surgical resection is the preferred management, especially for small tumors, while tyrosine kinase inhibitors, including imatinib mesylate and sunitinib malate, can serve as a neoadjuvant and/or adjuvant therapies.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Kurokawa R S-Editor: Wang JJ L-Editor: A P-Editor: Xing YX

References
1.  Demetri GD, von Mehren M, Antonescu CR, DeMatteo RP, Ganjoo KN, Maki RG, Pisters PW, Raut CP, Riedel RF, Schuetze S, Sundar HM, Trent JC, Wayne JD. NCCN Task Force report: update on the management of patients with gastrointestinal stromal tumors. J Natl Compr Canc Netw. 2010;8 Suppl 2:S1-41; quiz S42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 826]  [Cited by in F6Publishing: 778]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
2.  Foo WC, Liegl-Atzwanger B, Lazar AJ. Pathology of gastrointestinal stromal tumors. Clin Med Insights Pathol. 2012;5:23-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
3.  Nilsson B, Bümming P, Meis-Kindblom JM, Odén A, Dortok A, Gustavsson B, Sablinska K, Kindblom LG. Gastrointestinal stromal tumors: the incidence, prevalence, clinical course, and prognostication in the preimatinib mesylate era--a population-based study in western Sweden. Cancer. 2005;103:821-829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 803]  [Cited by in F6Publishing: 835]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
4.  Tryggvason G, Gíslason HG, Magnússon MK, Jónasson JG. Gastrointestinal stromal tumors in Iceland, 1990-2003: the icelandic GIST study, a population-based incidence and pathologic risk stratification study. Int J Cancer. 2005;117:289-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 242]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
5.  Menge F, Jakob J, Kasper B, Smakic A, Gaiser T, Hohenberger P. Clinical Presentation of Gastrointestinal Stromal Tumors. Visc Med. 2018;34:335-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 33]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
6.  Nowain A, Bhakta H, Pais S, Kanel G, Verma S. Gastrointestinal stromal tumors: clinical profile, pathogenesis, treatment strategies and prognosis. J Gastroenterol Hepatol. 2005;20:818-824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 103]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
7.  Corless CL, Heinrich MC. Molecular pathobiology of gastrointestinal stromal sarcomas. Annu Rev Pathol. 2008;3:557-586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 249]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
8.  Pink D, Schoeler D, Lindner T, Thuss-Patience PC, Kretzschmar A, Knipp H, Vanhoefer U, Reichardt P. Severe hypoglycemia caused by paraneoplastic production of IGF-II in patients with advanced gastrointestinal stromal tumors: a report of two cases. J Clin Oncol. 2005;23:6809-6811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
9.  Maynard MA, Marino-Enriquez A, Fletcher JA, Dorfman DM, Raut CP, Yassa L, Guo C, Wang Y, Dorfman C, Feldman HA, Frates MC, Song H, Jugo RH, Taguchi T, Hershman JM, Larsen PR, Huang SA. Thyroid hormone inactivation in gastrointestinal stromal tumors. N Engl J Med. 2014;370:1327-1334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 45]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
10.  Graadt van Roggen JF, van Velthuysen ML, Hogendoorn PC. The histopathological differential diagnosis of gastrointestinal stromal tumours. J Clin Pathol. 2001;54:96-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 171]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
11.  Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, Kawano K, Hanada M, Kurata A, Takeda M, Muhammad Tunio G, Matsuzawa Y, Kanakura Y, Shinomura Y, Kitamura Y. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998;279:577-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3215]  [Cited by in F6Publishing: 3030]  [Article Influence: 116.5]  [Reference Citation Analysis (0)]
12.  Zhao X, Yue C. Gastrointestinal stromal tumor. J Gastrointest Oncol. 2012;3:189-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 53]  [Reference Citation Analysis (0)]
13.  Heinrich MC, Corless CL, Duensing A, McGreevey L, Chen CJ, Joseph N, Singer S, Griffith DJ, Haley A, Town A, Demetri GD, Fletcher CD, Fletcher JA. PDGFRA activating mutations in gastrointestinal stromal tumors. Science. 2003;299:708-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1712]  [Cited by in F6Publishing: 1674]  [Article Influence: 79.7]  [Reference Citation Analysis (0)]
14.  Dagher R, Cohen M, Williams G, Rothmann M, Gobburu J, Robbie G, Rahman A, Chen G, Staten A, Griebel D, Pazdur R. Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clin Cancer Res. 2002;8:3034-3038.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  West RB, Corless CL, Chen X, Rubin BP, Subramanian S, Montgomery K, Zhu S, Ball CA, Nielsen TO, Patel R, Goldblum JR, Brown PO, Heinrich MC, van de Rijn M. The novel marker, DOG1, is expressed ubiquitously in gastrointestinal stromal tumors irrespective of KIT or PDGFRA mutation status. Am J Pathol. 2004;165:107-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 440]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
16.  Fletcher CD, Berman JJ, Corless C, Gorstein F, Lasota J, Longley BJ, Miettinen M, O'Leary TJ, Remotti H, Rubin BP, Shmookler B, Sobin LH, Weiss SW. Diagnosis of gastrointestinal stromal tumors: A consensus approach. Hum Pathol. 2002;33:459-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2231]  [Cited by in F6Publishing: 2102]  [Article Influence: 95.5]  [Reference Citation Analysis (1)]
17.  Kindblom LG, Remotti HE, Aldenborg F, Meis-Kindblom JM. Gastrointestinal pacemaker cell tumor (GIPACT): gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cells of Cajal. Am J Pathol. 1998;152:1259-1269.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Seidal T, Edvardsson H. Expression of c-kit (CD117) and Ki67 provides information about the possible cell of origin and clinical course of gastrointestinal stromal tumours. Histopathology. 1999;34:416-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 85]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
19.  Marrari A, Wagner AJ, Hornick JL. Predictors of response to targeted therapies for gastrointestinal stromal tumors. Arch Pathol Lab Med. 2012;136:483-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
20.  Dematteo RP, Gold JS, Saran L, Gönen M, Liau KH, Maki RG, Singer S, Besmer P, Brennan MF, Antonescu CR. Tumor mitotic rate, size, and location independently predict recurrence after resection of primary gastrointestinal stromal tumor (GIST). Cancer. 2008;112:608-615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 317]  [Cited by in F6Publishing: 339]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
21.  Miranda C, Nucifora M, Molinari F, Conca E, Anania MC, Bordoni A, Saletti P, Mazzucchelli L, Pilotti S, Pierotti MA, Tamborini E, Greco A, Frattini M. KRAS and BRAF mutations predict primary resistance to imatinib in gastrointestinal stromal tumors. Clin Cancer Res. 2012;18:1769-1776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 133]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
22.  Miettinen M, Lasota J, Sobin LH. Gastrointestinal stromal tumors of the stomach in children and young adults: a clinicopathologic, immunohistochemical, and molecular genetic study of 44 cases with long-term follow-up and review of the literature. Am J Surg Pathol. 2005;29:1373-1381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 183]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
23.  Agaram NP, Laquaglia MP, Ustun B, Guo T, Wong GC, Socci ND, Maki RG, DeMatteo RP, Besmer P, Antonescu CR. Molecular characterization of pediatric gastrointestinal stromal tumors. Clin Cancer Res. 2008;14:3204-3215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 208]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
24.  Milliron B, Mittal PK, Camacho JC, Datir A, Moreno CC. Gastrointestinal Stromal Tumors: Imaging Features Before and After Treatment. Curr Probl Diagn Radiol. 2017;46:17-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
25.  Wronski M, Cebulski W, Slodkowski M, Krasnodebski IW. Gastrointestinal stromal tumors: ultrasonographic spectrum of the disease. J Ultrasound Med. 2009;28:941-948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
26.  Kang HC, Menias CO, Gaballah AH, Shroff S, Taggart MW, Garg N, Elsayes KM. Beyond the GIST: mesenchymal tumors of the stomach. Radiographics. 2013;33:1673-1690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 92]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
27.  Hong X, Choi H, Loyer EM, Benjamin RS, Trent JC, Charnsangavej C. Gastrointestinal stromal tumor: role of CT in diagnosis and in response evaluation and surveillance after treatment with imatinib. Radiographics. 2006;26:481-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 132]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
28.  Chourmouzi D, Sinakos E, Papalavrentios L, Akriviadis E, Drevelegas A. Gastrointestinal stromal tumors: a pictorial review. J Gastrointestin Liver Dis. 2009;18:379-383.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Chen MY, Bechtold RE, Savage PD. Cystic changes in hepatic metastases from gastrointestinal stromal tumors (GISTs) treated with Gleevec (imatinib mesylate). AJR Am J Roentgenol. 2002;179:1059-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 92]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
30.  Benjamin RS, Choi H, Macapinlac HA, Burgess MA, Patel SR, Chen LL, Podoloff DA, Charnsangavej C. We should desist using RECIST, at least in GIST. J Clin Oncol. 2007;25:1760-1764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 404]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
31.  Choi H, Charnsangavej C, Faria SC, Macapinlac HA, Burgess MA, Patel SR, Chen LL, Podoloff DA, Benjamin RS. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J Clin Oncol. 2007;25:1753-1759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1094]  [Cited by in F6Publishing: 1073]  [Article Influence: 63.1]  [Reference Citation Analysis (0)]
32.  Sandrasegaran K, Rajesh A, Rushing DA, Rydberg J, Akisik FM, Henley JD. Gastrointestinal stromal tumors: CT and MRI findings. Eur Radiol. 2005;15:1407-1414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 158]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
33.  Yu MH, Lee JM, Baek JH, Han JK, Choi BI. MRI features of gastrointestinal stromal tumors. AJR Am J Roentgenol. 2014;203:980-991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
34.  Tirumani SH, Jagannathan JP, Krajewski KM, Shinagare AB, Jacene H, Ramaiya NH. Imatinib and beyond in gastrointestinal stromal tumors: A radiologist's perspective. AJR Am J Roentgenol. 2013;201:801-810.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
35.  Joensuu H, Martin-Broto J, Nishida T, Reichardt P, Schöffski P, Maki RG. Follow-up strategies for patients with gastrointestinal stromal tumour treated with or without adjuvant imatinib after surgery. Eur J Cancer. 2015;51:1611-1617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 57]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
36.  Van den Abbeele AD. The lessons of GIST--PET and PET/CT: a new paradigm for imaging. Oncologist. 2008;13 Suppl 2:8-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 193]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
37.  McAuliffe JC, Hunt KK, Lazar AJ, Choi H, Qiao W, Thall P, Pollock RE, Benjamin RS, Trent JC. A randomized, phase II study of preoperative plus postoperative imatinib in GIST: evidence of rapid radiographic response and temporal induction of tumor cell apoptosis. Ann Surg Oncol. 2009;16:910-919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 141]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
38.  DeMatteo RP, Ballman KV, Antonescu CR, Corless C, Kolesnikova V, von Mehren M, McCarter MD, Norton J, Maki RG, Pisters PW, Demetri GD, Brennan MF, Owzar K; American College of Surgeons Oncology Group (ACOSOG) Intergroup Adjuvant GIST Study Team for the Alliance for Clinical Trials in Oncology. Long-term results of adjuvant imatinib mesylate in localized, high-risk, primary gastrointestinal stromal tumor: ACOSOG Z9000 (Alliance) intergroup phase 2 trial. Ann Surg. 2013;258:422-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 119]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
39.  Blanke CD, Demetri GD, von Mehren M, Heinrich MC, Eisenberg B, Fletcher JA, Corless CL, Fletcher CD, Roberts PJ, Heinz D, Wehre E, Nikolova Z, Joensuu H. Long-term results from a randomized phase II trial of standard- versus higher-dose imatinib mesylate for patients with unresectable or metastatic gastrointestinal stromal tumors expressing KIT. J Clin Oncol. 2008;26:620-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 716]  [Cited by in F6Publishing: 721]  [Article Influence: 45.1]  [Reference Citation Analysis (0)]
40.  DeMatteo RP, Lewis JJ, Leung D, Mudan SS, Woodruff JM, Brennan MF. Two hundred gastrointestinal stromal tumors: recurrence patterns and prognostic factors for survival. Ann Surg. 2000;231:51-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1797]  [Cited by in F6Publishing: 1636]  [Article Influence: 68.2]  [Reference Citation Analysis (0)]
41.  Kurokawa R, Hagiwara A, Amemiya S, Gonoi W, Fujita N, Kurokawa M, Yamaguchi H, Nakai Y, Ota Y, Baba A, Kawahara T, Abe O. Imatinib-induced pancreatic hypertrophy in patients with gastrointestinal stromal tumor: Association with overall survival. Pancreatology. 2021;21:246-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
42.  Shinagare AB, Steele E, Braschi-Amirfarzan M, Tirumani SH, Ramaiya NH. Sunitinib-associated Pancreatic Atrophy in Patients with Gastrointestinal Stromal Tumor: A Toxicity with Prognostic Implications Detected at Imaging. Radiology. 2016;281:140-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
43.  Wolfe D, Kanji S, Yazdi F, Barbeau P, Rice D, Beck A, Butler C, Esmaeilisaraji L, Skidmore B, Moher D, Hutton B. Drug induced pancreatitis: A systematic review of case reports to determine potential drug associations. PLoS One. 2020;15:e0231883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 57]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]