[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (19)

Search Parameters:
Keywords = wild type GIST

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 5944 KiB  
Review
Gastrointestinal Stromal Tumors (GISTs) in Pediatric Patients: A Case Report and Literature Review
by Tudor-Alexandru Popoiu, Cãtãlin-Alexandru Pîrvu, Cãlin-Marius Popoiu, Emil Radu Iacob, Tamas Talpai, Amalia Voinea, Rãzvan-Sorin Albu, Sorina Tãban, Larisa-Mihaela Bãlãnoiu and Stelian Pantea
Children 2024, 11(9), 1040; https://doi.org/10.3390/children11091040 - 26 Aug 2024
Cited by 1 | Viewed by 1769
Abstract
Gastrointestinal stromal tumors (GISTs) are rare mesenchymal neoplasms that primarily affect adults, with pediatric cases constituting only 0.5–2.7% of the total. Pediatric GISTs present unique clinical, genetic, and pathological features that distinguish them from adult cases. This literature review aims to elucidate these [...] Read more.
Gastrointestinal stromal tumors (GISTs) are rare mesenchymal neoplasms that primarily affect adults, with pediatric cases constituting only 0.5–2.7% of the total. Pediatric GISTs present unique clinical, genetic, and pathological features that distinguish them from adult cases. This literature review aims to elucidate these differences, emphasizing diagnostic and therapeutic challenges. We discuss the resistance of pediatric GISTs to conventional chemotherapy and highlight the importance of surgical intervention, especially in emergency situations involving intra-abdominal bleeding. The review also explores the molecular characteristics of pediatric GISTs, including rare mutations such as quadruple-negative wild-type GIST with an FGF3 gene gain mutation. To illustrate these points, we conclude with a case from our clinic involving a 15-year-old female with multiple CD117-positive gastric GISTs and a quadruple-negative wild-type genetic profile who required urgent surgical intervention following a failed tumor embolization. This case underscores the critical need for early diagnosis and individualized therapeutic strategies combining oncologic and surgical care to improve outcomes in pediatric GIST patients. Full article
(This article belongs to the Special Issue Cutting-Edge Laparoscopic and Thoracoscopic Surgery in Children)
Show Figures

Figure 1

Figure 1
<p>Molecular characteristics of adult and pediatric GISTs.</p>
Full article ">Figure 2
<p>SDH immunohistochemistry—normal distribution of SDH in the bioptic tumor tissue.</p>
Full article ">Figure 3
<p>CD117-positive biopsy.</p>
Full article ">Figure 4
<p>Vascular invasion—hematoxiline–eozine 20×.</p>
Full article ">Figure 5
<p>Gastric tumor-GIST seen in CT scan before biopsy was obtained. In yellow you can see a large tumor mass. The blue arrow points to ascites present in the abdomen.</p>
Full article ">Figure 6
<p>A large peritoneal tumor mass (outlined in yellow) located in the left upper hemiabdomen, with a solid component that absorbs iodine located distally and a liquid component. The liver (outlined in green) shows the diffuse pathological uptake of the contrast substance at the periphery.</p>
Full article ">Figure 7
<p>MRI T1 sequence of the abdomen, coronal section in the venous phase: Secondary hepatic lesion with gadolinium uptake, subcentimetric (blue arrow). Tumor mass (green arrows) with a solid component showing gadolinium uptake (yellow arrows) and a liquid component.</p>
Full article ">
16 pages, 4605 KiB  
Article
Molecular Profiling of KIT/PDGFRA-Mutant and Wild-Type Gastrointestinal Stromal Tumors (GISTs) with Clinicopathological Correlation: An 18-Year Experience at a Tertiary Center in Kuwait
by Rola H. Ali, Ahmad R. Alsaber, Asit K. Mohanty, Abdulsalam Alnajjar, Eiman M. A. Mohammed, Mona Alateeqi, Hiba Jama, Ammar Almarzooq, Noelle Benobaid, Zainab Alqallaf, Amir A. Ahmed, Shakir Bahzad and Mohammad Alkandari
Cancers 2024, 16(16), 2907; https://doi.org/10.3390/cancers16162907 - 21 Aug 2024
Viewed by 1128
Abstract
In gastrointestinal stromal tumors (GISTs), identifying prototypical mutations in the KIT/PDGFRA oncogenes, or in rare alternate genes, is essential for prognostication and predicting response to tyrosine kinase inhibitors. Conversely, wild-type GISTs (WT-GIST), which lack known mutations, have limited treatment options. Data on the [...] Read more.
In gastrointestinal stromal tumors (GISTs), identifying prototypical mutations in the KIT/PDGFRA oncogenes, or in rare alternate genes, is essential for prognostication and predicting response to tyrosine kinase inhibitors. Conversely, wild-type GISTs (WT-GIST), which lack known mutations, have limited treatment options. Data on the mutational landscape of GISTs and their impact on disease progression are very limited in Kuwait. Using a targeted next-generation sequencing panel, we investigated the spectrum and frequency of KIT, PDGFRA, and RAS-pathway-related mutations in 95 out of 200 GISTs diagnosed at Kuwait Cancer Center from 2005 to 2023 and assessed their correlation with clinicopathological parameters. Among the 200 tumors (median age 55 years; 15–91), 54% originated in the stomach, 33% in the small bowel, 7% in the colorectum, 1.5% in the peritoneum, and 4.5% had an unknown primary site. Of the 95 molecularly profiled cases, 88% had a mutation: KIT (61%), PDGFRA (25%), NF1 (2%), and one NTRK1 rearrangement. Ten WT-GISTs were identified (stomach = 6, small bowel = 2, and colorectum = 2). WT-GISTs tended to be smaller (median 4.0 cm; 0.5–8.0) (p = 0.018), with mitosis ≤5/5 mm2, and were of lower risk (p = 0.019). KIT mutations were an adverse indicator of disease progression (p = 0.049), while wild-type status did not significantly impact progression (p = 0.934). The genetic landscape in this cohort mirrors that of global studies, but regional collaborations are needed to correlate outcomes with genetic variants. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

Figure 1
<p>Frequencies of <span class="html-italic">KIT</span> and <span class="html-italic">PDGFRA</span> mutations (<span class="html-italic">n</span> = 95). <span class="html-italic">KIT</span> exon 11 mutations are heterogeneous, with W557_K558del being the most common. <span class="html-italic">PDGFRA</span> exon 18 and <span class="html-italic">KIT</span> exon 9 show a predominance of one variant each: D842V and A502_Y503dup, respectively. TK1 = Tyrosine kinase domain 1; TK2 = Tyrosine kinase domain 2.</p>
Full article ">Figure 2
<p>Genomic and amino-acid sequences of <span class="html-italic">KIT</span> exon 11 mutations. (<b>A</b>) Codon positions. (<b>B</b>) Frequency of codons involved in mutation. * <span class="html-italic">KIT</span> 4q12 locus.</p>
Full article ">Figure 3
<p>Distribution of molecular alterations based on gastrointestinal locations (<span class="html-italic">n</span> = 95). * <span class="html-italic">NTRK</span>-fused spindle cell neoplasms are currently classified as a separate entity.</p>
Full article ">Figure 4
<p>Graphical summary of molecular and clinicopathological findings (<span class="html-italic">n</span> = 95), with each row representing an individual patient. * Metastatic at diagnosis; ** Tyrosine kinase inhibitors, in adjuvant and/or metastatic setting; *** <span class="html-italic">PDGFRA</span>-specific TKIs not available; PFS = progression-free survival; IHC = immunohistochemistry; NA = not available.</p>
Full article ">Figure 5
<p>Histopathology of <span class="html-italic">KIT/PDGFRA</span> wild-type GISTs with corresponding KIT immunostaining. (<b>A</b>,<b>D</b>) <span class="html-italic">NF1</span>-mutant in the duodenum with spindle cell morphology and diffuse KIT expression in a known neurofibromatosis type 1 patient. (<b>B</b>,<b>E</b>) <span class="html-italic">KIT/PDGFRA/RAS</span> wild-type in the colon with spindle cell morphology and diffuse KIT expression. (<b>C</b>,<b>F</b>) <span class="html-italic">KIT/PDGFRA/RAS</span> wild-type in the stomach with epithelioid morphology and faint KIT expression. All are at 20× magnification.</p>
Full article ">
16 pages, 1764 KiB  
Article
Utility of Clinical Next Generation Sequencing Tests in KIT/PDGFRA/SDH Wild-Type Gastrointestinal Stromal Tumors
by Ryan A. Denu, Cissimol P. Joseph, Elizabeth S. Urquiola, Precious S. Byrd, Richard K. Yang, Ravin Ratan, Maria Alejandra Zarzour, Anthony P. Conley, Dejka M. Araujo, Vinod Ravi, Elise F. Nassif Haddad, Michael S. Nakazawa, Shreyaskumar Patel, Wei-Lien Wang, Alexander J. Lazar and Neeta Somaiah
Cancers 2024, 16(9), 1707; https://doi.org/10.3390/cancers16091707 - 27 Apr 2024
Cited by 2 | Viewed by 2860
Abstract
Objective: The vast majority of gastrointestinal stromal tumors (GISTs) are driven by activating mutations in KIT, PDGFRA, or components of the succinate dehydrogenase (SDH) complex (SDHA, SDHB, SDHC, and SDHD genes). A small fraction of GISTs lack [...] Read more.
Objective: The vast majority of gastrointestinal stromal tumors (GISTs) are driven by activating mutations in KIT, PDGFRA, or components of the succinate dehydrogenase (SDH) complex (SDHA, SDHB, SDHC, and SDHD genes). A small fraction of GISTs lack alterations in KIT, PDGFRA, and SDH. We aimed to further characterize the clinical and genomic characteristics of these so-called “triple-negative” GISTs. Methods: We extracted clinical and genomic data from patients seen at MD Anderson Cancer Center with a diagnosis of GIST and available clinical next generation sequencing data to identify “triple-negative” patients. Results: Of the 20 patients identified, 11 (55.0%) had gastric, 8 (40.0%) had small intestinal, and 1 (5.0%) had rectal primary sites. In total, 18 patients (90.0%) eventually developed recurrent or metastatic disease, and 8 of these presented with de novo metastatic disease. For the 13 patients with evaluable response to imatinib (e.g., neoadjuvant treatment or for recurrent/metastatic disease), the median PFS with imatinib was 4.4 months (range 0.5–191.8 months). Outcomes varied widely, as some patients rapidly developed progressive disease while others had more indolent disease. Regarding potential genomic drivers, four patients were found to have alterations in the RAS/RAF/MAPK pathway: two with a BRAF V600E mutation and two with NF1 loss-of-function (LOF) mutations (one deletion and one splice site mutation). In addition, we identified two with TP53 LOF mutations, one with NTRK3 fusion (ETV6-NTRK3), one with PTEN deletion, one with FGFR1 gain-of-function (GOF) mutation (K654E), one with CHEK2 LOF mutation (T367fs*), one with Aurora kinase A fusion (AURKA-CSTF1), and one with FANCA deletion. Patients had better responses with molecularly targeted therapies than with imatinib. Conclusions: Triple-negative GISTs comprise a diverse cohort with different driver mutations. Compared to KIT/PDGFRA-mutant GIST, limited benefit was observed with imatinib in triple-negative GIST. In depth molecular profiling can be helpful in identifying driver mutations and guiding therapy. Full article
Show Figures

Figure 1

Figure 1
<p>Clinical characteristics and outcomes in triple-negative GIST. (<b>A</b>) Distribution of age at diagnosis. (<b>B</b>) Distribution of tumor size at diagnosis. (<b>C</b>) Distribution of mitotic count from original biopsy or resected specimen. In (<b>A</b>–<b>C</b>), bars represent means ± SD. (<b>D</b>) Anatomic distribution of triple-negative GIST cases. (<b>E</b>) On the left in colored bars are the initial disease stage. Black and gray bars on the right indicate the percent of patients that remained with localized disease versus those that developed recurrent or metastatic disease. Bars represent percentages plus standard error of proportion.</p>
Full article ">Figure 2
<p>Genomics of triple-negative GIST. (<b>A</b>) Map of the genomic alterations in triple-negative GIST patients. Each row represents a patient. Each column represents a clinical feature (left 3 columns) or gene, as indicated. White boxes indicate that the gene was profiled but that no alteration was found, and gray boxes indicate that the gene was not profiled. (<b>B</b>) Number of somatic mutations detected by clinical sequencing assays. Each dot represents a single tumor, and bars represent mean ± SD. (<b>C</b>) Distribution of the most commonly altered genes in the cohort. (<b>D</b>) Percentage of tumors with each hypothesized driver mutation. In (<b>C</b>,<b>D</b>), percentages plus standard error of proportion are plotted.</p>
Full article ">Figure 3
<p>Response to treatment in triple-negative GIST. (<b>A</b>) Progression-free survival while on imatinib (n = 15 patients) versus molecularly matched treatments (n = 3 patients). <span class="html-italic">p</span> = 0.21. (<b>B</b>) Swimmer’s plot showing timeline of indicated therapies. Purported driver mutation is shown in the column on the left.</p>
Full article ">Figure 4
<p>Survival outcomes in triple-negative GIST. Overall survival (<b>A</b>), recurrence-free survival (<b>B</b>), and progression-free survival (<b>C</b>) of patients with triple- negative GIST. Recurrence-free survival was calculated in patients with initially localized disease from the date of histologic diagnosis to the date of recurrence, death, or the latest follow-up. Progression-free survival was calculated from the start of therapy to the date of recurrence, death, or the latest follow-up.</p>
Full article ">
13 pages, 8120 KiB  
Case Report
ETV6::NTRK3 Fusion-Positive Wild-Type Gastrointestinal Stromal Tumor (GIST) with Abundant Lymphoid Infiltration (TILs and Tertiary Lymphoid Structures): A Report on a New Case with Therapeutic Implications and a Literature Review
by Isidro Machado, Reyes Claramunt-Alonso, Javier Lavernia, Ignacio Romero, María Barrios, María José Safont, Nuria Santonja, Lara Navarro, José Antonio López-Guerrero and Antonio Llombart-Bosch
Int. J. Mol. Sci. 2024, 25(7), 3707; https://doi.org/10.3390/ijms25073707 - 26 Mar 2024
Cited by 3 | Viewed by 1666
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract, with proto-oncogene, receptor tyrosine kinase (c-kit), or PDGFRα mutations detected in around 85% of cases. GISTs without c-kit or platelet-derived growth factor receptor alpha (PDGFRα) [...] Read more.
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract, with proto-oncogene, receptor tyrosine kinase (c-kit), or PDGFRα mutations detected in around 85% of cases. GISTs without c-kit or platelet-derived growth factor receptor alpha (PDGFRα) mutations are considered wild-type (WT), and their diverse molecular alterations and biological behaviors remain uncertain. They are usually not sensitive to tyrosine kinase inhibitors (TKIs). Recently, some molecular alterations, including neurotrophic tyrosine receptor kinase (NTRK) fusions, have been reported in very few cases of WT GISTs. This novel finding opens the window for the use of tropomyosin receptor kinase (TRK) inhibitor therapy in these subtypes of GIST. Herein, we report a new case of NTRK-fused WT high-risk GIST in a female patient with a large pelvic mass (large dimension of 20 cm). The tumor was removed, and the histopathology displayed spindle-predominant morphology with focal epithelioid areas, myxoid stromal tissue, and notable lymphoid infiltration with tertiary lymphoid structures. Ten mitoses were quantified in 50 high-power fields without nuclear pleomorphism. DOG1 showed strong and diffuse positivity, and CD117 showed moderate positivity. Succinate dehydrogenase subunit B (SDHB) was retained, Pan-TRK was focal positive (nuclear pattern), and the proliferation index Ki-67 was 7%. Next-generation sequencing (NGS) detected an ETV6::NTRK3 fusion, and this finding was confirmed by fluorescence in situ hybridization (FISH), which showed NTRK3 rearrangement. In addition, an RB1 mutation was found by NGS. The follow-up CT scan revealed peritoneal nodules suggestive of peritoneal dissemination, and Entrectinib (a TRK inhibitor) was administered. After 3 months of follow-up, a new CT scan showed a complete response. Based on our results and the cases from the literature, GISTs with NTRK fusions are very uncommon so far; hence, further screening studies, including more WT GIST cases, may increase the possibility of finding additional cases. The present case may offer new insights into the potential introduction of TRK inhibitors as treatments for GISTs with NTRK fusions. Additionally, the presence of abundant lymphoid infiltration in the present case may prompt further research into immunotherapy as a possible additional therapeutic option. Full article
Show Figures

Figure 1

Figure 1
<p>Genomic structures of neurotrophic tyrosine receptor kinase (<span class="html-italic">NTRK3</span>) (<b>A</b>) and ETS Variant Transcription Factor 6 (<span class="html-italic">ETV6)</span> (<b>B</b>) are shown, illustrating exons encoding the canonical isoforms as described in the Genome Browser v461 software. The regions of the corresponding mRNAs encoding functional domains are marked. <span class="html-italic">NTRK3</span> stands for Neurotrophic Tyrosine Kinase Receptor Type 3 (NCBI Gene ID: 4916), and <span class="html-italic">ETV6</span> stands for ETS Variant Transcription Factor 6 (NCBI Gene ID: 2120).</p>
Full article ">Figure 2
<p>Chromosome 15 diagram, ISCN 2009, and localization of LSP neurotrophic tyrosine receptor kinase <span class="html-italic">NTRK3</span> 5′ (Green) and LSP <span class="html-italic">NTRK3</span> 3′ (Red) FISH probes on 15q25.3 chromosome position.</p>
Full article ">Figure 3
<p>(<b>A</b>) Coronal and (<b>B</b>) axial. Computerized tomography displays a large intra-abdominal and pelvic tumor with necrosis attached to the small bowel. (<b>C</b>) Peritoneal carcinomatosis (arrow) in CT scan of follow-up. (<b>D</b>) CT scan after 3 months with Entrectinic treatment, showing a complete response.</p>
Full article ">Figure 4
<p>(<b>A</b>–<b>D</b>) Microscopic examination with hematoxylin and eosin (H&amp;E) displays a mesenchymal neoplasm with spindle-predominant morphology, with focal epithelioid shape, ill-defined eosinophilic cytoplasm, myxoid stromal tissue, and remarkable lymphoid infiltration with tumor-infiltrating lymphocytes (TILs) and focal tertiary lymphoid structures, H&amp;E. (<b>A</b>) 40×, (<b>B</b>) 400×, and (<b>C</b>,<b>D</b>) 200×.</p>
Full article ">Figure 5
<p>(<b>A</b>) Diffuse and moderate CD117 cytoplasmic immunoreactivity, 100×. (<b>B</b>) Strong and diffuse DOG1 cytoplasmic positivity, 200×. (<b>C</b>) Patchy CD34 positivity, 100×. (<b>D</b>,<b>E</b>) Focal and nuclear Pan-TRK immunoreactivity, 400×.</p>
Full article ">Figure 6
<p>(<b>A</b>) CD8 positivity in tumor-infiltrating lymphocytes (TILs), 100×. (<b>B</b>) CD20 immunoreactivity in B-cells from tertiary lymphoid structures, 200×. (<b>C</b>) CD138 positivity in plasma cells, 100×. (<b>D</b>) CD163 positivity in the myeloid/histiocyte population, 100×.</p>
Full article ">Figure 7
<p>(<b>A</b>) Representation of the fusion gene (<span class="html-italic">ETV6::NTRK3</span>) obtained by next-generation sequencing (NGS), showing the exons and domains involved in the resultant fusion gene. HLH: Helix-loop-helix domain, TK: tyrosine kinase domain. (<b>B</b>) Integrative Genomics Viewer (IGV) displaying the <span class="html-italic">ETV6::NTRK3</span> fusion gene.</p>
Full article ">Figure 8
<p>Neurotrophic tyrosine receptor kinase type 3(<span class="html-italic">NTRK3)</span> FISH analysis with a break-apart probe. (<b>A</b>) Positive nuclei for the rearrangement of the <span class="html-italic">NTRK3</span> gene, indicated by white arrows. The positive signal pattern corresponds to an atypical pattern with an extra <span class="html-italic">NTRK3</span> 3′ signal (red). Additionally, there are abnormal signal patterns with only one red signal and one normal nucleus with overlapping signals (<span class="html-italic">NTRK3</span> 5′ green and <span class="html-italic">NTRK3</span> 3′ red). (<b>B</b>) Two positive nuclei, indicated by white arrows, and one nucleus with a normal signal pattern. The positive nuclei present a typical positive signal pattern with separated <span class="html-italic">NTRK3</span> 3′ (red) and <span class="html-italic">NTRK3</span> 5′ (green) signals.</p>
Full article ">
7 pages, 2398 KiB  
Case Report
Familial Gastrointestinal Stromal Tumor Associated with Zebra-like Pigmentation
by Takuma Hayashi and Ikuo Konishi
Biomedicines 2023, 11(6), 1590; https://doi.org/10.3390/biomedicines11061590 - 30 May 2023
Viewed by 1638
Abstract
Purpose: According to clinical studies, gastrointestinal stromal tumors (GISTs) are predominantly sporadic. GISTs associated with familial syndromes are very rare, and most patients exhibit wild-type KIT and platelet-derived growth factor alpha (PDGFRA). To date, GISTs associated with germline KIT pathogenic variants have been [...] Read more.
Purpose: According to clinical studies, gastrointestinal stromal tumors (GISTs) are predominantly sporadic. GISTs associated with familial syndromes are very rare, and most patients exhibit wild-type KIT and platelet-derived growth factor alpha (PDGFRA). To date, GISTs associated with germline KIT pathogenic variants have been observed in only 30 kindreds worldwide. The efficacy of imatinib, a multityrosine kinase inhibitor, in patients with GIST presenting germline KIT variants has been poorly reported, and the efficacy in clinical trials of treatments with tyrosine kinase inhibitors remains unclear. Therefore, imatinib is not yet recommended for treating GIST patients with germline KIT variants. Experimental Design: We performed cancer genomic testing on samples from a 32-year-old male patient with advanced GISTs throughout the upper stomach and cutaneous hyperpigmentation to determine diagnosis and treatment strategies. Results: We detected a germline W557R pathogenic variant of KIT. The patient was diagnosed with familial multinodular GIST based on the clinical findings and familial history of malignant tumors. Treatment with imatinib resulted in long-term regression of GISTs. Conclusions: Pathogenic variants detected by cancer genome testing can be used to diagnose malignant tumors and select new therapeutic agents for patients with advanced malignancies. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

Figure 1
<p>Pathological and contrast-enhanced computed tomography (CT) analyses. (<b>A</b>) Results of the pathological examination. Magnification: 20×. Scale bar = 100 μm. (<b>B</b>). Contrast-enhanced CT revealed a submucosal tumor in the upper stomach, a tumor in the liver, and multiple tumors in the abdominal cavity. Liver metastases of gastrointestinal stromal tumors (GISTs) are indicated by red dotted lines.</p>
Full article ">Figure 2
<p>Picture of the patient’s hands before treatment with imatinib mesylate. The back of both hands exhibited zebra-like pigmentation.</p>
Full article ">
13 pages, 1158 KiB  
Review
SDHA Germline Mutations in SDH-Deficient GISTs: A Current Update
by Angela Schipani, Margherita Nannini, Annalisa Astolfi and Maria A. Pantaleo
Genes 2023, 14(3), 646; https://doi.org/10.3390/genes14030646 - 4 Mar 2023
Cited by 7 | Viewed by 3711
Abstract
Loss of function of the succinate dehydrogenase complex characterizes 20–40% of all KIT/PDGFRA-negative GIST. Approximately half of SDH-deficient GIST patients lack SDHx mutations and are caused by a hypermethylation of the SDHC promoter, which causes the repression of SDHC transcription and depletion [...] Read more.
Loss of function of the succinate dehydrogenase complex characterizes 20–40% of all KIT/PDGFRA-negative GIST. Approximately half of SDH-deficient GIST patients lack SDHx mutations and are caused by a hypermethylation of the SDHC promoter, which causes the repression of SDHC transcription and depletion of SDHC protein levels through a mechanism described as epimutation. The remaining 50% of SDH-deficient GISTs have mutations in one of the SDH subunits and SDHA mutations are the most common (30%), with consequent loss of SDHA and SDHB protein expression immunohistochemically. SDHB, SDHC, and SDHD mutations in GIST occur in only 20–30% of cases and most of these SDH mutations are germline. More recently, germline mutations in SDHA have also been described in several patients with loss of function of the SDH complex. SDHA-mutant patients usually carry two mutational events at the SDHA locus, either the loss of the wild type allele or a second somatic event in compound heterozygosis. This review provides an overview of all data in the literature regarding SDHA-mutated GIST, especially focusing on the prevalence of germline mutations in SDH-deficient GIST populations who harbor SDHA somatic mutations, and offers a view towards understanding the importance of genetic counselling for SDHA-variant carriers and relatives. Full article
(This article belongs to the Special Issue Genetic Predispositions to Tumors of the Digestive System)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the SDH complex in physiological (<b>a</b>) and pathological (<b>b</b>) condition. SDH, succinate dehydrogenase; Q,ubiquinone; QH2,Ubiquinol; Fe–S, iron-sulfur; FAD, flavin adenine dinucleotide; FADH2, dihydroflavine-adenine dinucleotide; e, electron; TET, ten–eleven translocation; Me, methyl group; PHD, prolyl-hydroxylase domain proteins; Ub, ubiquitin; HIF, hypoxia-inducible factor.</p>
Full article ">Figure 2
<p>Distribution of reported <span class="html-italic">SDHA</span> gene mutations in GIST. Mutations are annotated at the cDNA (<b>a</b>) and protein level (<b>b</b>).</p>
Full article ">
11 pages, 1917 KiB  
Article
Small Gastric Stromal Tumors: An Underestimated Risk
by Jintao Guo, Qichao Ge, Fan Yang, Sheng Wang, Nan Ge, Xiang Liu, Jing Shi, Pietro Fusaroli, Yang Liu and Siyu Sun
Cancers 2022, 14(23), 6008; https://doi.org/10.3390/cancers14236008 - 6 Dec 2022
Cited by 5 | Viewed by 2078
Abstract
Background and Objectives: Small gastrointestinal stromal tumors (GISTs) are defined as tumors less than 2 cm in diameter, which are often found incidentally during gastroscopy. There is controversy regarding the management of small GISTs, and a certain percentage of small GISTs become malignant [...] Read more.
Background and Objectives: Small gastrointestinal stromal tumors (GISTs) are defined as tumors less than 2 cm in diameter, which are often found incidentally during gastroscopy. There is controversy regarding the management of small GISTs, and a certain percentage of small GISTs become malignant during follow-up. Previous studies which used Sanger targeted sequencing have shown that the mutation rate of small GISTs is significantly lower than that of large tumors. The aim of this study was to investigate the overall mutational profile of small GISTs, including those of wild-type tumors, using whole-exome sequencing (WES) and Sanger sequencing. Methods: Thirty-six paired small GIST specimens, which were resected by endoscopy, were analyzed by WES. Somatic mutations identified by WES were confirmed by Sanger sequencing. Sanger sequencing was performed in an additional 38 small gastric stromal tumor samples for examining hotspot mutations in KIT, PDGFRA, and BRAF. Results: Somatic C-KIT/PDGFRA mutations accounted for 81% of the mutations, including three novel mutation sites in C-KIT at exon 11, across the entire small gastric stromal tumor cohort (n = 74). In addition, 15% of small GISTs harbored previously undescribed BRAF-V600E hotspot mutations. No significant correlation was observed among the genotype, pathological features, and clinical classification. Conclusions: Our data revealed a high overall mutation rate (~96%) in small GISTs, indicating that genetic alterations are common events in early GIST generation. We also identified a high frequency of oncogenic BRAF-V600E mutations (15%) in small GISTs, which has not been previously reported. Full article
(This article belongs to the Topic Soft Tissue Sarcomas: Treatment and Management)
Show Figures

Figure 1

Figure 1
<p>Flow chart of the selection procedure.</p>
Full article ">Figure 2
<p>High frequency of oncogenic mutations in 36 small gastrointestinal stromal tumor (GIST) samples identified by whole-exome sequencing (WES). (<b>a</b>) Selected driver genes, by comparing somatic mutations and known driver genes in the database; (<b>b</b>) mutation distribution in the KIT molecular structure diagram, with novel mutations marked.</p>
Full article ">Figure 3
<p>Percentage of classic oncogenic mutations in all 74 samples.</p>
Full article ">Figure 4
<p>Mutation alleles based on Sanger sequencing. (<b>a</b>) Validation of KIT novel mutations using PCR-based Sanger sequencing; (<b>b</b>) validation of BRAF mutations using PCR-based Sanger sequencing.</p>
Full article ">
16 pages, 1061 KiB  
Review
Current Molecular Profile of Gastrointestinal Stromal Tumors and Systemic Therapeutic Implications
by Maria Cecilia Mathias-Machado, Victor Hugo Fonseca de Jesus, Leandro Jonata de Carvalho Oliveira, Marina Neumann and Renata D’Alpino Peixoto
Cancers 2022, 14(21), 5330; https://doi.org/10.3390/cancers14215330 - 29 Oct 2022
Cited by 12 | Viewed by 3249
Abstract
Gastrointestinal stromal tumors (GISTs) are malignant mesenchymal tumors arising from the intestinal pacemaker cells of Cajal. They compose a heterogenous group of tumors due to a variety of molecular alterations. The most common gain-of-function mutations in GISTs are either in the KIT (60–70%) [...] Read more.
Gastrointestinal stromal tumors (GISTs) are malignant mesenchymal tumors arising from the intestinal pacemaker cells of Cajal. They compose a heterogenous group of tumors due to a variety of molecular alterations. The most common gain-of-function mutations in GISTs are either in the KIT (60–70%) or platelet-derived growth factor receptor alpha (PDGFRA) genes (10–15%), which are mutually exclusive. However, a smaller subset, lacking KIT and PDGFRA mutations, is considered wild-type GISTs and presents distinct molecular findings with the activation of different proliferative pathways, structural chromosomal and epigenetic changes, such as inactivation of the NF1 gene, mutations in the succinate dehydrogenase (SDH), BRAF, and RAS genes, and also NTRK fusions. Currently, a molecular evaluation of GISTs is imperative in many scenarios, aiding in treatment decisions from the (neo)adjuvant to the metastatic setting. Here, we review the most recent data on the molecular profile of GISTs and highlight therapeutic implications according to distinct GIST molecular subtypes. Full article
(This article belongs to the Special Issue Updates on the Molecular Profile of Gastrointestinal Stromal Tumors)
Show Figures

Figure 1

Figure 1
<p>Proposed stepwise molecular testing for GISTs (when NGS is not promptly available). Legend: Initial test for the presence of <span class="html-italic">KIT</span> and <span class="html-italic">PDGFRA</span> mutations. If negative, test for SDH status by IHC. If SDH-proficient, patients should be tested for other genetic alterations. If SDH-deficient, test for mutations in <span class="html-italic">SDH A/B/C/D</span>. When mutated, consider germline testing for CSS. If not mutated, consider SDH C promoter methylation testing.</p>
Full article ">Figure 2
<p>Location and frequencies of most common KIT and PDGFRA mutations.</p>
Full article ">
14 pages, 1115 KiB  
Review
Latest Advances in the Management of Pediatric Gastrointestinal Stromal Tumors
by Marta Andrzejewska, Jakub Czarny and Katarzyna Derwich
Cancers 2022, 14(20), 4989; https://doi.org/10.3390/cancers14204989 - 12 Oct 2022
Cited by 8 | Viewed by 3138
Abstract
Gastrointestinal stromal tumor is the most common mesenchymal neoplasm of the gastrointestinal tract, usually found in elderly adults. It is infrequent among pediatric patients and usually differs biologically from adult-type diseases presenting mutations of KIT and PDGFR genes. In this population, more frequent [...] Read more.
Gastrointestinal stromal tumor is the most common mesenchymal neoplasm of the gastrointestinal tract, usually found in elderly adults. It is infrequent among pediatric patients and usually differs biologically from adult-type diseases presenting mutations of KIT and PDGFR genes. In this population, more frequent is the wild-type GIST possessing SDH, TRK, RAS, NF1 mutations, among others. Both tumor types require individualized treatment with kinase inhibitors that are still being tested in the pediatric population due to the different neoplasm biology. We review the latest updates to the management of pediatric gastrointestinal tumors with a particular focus on the advances in molecular biology of the disease that enables the definition of possible resistance. Emerging treatment with kinase inhibitors that could serve as targeted therapy is discussed, especially with multikinase inhibitors of higher generation, the effectiveness of which has already been confirmed in the adult population. Full article
Show Figures

Figure 1

Figure 1
<p>Choice of kinase inhibitors in the management of pediatric gastrointestinal stromal tumor.</p>
Full article ">Figure 2
<p>Management algorithm when a gastrointestinal stromal tumor in a pediatric patient is diagnosed.</p>
Full article ">
15 pages, 2348 KiB  
Article
Integrated Antitumor Activities of Cellular Immunotherapy with CIK Lymphocytes and Interferons against KIT/PDGFRA Wild Type GIST
by Erika Fiorino, Alessandra Merlini, Lorenzo D’Ambrosio, Ilaria Cerviere, Enrico Berrino, Caterina Marchiò, Lidia Giraudo, Marco Basiricò, Annamaria Massa, Chiara Donini, Valeria Leuci, Ramona Rotolo, Federica Galvagno, Letizia Vitali, Alessia Proment, Soldano Ferrone, Alberto Pisacane, Ymera Pignochino, Massimo Aglietta, Giovanni Grignani, Giulia Mesiano and Dario Sangioloadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2022, 23(18), 10368; https://doi.org/10.3390/ijms231810368 - 8 Sep 2022
Cited by 11 | Viewed by 2033
Abstract
Gastrointestinal stromal tumors (GISTs) are rare, mesenchymal tumors of the gastrointestinal tract, characterized by either KIT or PDGFRA mutation in about 85% of cases. KIT/PDGFRA wild type gastrointestinal stromal tumors (wtGIST) account for the remaining 15% of GIST and represent an unmet medical [...] Read more.
Gastrointestinal stromal tumors (GISTs) are rare, mesenchymal tumors of the gastrointestinal tract, characterized by either KIT or PDGFRA mutation in about 85% of cases. KIT/PDGFRA wild type gastrointestinal stromal tumors (wtGIST) account for the remaining 15% of GIST and represent an unmet medical need: their prevalence and potential medical vulnerabilities are not completely defined, and effective therapeutic strategies are still lacking. In this study we set a patient-derived preclinical model of wtGIST to investigate their phenotypic features, along with their susceptibility to cellular immunotherapy with cytokine-induced killer lymphocytes (CIK) and interferons (IFN). We generated 11 wtGIST primary cell lines (wtGISTc). The main CIK ligands (MIC A/B; ULBPs), along with PD-L1/2, were expressed by wtGISTc and the expression of HLA-I molecules was preserved. Patient-derived CIK were capable of intense killing in vitro against wtGISTc resistant to both imatinib and sunitinib. We found that CIK produce a high level of granzyme B, IFNα and IFNγ. CIK-conditioned supernatant was responsible for part of the observed tumoricidal effect, along with positive bystander modulatory activities enhancing the expression of PD-L1/2 and HLA-I molecules. IFNα, but not In, had direct antitumor effects on 50% (4/8) of TKI-resistant wtGISTc, positively correlated with the tumor expression of IFN receptors. wtGIST cells that survived IFNα were still sensitive to CIK immunotherapy. Our data support the exploration of CIK immunotherapy in clinical studies for TKI-resistant wtGIST, proposing reevaluation for IFNα within this challenging setting. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic summary of the experimental platform from GIST patients. m = months.</p>
Full article ">Figure 2
<p>CD117 and DOG1 expression in GIST samples and corresponding wtGISTc. CD117 (<b>A</b>) and DOG1 (<b>B</b>) immunohistochemistry staining (magnification 40×) in two representative surgical samples from GIST patients (diffuse expression on the left, focal expression on the right) and the corresponding wtGISTc. S012 and S108: unique patient numbers.</p>
Full article ">Figure 3
<p>Sensitivity to imatinib and sunitinib of wtGISTc in vitro. wtGISTc resulted resistant to imatinib (IC50 17 ± 8 µM, mean ± SEM) (<b>A</b>) and moderately sensitive to sunitinib (IC50 6 ± 2 µM, mean ± SEM) (<b>B</b>). A498 RCC line was used as a positive control of sunitinib sensitivity.</p>
Full article ">Figure 4
<p>Phenotype and main subsets of patient-derived CIK lymphocytes. Representative phenotype of CIK at the end of their ex vivo expansion; mature CIK were mainly CD3<sup>+</sup>CD8<sup>+</sup> with a relevant CD3<sup>+</sup>CD56<sup>+</sup> double-positive subset. The receptors mostly imputed to cancer cell recognition, NKG2D and DNAM-1, were expressed on a high percentage of cells (<b>A</b>). Distribution of main lymphocyte subsets with effector memory (EM), effector memory-RA<sup>+</sup> (EM RA), central memory (CM), and naïve (<b>B</b>).</p>
Full article ">Figure 5
<p>(<b>A</b>) Antitumor activity of CIK against wtGISTc resistant to TKI. Patient-derived CIK efficiently killed in vitro wtGISTc resistant to imatinib. The CIK killing ability was retained when sequentially tested against wtGISTc that survived treatment with sunitinib. (<b>B</b>) CIK-conditioned medium is endowed with tumoricidal activity against wtGISTc in vitro. CIK-conditioned supernatants, collected after 72 h co-culture of CIK with wtGISTc, was shown to be capable of cancer cell killing activity even in the absence of a direct CIK-tumor contact. (<b>C</b>,<b>D</b>) Direct and indirect modulation of HLA-I and β2M in wtGISTc by CIK lymphocytes. The membrane expression of both HLA-I (<b>C</b>) and β2M (<b>D</b>) on wtGIST was enhanced after treatment with CIK or CIK-conditioned medium (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.005, nonparametric <span class="html-italic">t</span>-test). (<b>E</b>) Soluble factors produced by activated CIK lymphocytes. Granzyme B, IFNα and IFNγ are intensely produced by CIK following contact with wtGIST (10:1 E:T).</p>
Full article ">Figure 6
<p>Sensitivity of wtGISTc to IFNα and IFNγ. After treatment, all wtGISTc resulted resistant to IFNγ and 4 out of 8 wtGISTc were relatively sensitive to IFNα (mean tumor cell death 42%).</p>
Full article ">Figure 7
<p>IFNα and IFNγ receptors (subunit 1) in wtGISTc. wtGISTc were confirmed to express IFNα/ɣ R1 even if at a lower level than monocytes, used as positive controls (<b>A</b>). The expression levels of IFNαR1 mRNA were significantly higher in wtGISTc sensitive to IFNα (<b>B</b>). All values are represented in fold change and normalized on three housekeeping genes (GAPDH, HPRT and PGK). (*** <span class="html-italic">p</span> &lt; 0.001, nonparametric <span class="html-italic">t</span>-test).</p>
Full article ">Figure 8
<p>Modulation of HLA-I, β2-microglobulin, PD-L1 and PD-L2 expression mediated by IFNs in wtGISTc. IFNs determined a significant enhancement in the expression of HLA-I/B2M and immune checkpoints (PDL-1/2) in wtGISTc. All the reported values were compared to untreated controls and expressed in MFI. (**** <span class="html-italic">p</span> &lt; 0.0001; ** <span class="html-italic">p</span> &lt; 0.005; * <span class="html-italic">p</span> &lt; 0.05; nonparametric <span class="html-italic">t</span>-test).</p>
Full article ">
9 pages, 1761 KiB  
Case Report
Metastatic SDH-Deficient GIST Diagnosed during Pregnancy: Approach to a Complex Case
by Anas Chennouf, Elie Zeidan, Martin Borduas, Maxime Noël-Lamy, John Kremastiotis and Annie Beaudoin
Curr. Oncol. 2022, 29(8), 5933-5941; https://doi.org/10.3390/curroncol29080468 - 20 Aug 2022
Cited by 3 | Viewed by 2636
Abstract
Gastrointestinal stromal tumors (GISTs) account for 1% of GI neoplasms in adults, and epidemiological data suggest an even lower occurrence in pregnant women. The majority of GISTs are caused by KIT and PDGFRA mutations. This is not the case in women of childbearing [...] Read more.
Gastrointestinal stromal tumors (GISTs) account for 1% of GI neoplasms in adults, and epidemiological data suggest an even lower occurrence in pregnant women. The majority of GISTs are caused by KIT and PDGFRA mutations. This is not the case in women of childbearing age. Some GISTs do not have a KIT/PDGFRA mutation and are classified as wild-type (WT) GISTs. WT-GIST includes many molecular subtypes including SDH deficiencies. In this paper, we present the first case report of a metastatic SDH-deficient GIST in a 23-year-old pregnant patient and the challenges encountered given her concurrent pregnancy. Our patient underwent a surgical tumor resection of her gastric GIST as well as a lymphadenectomy a week after induction of labor at 37 + 1 weeks. She received imatinib, sunitinib as well as regorafenib afterward. These drugs were discontinued because of disease progression despite treatment or after side effects were reported. Hence, she is currently under treatment with ripretinib. Her last FDG-PET showed a stable disease. This case highlights the complexity of GI malignancy care during pregnancy, and the presentation and management particularities of metastatic WT-GISTs. This case also emphasizes the need for a multidisciplinary approach and better clinical guidelines for offering optimal management to women in this specific context. Full article
(This article belongs to the Section Gastrointestinal Oncology)
Show Figures

Figure 1

Figure 1
<p>Axial (<b>A</b>) and coronal (<b>B</b>) contrast-enhanced CT view showing an 8.3 cm polylobate submucosal lesion with cystic components (yellow arrows) consistent with a GIST.</p>
Full article ">Figure 2
<p>Pathological cut of gastric GIST with immunohistochemistry stain for CD117/c-kit.</p>
Full article ">Figure 3
<p>HE stains showing GIST lymph node metastasis.</p>
Full article ">Figure 4
<p>Nine-month follow-up FDG-TEP scan after introduction of imatinib, showing disease progression with new liver metastasis.</p>
Full article ">Figure 5
<p>Timeline of the patient’s diagnosis and treatments.</p>
Full article ">
22 pages, 908 KiB  
Review
Treatment of Gastrointestinal Stromal Tumors (GISTs): A Focus on Younger Patients
by Monika Dudzisz-Śledź, Anna Klimczak, Elżbieta Bylina and Piotr Rutkowski
Cancers 2022, 14(12), 2831; https://doi.org/10.3390/cancers14122831 - 8 Jun 2022
Cited by 8 | Viewed by 2980
Abstract
Gastrointestinal stromal tumors (GISTs) originate from Cajal’s cells and are the most common mesenchymal neoplasms of the gastrointestinal tract. GISTs in young adults, i.e., patients before the age of 40, are rare and differ from those in older patients and GISTs in children [...] Read more.
Gastrointestinal stromal tumors (GISTs) originate from Cajal’s cells and are the most common mesenchymal neoplasms of the gastrointestinal tract. GISTs in young adults, i.e., patients before the age of 40, are rare and differ from those in older patients and GISTs in children in terms of the molecular and clinical features, including the location and type of mutations. They often harbor other molecular abnormalities than KIT and PDGFRA mutations (wild-type GISTs). The general principles of therapeutic management in young patients are the same as in the elderly. Considering some differences in molecular abnormalities, molecular testing should be the standard procedure to allow appropriate systemic therapy if needed. The optimal treatment strategy should be established by a multidisciplinary team experienced in sarcoma treatment. The impact of treatment on the quality of life and daily activities, including the impact on work, pregnancy, and fertility, in this patient population should be especially taken into consideration. Full article
(This article belongs to the Special Issue Young-Onset GI Cancer)
Show Figures

Figure 1

Figure 1
<p>Advanced wild-type GIST originating in the stomach in young adult women treated for 18 years.</p>
Full article ">Figure 2
<p>CT scan before (<b>A</b>) and after (<b>B</b>) resection of residual metastatic lesion in the abdominal cavity in young women with GIST with <span class="html-italic">KIT</span> exon 9 deletion mutation arising from the small intestine, after treatment with TKIs.</p>
Full article ">
10 pages, 955 KiB  
Article
Tropomyosin-Related Kinase Fusions in Gastrointestinal Stromal Tumors
by Ji Hyun Lee, Su-Jin Shin, Eun-Ah Choe, Jungyoun Kim, Woo Jin Hyung, Hyo Song Kim, Minkyu Jung, Seung-Hoon Beom, Tae Il Kim, Joong Bae Ahn, Hyun Cheol Chung and Sang Joon Shin
Cancers 2022, 14(11), 2659; https://doi.org/10.3390/cancers14112659 - 27 May 2022
Cited by 8 | Viewed by 2109 | Correction
Abstract
The canonical mutations in gastrointestinal stromal tumors (GISTs) are typically activating mutations in KIT and platelet-derived growth factor receptor alpha (PDGFRA). GISTs with non-canonical mutations are a heterogeneous group. Here, we examined tropomyosin-related kinase (TRK) fusion in GIST cases without KIT/PDGFRA mutations ( [...] Read more.
The canonical mutations in gastrointestinal stromal tumors (GISTs) are typically activating mutations in KIT and platelet-derived growth factor receptor alpha (PDGFRA). GISTs with non-canonical mutations are a heterogeneous group. Here, we examined tropomyosin-related kinase (TRK) fusion in GIST cases without KIT/PDGFRA mutations (KIT/PDGFRA wild-type (WT) GISTs). We retrospectively analyzed patients who were diagnosed with GISTs at the Yonsei Cancer Center, Severance Hospital, between January 1998 and December 2016. Thirty-one patients with KIT/PDGFRA WT GISTs were included in the analysis. TRK expression in tumor samples was assessed by pan-TRK immunohistochemistry (IHC), and the neurotrophic tyrosine receptor kinase (NTRK: the gene encoding TRK) rearrangement was analyzed by fluorescence in situ hybridization (FISH). IHC analyses revealed that five cases in this cohort exhibited a weak to moderate TRK expression. NTRK1 fusions were detected in three tumor samples, and two samples harbored NTRK3 fusions. The remaining 26 samples did not harbor NTRK fusions. Two types of NTRK fusions were detected, and the overall NTRK fusion frequency in KIT/PDGFRA WT GIST cases was 16% (5/31). Our data provide insights into the molecular alterations underpinning KIT/PDGFRA WT GISTs. More effort should be devoted to improve methods to identify this distinct disease subtype within the KIT/PDGFRA WT GIST group. Full article
Show Figures

Figure 1

Figure 1
<p>Representative images of pan-TRK expression in samples of WT GIST. IHC results showing samples in which the intensity of cytoplasmic staining was scored as negative (<b>A</b>), weakly positive (<b>B</b>), and moderately positive (<b>C</b>). There were no cases scored as strongly positive. Each scale bar is 100 μM. TRK: tropomyosin-related kinase; WT: wild type; GIST: gastrointestinal stromal tumor; IHC: immunohistochemistry.</p>
Full article ">Figure 2
<p>Representative images of <span class="html-italic">NTRK</span> fusion detected by FISH. Tumor tissues were stained with dual-color FISH probes. The red and green signals represent upstream and downstream probes, respectively. FISH results showing (<b>A</b>) <span class="html-italic">NTRK1</span> and (<b>B</b>) <span class="html-italic">NTRK3</span> fusions. Each scale bar is 10 μM. Circles indicate gene rearrangements. NTRK: neurotrophic tyrosine receptor kinase; FISH: fluorescence in situ hybridization.</p>
Full article ">
8 pages, 4036 KiB  
Article
Rare Occurrence of Microsatellite Instability in Gastrointestinal Stromal Tumors
by Joonhong Park, Hae Jung Sul and Jeong Goo Kim
Medicina 2021, 57(2), 174; https://doi.org/10.3390/medicina57020174 - 18 Feb 2021
Cited by 3 | Viewed by 2312
Abstract
Background and Objectives: This study aimed to objectively determine microsatellite instability (MSI) status using a next-generation sequencing (NGS)-based MSI panel and to resolve the discrepancy regarding whether or not MSI is a rare phenomenon, irrespective of diverse genomic alterations in gastrointestinal stromal tumors [...] Read more.
Background and Objectives: This study aimed to objectively determine microsatellite instability (MSI) status using a next-generation sequencing (NGS)-based MSI panel and to resolve the discrepancy regarding whether or not MSI is a rare phenomenon, irrespective of diverse genomic alterations in gastrointestinal stromal tumors (GISTs). Materials and Methods: Genomic DNA was subjected to MSI panel sequencing using an Ion AmpliSeq Microsatellite Instability Assay, as well as to cancer gene panel sequencing using an Oncomine Focus DNA Assay. Results: All of our GIST patients showed microsatellite-stable (MSS) status, which confirmed that MSI status did not affect the molecular pathogenesis of GIST. The KIT gene (79%, 38/48) was the most frequently mutated gene, followed by the PDGFRA (8%, 4/48), PIK3CA (8%, 4/48), and ERBB2 (4%, 2/48) mutations. KIT exon 11 mutant patients were more favorable in responding to imatinib than those with exon 9 mutant or wild-type GISTs, and compared to non-KIT exon 11 mutant GISTs (p = 0.041). The NGS-based MSI panel with MSICall confirmed a rare phenomenon of microsatellite instability in GISTs irrespective of diverse genomic alterations. Conclusion: Massively parallel sequencing can simultaneously provide the MSI status as well as the somatic mutation profile in a single test. This combined approach may help us to understand the molecular pathogenesis of GIST carcinogenesis and malignant progression. Full article
(This article belongs to the Special Issue Research on Cancer Biology)
Show Figures

Figure 1

Figure 1
<p>Distribution of microsatellite instability (MSI) scores generated from the Ion AmpliSeq Microsatellite Instability Assay in 48 gastrointestinal stromal tumors.</p>
Full article ">Figure 2
<p>Frequencies of somatic mutation types detected in the various genes by the Oncomine Focus DNA Assay in 48 gastrointestinal stromal tumors. Gene identities are depicted on the x-axis, and the number of mutations is depicted on the y-axis.</p>
Full article ">Figure 3
<p>Distribution of somatic mutations in the KIT and PDGFRA functional domains. The somatic mutations in KIT and PDGFRA identified in our study are shown. The types of mutations detected within the domains are indicated above. Boxes represent functional domains: I–V, 5 immunoglobulin-like domain; TM, transmembrane domain; JM, juxtamembrane domain; TK1, tyrosine kinase domain 1; KI, kinase insert domain; TK2, tyrosine kinase domain 2.</p>
Full article ">
23 pages, 5544 KiB  
Review
Update on Molecular Genetics of Gastrointestinal Stromal Tumors
by Iva Brčić, Alexandra Argyropoulos and Bernadette Liegl-Atzwanger
Diagnostics 2021, 11(2), 194; https://doi.org/10.3390/diagnostics11020194 - 28 Jan 2021
Cited by 47 | Viewed by 6552
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. The majority are sporadic, solitary tumors that harbor mutually exclusive KIT or PDGFRA gain-of-function mutations. The type of mutation in addition to risk stratification corresponds to the biological behavior [...] Read more.
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. The majority are sporadic, solitary tumors that harbor mutually exclusive KIT or PDGFRA gain-of-function mutations. The type of mutation in addition to risk stratification corresponds to the biological behavior of GIST and response to treatment. Up to 85% of pediatric GISTs and 10–15% of adult GISTs are devoid of these (KIT/PDGFRA) mutations and are referred to as wild-type GISTs (wt-GIST). It has been shown that these wt-GISTs are a heterogeneous tumor group with regard to their clinical behavior and molecular profile. Recent advances in molecular pathology helped to further sub-classify the so-called “wt-GISTs”. Based on their significant clinical and molecular heterogeneity, wt-GISTs are divided into a syndromic and a non-syndromic (sporadic) subgroup. Recently, the use of succinate dehydrogenase B (SDHB) by immunohistochemistry has been used to stratify GIST into an SDHB-retained and an SDHB-deficient group. In this review, we focus on GIST sub-classification based on clinicopathologic, and molecular findings and discuss the known and yet emerging prognostic and predictive genetic alterations. We also give insights into the limitations of targeted therapy and highlight the mechanisms of secondary resistance. Full article
(This article belongs to the Special Issue Molecular Classification of Soft Tissue and Bone Tumors)
Show Figures

Figure 1

Figure 1
<p>Morphology and immunohistochemical findings in GIST (Gastrointestinal Stroma Tumor). (<b>A</b>) Spindle cell GIST (inset: IHC DOG1+). (<b>B</b>) Epithelioid GIST (inset: IHC DOG1+). (<b>C</b>) Spindle cell GISTs with nuclear palisading, (<b>D</b>) GIST with storiform growth pattern, and (<b>E</b>) GIST with prominent paranuclear vacuolation. (<b>F</b>) Epithelioid GIST with a prominent plasmacytoid morphology. (<b>G</b>) Pleomorphic GIST (inset: IHC KIT+). (<b>H</b>) Dedifferentiated GIST (inset: IHC DOG1− and KIT−). IHC (immunohistochemistry); −(negative); +(positive).</p>
Full article ">Figure 2
<p>Sub-classification of GISTs into a succinate dehydrogenase (SDH)-competent and an SDH-deficient group by using an SDHB IHC <sup>#</sup>. Legend: CSS: Carney–Stratakis Syndrome; CT: Carney triad; CT *: in some cases, mutations described [<a href="#B9-diagnostics-11-00194" class="html-bibr">9</a>]; wt: wild type. Grey rectangle: DNA- and RNA sequencing in a specialized center.</p>
Full article ">Figure 3
<p>Graphical representation of <span class="html-italic">KIT</span> and <span class="html-italic">PDGFRA</span> transmembrane tyrosine kinase receptors with frequency and localization/distribution of primary mutations found in sporadic GIST (adapted from [<a href="#B53-diagnostics-11-00194" class="html-bibr">53</a>,<a href="#B54-diagnostics-11-00194" class="html-bibr">54</a>]).</p>
Full article ">Figure 4
<p>Secondary KIT mutations and predictive response to most frequently used TKIs (IM:imatinib; SU: sunitinib; RE: regorafenib). Adapted from [<a href="#B80-diagnostics-11-00194" class="html-bibr">80</a>].</p>
Full article ">Figure 5
<p>Diagnostic algorithm in wild-type gastrointestinal stromal tumors (wt-GISTs). Legend: Assoc.: associated; CSS: Carney–Stratakis Syndrome; CT: Carney triad; IHC: immunohistochemistry; PC: pulmonary chondroma; PGL: paraganglioma; SDHB: Succinate dehydrogenase B; wt: wild type; *: rarely reported.</p>
Full article ">Figure 6
<p>Diagnostic and molecular testing algorithm for GIST. * Our GIST mutation-panel includes following genes: PDGFA, KRAS, NRAS, HRAS, BRAF, KIT (Exon 8,9,10,11,12,13,17,18), PDGFRB (Exon 12,13,14,17,18), TP53 (Exons 4–10), SDHA, SDHB, SDHC, SDHD, NF1, CDKN2A and RB1. ** If available, mutation-panel is performed. TKI: Tyrosin Kinase Inhibitor.</p>
Full article ">Figure 7
<p>Immunohistochemical staining with pan-TRK antibody in GIST shows diffuse cytoplasmic and membranous expression. (Scale bar shows 0.1 mm).</p>
Full article ">Figure 8
<p>Morphology and immunohistochemical findings in SDHB-deficient GIST. (<b>A</b>) The gastric tumor with multilobulated/plexiform growth pattern. (<b>B</b>) The tumor is composed of epithelioid cells with a syncytial appearance. (<b>C</b>,<b>D</b>) Immunohistochemically, the tumor shows positivity for KIT and DOG1 (<b>C</b>), while the expression of SDHB is lost (<b>D</b>); the cytoplasmic stain of the endothelial cells shows the positive internal control).</p>
Full article ">
Back to TopTop