Gastrointestinal mesenchymal tumors





Gastrointestinal stromal tumor


Gastrointestinal stromal tumors (GISTs) are soft tissue tumors that either express C-kit/CD117 protein or have C-KIT or platelet-derived growth factor receptor-α ( PDGFRA ) mutations and show spindle cell or epithelioid morphology. The earlier literature attempted to classify them as smooth muscle or nerve sheath tumors. The term stromal tumor was introduced in 1983, after Mazur and Clark failed to find ultrastructural evidence of smooth muscle or nerve sheath differentiation in several gastric tumors.


GISTs have been shown to display differentiation toward interstitial cells of Cajal (ICC), which normally govern gut motility. The availability of specific antibodies and clarification of their immunohistochemical profile has facilitated uniform diagnosis.


Clinical features


GISTs comprise 5% to 10% of all sarcomas and represent about 1% of all GI malignancy. In the United States, 3300 to 4350 new GISTs are reported annually. About 20% to 25% of gastric GISTs are malignant, and the number is higher for intestinal GISTs, 40% to 50%. They are most common in adults 50 to 60 years of age. These tumors vary in differentiation and prognosis according to their location within the GI tract. GISTs are rare in the esophagus, 50% to 70% involve the stomach, 25% to 40% the small intestine (of which 10% to 20% arise in the duodenum, 27% to 37% in the jejunum, and 27% to 53% in the ileum), and less than 10% are colorectal (50% colonic, 50% rectal). GIST-type tumors arising in the omentum, peritoneum, and retroperitoneum have been identified, comprising 6.7% of the large Armed Forces Institute of Pathology (AFIP) series of 1008 GISTs.


Tumors present with primary mass, pain, hemorrhage, or metastasis, and two thirds exceed 5 cm in diameter at presentation. GISTs often metastasize to the abdominal cavity and liver, rarely to bone, soft tissue, skin, lymph nodes, and lungs. Metastasis can occur more than 10 years after surgical resection, underscoring a need for long-term follow-up.


There are three hereditary syndromes of which GIST is a component. First, GIST is found in patients with neurofibromatosis-1 (NF-1). NF-1 is an autosomal dominant genetic disorder resulting from a defect in the NF1 gene on chromosome 17; affected patients have a deficiency of neurofibromin protein. The clinical features of NF-1 include café-au-lait spots, freckling, neurofibromas, malignant peripheral nerve sheath tumors, ganglioneuromas, and GISTs. GISTs arising in the setting of NF-1 tend to be multiple, typically involving the small intestine, and lack PDGFRA and C-KIT mutations. Instead, GISTs in NF-1 have been shown to have somatic inactivation of their wild-type NF1 allele, leading to inactivation of neurofibromin, and causing activation of MAP-kinase pathway. Second, familial GIST, in patients with germline C-KIT or PDGFRA mutations, manifests as a syndrome of GIST, hyperpigmentation, urticaria pigmentosa, mastocytosis, dysphagia, and hyperplasia of ICC. Of the documented families, most have C-KIT gene alterations but the PDGFRA gene is occasionally affected. Third, GIST is a component of Carney’s triad (gastric GIST, paraganglioma, and pulmonary chondroma). GIST in Carney’s triad occurs in young individuals, has epithelioid morphology, and shows a strong female predominance. No specific genetic alteration has been identified for Carney’s triad.




GASTROINTESTINAL STROMAL TUMORS—FACT SHEET


Definition





  • Gastrointestinal stromal tumor (GIST) is a soft tissue tumor arising from the GI tract that typically expresses CD117 or has C-KIT or platelet-derived growth factor receptor α (PDGFRA) mutations and shows spindle cells or epithelioid morphology



Incidence and location





  • GIST is extremely rare in the esophagus. It is commonly found in the stomach and intestine



  • Up to 5000 cases per year in the United States



Morbidity and mortality





  • Intestinal GIST is more likely to be malignant than gastric GIST



Gender, race, and age distribution





  • Adults with no gender predominance; may be overrepresented in African-Americans



Clinical features





  • Site-specific presentation of mural mass



Prognosis and therapy





  • Median survival for esophagus, 29 months; in stomach, 20% of patients die of disease; in the small bowel, about 40% of patients die of disease; similar for colon; appendix tumors rare



  • Treatment includes imatinib and newer tyrosine kinase inhibitors



  • Patients with exon 11 mutations in the C-KIT gene are most likely to respond to this treatment




Radiologic features


Radiologic features are not specific, but most large mural gastric masses are GISTs. An example of a gastric epithelioid GIST is seen in Figure 7-1 .




FIGURE 7-1


Imaging study from a patient with a large gastric stromal tumor. It was mistaken for a primary liver lesion by the radiologist.


Pathologic features


Gross findings


Tumors vary in diameter from less than 1 cm to greater than 20 cm, and can be submucosal, intramuscular, or subserosal, although most are centered in the muscularis propria. Generally GISTs are relatively well marginated. They can be solid or cystic with variable hemorrhage and necrosis, including mucosal ulceration and tumor cavitation. Figure 7-2 shows a small intestinal GIST.




FIGURE 7-2


Small intestinal stromal tumor. This lesion is malignant with metastases to the liver.


Microscopic findings


There are spindle, epithelioid, and (rare) pleomorphic forms in a variety of patterns and with modifications resulting from stromal features. The two principal cell types are spindle and epithelioid ( Figs. 7-3 through 7-7 ), which can coexist in varying proportions. About 70% of gastric GISTs and most of those in the small and large intestine are spindled. Most esophageal and rectal GISTs are malignant spindle cell tumors. The spindle cells are relatively short, are often uniform, and have tapered nuclei with amphophilic or eosinophilic slightly fibrillary cytoplasm. They form cellular sheets and fascicles with whorled, storiform, or palisaded patterns. The epithelioid cells have more abundant cytoplasm, with perinuclear halos and well-defined cell borders. Epithelioid GISTs have also been termed leiomyoblastomas or epithelioid smooth muscle tumors . Clear cell, signet ring, oncocytic, and plasmacytoid variants occur, but multinucleated cells are uncommon. Mitoses vary from none to many. Truly pleomorphic tumors are rare, although both pleomorphism and even unusual differentiation (e.g., skeletal muscle) can be seen in lesions that have been treated with imatinib or related tyrosine kinase inhibitors. The stroma is scanty, but fibrous septa sometimes delineate tumor nests in an organoid pattern. Myxoid or cystic change or hyalinization is sometimes seen, and there can be a variable lymphoplasmacytic inflammatory infiltrate.




FIGURE 7-3


Gastric stromal tumors exhibiting paranuclear vacuoles. A, Such vacuoles are also a feature of smooth muscle tumors, but they are seldom so numerous in true smooth muscle tumors. B, Myxoid stroma is seen, a feature that would not be present in a smooth muscle tumor, and the nuclei have more tapered ends than smooth muscle tumor nuclei.



FIGURE 7-4


Small intestinal GIST. Corresponds to the gross lesion depicted in Figure 7-1 and is highly cellular and mitotically active.



FIGURE 7-5


Epithelioid gastric GIST. Such tumors have been termed “leiomyoblastomas” in the past.



FIGURE 7-6


Epithelioid gastric GIST. On the left portion of the field, this lesion has invaded the mucosa, a feature associated with a poor prognosis. Note that the tumor is centered in the muscularis propria.



FIGURE 7-7


Epithelioid gastric GIST. Note the prominent cytoplasmic vacuoles and uniform nuclei; nuclear pleomorphism is unusual in GISTs and its presence should prompt the pathologist to consider other diagnoses.




GASTROINTESTINAL STROMAL TUMORS—PATHOLOGIC FEATURES


Gross findings





  • Mural mass with variable appearances, including cysts



Microscopic findings





  • Spindle cell or epithelioid, sometimes with “skeinoid” fibers in stroma (particularly in small intestine); sometimes cells have paranuclear vacuoles



Immunohistochemistry





  • Positive for CD117, DOG1, CD34, variable actin, S-100 protein, and PDGFRA



Molecular testing





  • Most mutations in C-KIT in exon 11, and most mutations in PDGFRA in exon 18



  • Some mutations predictive of response to targeted therapy



  • Laboratories that offer testing can be found at http://www.amptestdirectory.org



Differential diagnosis





  • Smooth muscle (desmin-positive, CD117–) and neural (S-100+, CD117–) tumors



  • Sometimes fibromatosis/desmoid has false-positive results for CD117/C-kit




Ancillary studies


Ultrastructural findings


Early studies failed to reveal well-developed smooth muscle differentiation. A minority (about 20%) of GISTs show focally incomplete features of smooth muscle differentiation, with arrays of cytoplasmic intermediate filaments and dense bodies. Many GISTs show neuronal-like differentiation with interdigitating neurite-like cytoplasmic processes that contain microtubules, smooth endoplasmic reticulum, and intermediate filaments. Dense core neuroendocrine granules, some of which are also associated with the prominent Golgi apparatus, and synapse-like structures have been described, but these are not usual features of ICC and might represent entrapped autonomic nerve. There are numerous mitochondria and occasional cell-to-cell junctions (including with smooth muscle cells and neurons).


Skeinoid fibers, which are amorphous periodic acid-Schiff–positive foci of haphazardly arranged modified collagen fibers with a periodicity of 45 nm, are seen rarely, usually in (small) intestinal tumors, benign and malignant, including those in the duodenum. Some GISTs, including CD117/CD34-positive cases, show no ultrastructural differentiation.


Immunohistochemistry


In the 1980s and early 1990s, immunohistochemical studies attempted to confirm supposed smooth muscle, neural, or dual differentiation in GISTs and to relate immunophenotype to behavior. Varying proportions were shown to have smooth muscle actin (SMA), muscle-specific actin (MSA), and desmin, with smaller numbers of cases displaying S-100 protein positivity. Significant numbers remained, however, with no specific markers or only vimentin. Gastrointestinal autonomic neural tumors (GANTs) were variably immunoreactive for neuron-specific enolase (NSE), neurofilaments, synaptophysin, and S-100 protein. In 1994, CD34 was shown to be a reliable marker for the majority of GISTs, and from 1998, CD117 positivity became definitional ( Fig. 7-8 ).




FIGURE 7-8


CD117/C-kit expression in GIST, with cytoplasmic and membranous labeling. CD117 staining is seen regardless of malignant potential in the vast majority of stromal tumors.


CD117 (KIT), the product of the C-KIT gene, is expressed among normal ICCs, mast cells, melanocytes, a variety of epithelia, fetal endothelial cells, and a subset of CD34-positive hematopoietic stem cells. Staining is usually cytoplasmic, but can be membranous in mast cells, melanocytes, and germ cells.


With immunohistochemistry, CD117 is positive (diffuse cytoplasmic staining with membranous accentuation) in the vast majority of benign and malignant GISTs. GISTs with spindle cell and epithelioid morphology are both positive, although less intensely in the latter. Smooth muscle tumors (SMA and desmin positive) and schwannomas (S-100 protein positive) are negative for CD117.


A variety of commercial antibodies to CD117 are available, with varying specificity and sensitivity. For example, using the rabbit polyclonal CD117 antibody marketed by Santa Cruz Biotechnology (C-19, sc-168) positivity is essentially confined to GISTs, among the similar tumors in the differential diagnosis. The rabbit polyclonal antibody A4502, manufactured by Dako (Carpinteria, CA), however, has been reported to stain (the cytoplasm of) lesional fibroblasts and myofibroblasts, including some examples of fibromatosis, and solitary fibrous tumor; awareness of this may prevent misdiagnosis of such tumors as GISTs. These “falsely positive” tumors lack kit mutations and respond only minimally to imatinib treatment.


A small number (2% to 10%) of otherwise typical GISTs are CD117 negative. These are predominantly epithelioid in cell morphology. It has been shown that protein kinase C theta (PKCT) is constitutively activated in all GISTs, including those that are C-kit negative, although by immunoblotting, PKCT expression in these is 50% lower than in C-kit-positive GISTs. Virtually all GISTs were positive with an antibody to PKCT phospho Thr 538 . DOG1 (discovered on GIST-1) antibodies also label most GISTs and may be helpful in that they tend to label GISTS that lack CD117 expression.


CD34 is positive in 47% to 100% of GISTs, and its expression varies with location within the GI tract. Miettinen and associates found, among CD117-positive tumors, 100%, 90%, 47%, 65%, 96%, and 64% of cases to be CD34-positive in esophagus, stomach, small intestine, colon, rectum, and extraintestinal locations, respectively. Among malignant gastric and small intestinal GISTs, 88% and 55% were CD34-positive. Tumors that are CD34-positive are almost always CD117-positive, and most CD34-negative tumors are CD117 positive. CD117 is sometimes lost in metastases. It should be noted that CD34 is also positive in occasional smooth muscle tumors, including epithelioid variants.


Antibodies against platelet-derived growth factor receptors α and β (PDGFRA and PDGFRB), which are produced by Santa Cruz Biotechnology (pAb, no.sc-338 for PDGFRA and pAb, no. sc-339 for PDGFRB), are also available and label a subset of CD117-negative GISTs. However, some fibromatoses cases also show labeling with PDGFRA and PDGFRB antibodies.


SMA is found only focally in up to half of GISTs in an inverse frequency of expression to that of CD34 (the two sometimes showing a mosaic distribution); benign GISTs of the small intestine have the highest proportion of SMA positivity. h-Caldesmon (an actin-binding cytoskeleton-associated protein) is positive in about 80% of GISTs, supporting myoid differentiation, and calponin in about 25%. Desmin is focally positive in up to 20% of tumors of the esophagus and omentum/peritoneum and in a small proportion of benign (but not in malignant) GISTs in the stomach and small intestine. Cytokeratins are rarely positive but are occasionally seen in malignant epithelioid GISTs.


A few have neurogenic differentiation, with positivity for S-100 protein (especially in small bowel tumors), PGP9.5, and NSE, and a few of these additionally express SMA, implying both neurogenic and myoid differentiation. Both GISTs and ICCs express embryonic smooth muscle myosin heavy chain messenger RNA.


Fine-needle aspiration biopsy


Fine-needle aspiration can be used to diagnose GISTs with appropriate immunohistochemistry but cannot be used to assess malignant potential.


Molecular studies


Two receptors in the type III receptor tyrosine kinase subfamily have been shown to play an important role in the pathogenesis of GIST: (1) C-kit receptor and (2) platelet-derived growth factor receptor-α ( PDGFRA ). The genes encoding for these two receptors are both located in chromosome 4. The protein structure of these receptors are also similar, including an extracellular (EC) domain, transmembrane (TM) domain, juxtamembrane (JM) domain, and tyrosine kinase (TK) domains. As a result of their similarities, activating mutation in either one of these two genes will lead to activation of similar signal transduction pathways leading to GIST formation.


In sporadic GISTs, 85% to 90% of the cases have mutations in the C-KIT gene while 10% to 15% of the remaining cases do not. The mutations of C-KIT gene occur in order of decreasing frequency: exon 11, exon 9, exon 13, and exon 17. The types of mutations in exon 11 include: in-frame deletions (60% to 70%) of one to several codons at 5′ end of codon 11 and region between codons Gln 550 to Glu 561 ; missense point mutations (20% to 30%) at Trp 557 , Val 559 , Val 560 , and Leu 576 ; and internal tandem duplications of 1 to more than 20 codons at 3′ end of codon 11. The clinical significance associated with mutations in exon 11 is that they may play a role in predicting response to treatment. Most mutations of the C-KIT gene occur at this exon (juxtamembrane domain). The juxtamembrane domain is an intracytoplasmic domain located between the transmembrane and tyrosine kinase domains and its function, after activation, is to induce dimerization of two C-kit receptors, which leads activation of the downstream signal transduction pathways.


At exon 9, encoding the extracellular domain, most of the cases show 6-nucleotide duplications encoding Ala 502 -Tyr 503 and this mutation is usually encountered in intestinal GISTs. At exon 13, the most common mutation is the 1945A>G substitution leading to Lys 642 Glu. At exon 17 encoding the tyrosine kinase II domain, the most common mutation is the 2487T>A substitution leading to Asn 822 Lys. Interestingly, seminoma and mastocytoma are two neoplasms that also express CD117; these both commonly have mutations in exon 17. In contrast, CD117-positive GIST rarely shows mutations at codon 17 and more commonly has exon 11 mutations.


As noted, 10% to 15% of sporadic GIST lack C-KIT gene mutations. About 35% to 63% of these cases have mutations in the PDGFRA gene. Mutations of PDGFRA gene occur, in order of decreasing frequency, in exon 18 (>80%), exon 12, and exon 14. In exon 18, a missense mutation leading to Asp 842 Val is commonly seen. In exon 12, a missense mutation is reported, Val 561 Asp. In exon 14, a missense mutation is seen, Asn 659 Lys/Tyr.


Understanding the specific mutations in C-KIT and PDGFRA genes is important because of the following five clinical implications. First, in terms of prognosis, GISTs with exon 11 mutations are associated with longer event-free and overall survival than GISTs with exon 9 mutations and GISTs with no detectable kinase mutation. Second, GISTs with exon 11 mutations have a better response to imatinib than GISTs with exon 9 mutation in C- KIT gene, GISTs with PDGFRA mutation, and wild-type GISTs. Third, in terms of dosing of imatinib, cases of metastatic/advanced GISTs with exon 9 mutation require higher doses of imatinib to achieve a response. Fourth, knowing the mutation status in the C-KIT and PDGFRA genes occasionally identifies patients who have unidentified genetic diseases. Specifically, in patients with wild-type GISTs, there is enrichment for NF-1 and Carney triad. Finally, when GISTs show resistance to first-line tyrosine kinase inhibitor such as imitanib, identification of secondary mutations can help in choosing a second-line tyrosine kinase inhibitor (e.g., sunitinib) for treatment. An excellent resource for arranging for molecular testing of GISTs, as well as other molecular tests, is found through the Association for Molecular Pathology at http://www.amptestdirectory.org/ .


Differential diagnosis


The differential diagnosis includes the other mesenchymal tumors discussed in this chapter, but in most cases, the differential diagnosis should include smooth muscle tumors, schwannomas, and occasionally fibromatosis.


Prognosis and therapy


Behavior


General


Perfect separation of benign and malignant GISTs cannot be achieved. However, the main prognostic factors identified have been mitotic count and tumor size. Both size and mitotic activity are continuous variables, and researchers have taken different cut-off points. Guidelines using combinations of maximum dimension and mitotic count for defining risk have recently been proposed as in Table 7-1 , but are imperfect because they do not take site into account. Refined schemes have been developed for gastric and small bowel GISTs ( Tables 7-2 through 7-4 ).



TABLE 7-1

Proposed Guidelines Using Combinations of Maximum Dimension and Mitotic Count for Defining Risk
































Size (cm) Mitotic Count/50 hpf
Very low risk <2 <5
Low risk 2-5 <5
Intermediate risk <5
5-10
6-10
<5
High risk >5 >5
>10 Any
Any >10

hpf, high-power field.


TABLE 7-2

Prognosis of Gastric versus Small Bowel GISTs
































Stomach GISTs; 20% Tumor-Related Deaths Small Intestinal GISTs; 40% Tumor-Related Deaths
Size/mitoses/50 hpf Outcome (with metastases) Size/mitoses/50 hpf Outcome (with metastases)
<10 cm, <5 mitoses/50 hpf 3% <5 cm, <5 mitoses/50 hpf 3%
>10 cm, ≥5 mitoses/50 hpf 86% >10 cm, ≥5 mitoses/50 hpf 86%
>10 cm, <5 mitoses/50 hpf 11% >10 cm, <5 mitoses/50 hpf or <5 cm, ≥5 mitoses/50 hpf >50%
<5 cm, ≥5 mitoses/50 hpf 15% 5-10 cm, ≥5 mitoses/50 hpf 24%

hpf, high-power field.

Data from , Lasota J: Gastrointestinal stromal tumors: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med 2006; 130:1466-1478.


TABLE 7-3

Suggested Prognostic Criteria for Resected GISTs Arising in the Stomach


































Category Group Size and Mitotic Activity Prognostic Features
Benign 1 ≤2 cm, ≤5 mitoses/50 hpf No tumor-related mortality detected
Probably benign 2
3a
>2 cm, ≤5 cm, ≤5 mitoses/50 hpf
>5 cm, ≥10 cm, ≥5 mitoses/50 hpf
Very low malignant potential, <3% progressive disease
Uncertain or low malignant potential 4 ≤2 cm, >5 mitoses/50 hpf No progressive disease (too few cases available to reliably determine prognosis)
Low to moderate malignant potential 3b
5
>10 cm, ≥5 mitoses/50 hpf
>2 cm, ≤5 cm, >5 mitoses/50 hpf
12%-15% tumor-related mortality
High malignant potential 6a
6b
>5, ≤10 cm, >5 mitoses/50 hpf
>10 cm, >5 mitoses/50 hpf
49%-86% tumor-related mortality

Data from , Lasota J: Gastrointestinal stromal tumors: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med 2006; 130:1466-1478.


TABLE 7-4

Suggested Prognostic Criteria for Resected GISTs Arising in the Jejunum and Ileum

















































Category Group Size And Mitotic Activity Prognostic Features
Practically benign 1 <2 cm, ≤5 mitoses/50 hpf No evidence of progressive disease
Low malignant potential 2 >2 cm, ≤5 cm, <5 mitoses/50 hpf <5% progressive disease
Moderate malignant potential 3a >5 cm, ≤10 cm, ≤5 mitoses/50 hpf 10%-30% progressive disease
High malignant potential 3b >10 cm, ≤5 mitoses/50 hpf ≥50% progressive disease/tumor-related mortality
4 ≤2 cm, >5 mitoses/50 hpf
5 >2 cm, <5 cm, >5 mitoses/50 hpf
6a >5 cm, ≤10 cm, >5 mitoses/50 hpf
6b >10 cm, >5 mitoses/50 hpf

Data from Miettinen M, Lasota J: Gastrointestinal stromal tumors: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med 2006; 130:1466-1478.


Site-specific analyses, including a large AFIP series of 1008 cases, have shown location, size, mitotic count, and age to be independent prognostic variables. Small intestinal tumors are the most aggressive. However, in no site are size and mitotic count alone sufficient for long-term prediction of behavior, so no definite criteria proposed for diagnosis of malignancy are predictive of outcome. For unclear reasons, malignant examples appear to be overrepresented in African-Americans.


Regional


Esophagus . GISTs account for a minority of esophageal stromal tumors (and leiomyomas for the majority), and involve the lower third or gastroesophageal junction, predominantly in males. They can be spindled or epithelioid and display the usual variety of patterns. All are CD117 and CD34 positive, and about 15% have SMA or desmin. The majority are malignant. In the series of Miettinen and coworkers, 9 of 16 patients died of disease, including all with tumors larger than 10 cm, and none with tumors smaller than 5 cm. Median survival was 29 months. However, a subset of GISTs of the gastroesophageal junction is minute, multiple, and presumably does not progress, because these “seedling” GISTS are encountered commonly (approximately 10%) in autopsies and resections when the entire gastroesophageal junction is embedded ( Fig. 7-9 )




FIGURE 7-9


“ Seedling” GISTs of the gastroesophageal junction are common and apparently do not progress to clinically evident lesions. Several small nodules are seen on H&E (A), and highlighted by CD117 (B) and CD34 (C) staining.


Stomach . About 20% are result in patient deaths. The consensus is that 5 mitoses per 50 high-power fields (hpf) and size greater than 5 cm are adverse prognostic factors. The 5-year survival rate of gastric GISTs is about 80%, with improvement in completely resected cases. There is no evidence that radical surgery improves survival, so the least extensive surgical procedure compatible with complete excision is advisable. Gastric GISTs are more frequent in males.


Epithelioid GISTs (formerly called leiomyoblastomas ) comprise about 10% of gastric GISTs, of which about 80% are benign. Large tumors in the fundus or cardiac area and posterior wall are more likely to be malignant.


Duodenum. Duodenal GISTs are most common in the second part, and 35% to 50% are malignant. Miettinen and associates studied 156 duodenal GISTs, with the following results: 12 patients whose tumors were less than 2 cm with mitoses less than or equal to 5/50hpf had no recurrence, metastases, or tumor-related death during follow-up of 6 years. However, 18 of 21 patients (86%) with tumors greater than 5 cm with greater than 5 mitoses/50 hpf had metastases and disease-related death with a median survival time of 21 months.


Jejunum and Ileum. About 40% result in patient deaths. These GISTs can be spindled, epithelioid, or mixed; mixed and epithelioid tumors are associated with worse behavior than spindled tumors. The presence of skeinoid fibers, PAS-reactive thick collagen fibers, present in about 45% of the cases, is a favorable prognostic factor. As in other sites, the most important factors determining the prognosis are tumor size and mitotic index. Small (<5 cm) with few mitoses (≤5 mitoses/50 hpf), convey an excellent prognosis. In contrast, large (>10 cm) mitotically active (>10 mitoses/50 hpf) GISTs are highly aggressive. Most small intestinal GISTs have mutation in exon 11 of C-KIT with few cases showing mutations in exons 9 and 17 of C-KIT . These mutations lack prognostic significance. PDGFRA mutations are rare in small bowel GISTs.


Colon . Tumors are most common in adults in the sixth decade of life in the ascending and descending portions of the colon and usually present with pain or a mass. Except for small subserosal lesions, they are typically transmural tumors with intraluminal and outward-bulging components. Morphologically they are heterogeneous, with spindle cells in fascicles, palisades or storiform arrangement, and sometimes an organoid pattern; a minority have epithelioid cells in varying proportions.


Moyana and colleagues and Tworek and associates have taken a size of 5 cm and 5 mitoses per 10 hpf as thresholds for malignancy, but Miettinen and coworkers found lower levels. In their series, tumors smaller than 1 cm did not recur, whereas in larger tumors 20% with minimal mitoses and all with more than 5 mitoses per 50 hpf metastasized or caused patient deaths. Interestingly, the few cases with skeinoid fibers had a better prognosis.


Appendix. GISTs of the appendix are rare. Miettinen and Sobin described four cases in men 56 to 72 years of age (mean 63 years). Two tumors occurred in patients who had undergone surgery for appendicitis-like symptoms: one was an incidental finding during surgery for a malignant gastric epithelioid GIST and one was an incidental autopsy finding. One was a polypoid GIST projected outward from the proximal part of the appendix. Three tumors were partially obliterating nodules, eccentrically expanding the appendiceal wall. All four were spindle cell tumors, and three of them contained extracellular collagen globules (skeinoid fibers); none had atypia or mitotic activity (< 1 mitosis per 50 hpf). Follow-up revealed death resulting from cardiovascular disease in one case (4 years after appendectomy) and liver failure because of malignant gastric epithelioid GIST metastatic to the liver in another case 15 years after the appendectomy.


Anorectum . These are rare tumors. St. Mark’s Hospital in London (a specialist lower GI institution) had only 25 “leiomyosarcomas” of the rectum in a 25-year period. Thirty-two percent to 54% of rectal GISTs are malignant, the smallest being 1.6 cm, but most exceed 5 cm and have greater than 5 mitoses per 50 hpf. In the series of Miettinen and colleagues of 133 anorectal GISTs, 54% recurred or metastasized (to liver, lung, or bone), sometimes after a long interval. Seventy percent of patients whose tumors were larger than 5 cm and with greater than 5 mitoses per 50 hpf died. One tumor smaller than 2 cm and with fewer than 5 mitoses per 50 hpf recurred, but no patients died. Eighteen of 29 cases (62%) had GIST-specific C-KIT mutations, mainly in exon 11. Survival rates are around 60% at 5 years and 20% to 50% at 10 years (for patients with curative rather than palliative resections).


Extragastrointestinal GISTs. These GISTs have been described by a number of observers. Some data suggest that single omental GISTs unattached to the GI tract resemble gastric lesions, raising the possibility that they are gastric GISTs that have extended into the omentum. In contrast, multiple omental GISTs are more reminiscent of small bowel GISTs and thus may arise in the small bowel and metastases/extensions. Solitary examples are associated with a better prognosis than multiple ones. Kit reactive Cajal cells are absent in the omentum.


Treatment


Complete excision is the initial treatment. Gene product-targeted therapy, utilizing STI571, a 2-phenylaminopyrimidine derivative that is a selective inhibitor of C-KIT , and PDGFR tyrosine kinases has been ongoing since 2002. The first case report was published in 2001. The initial response is rapid and relatively free of side effects. Treated tumors show small pyknotic nuclei in an eosinophilic myxoid background, unusual pleomorphic cells, or even heterologous differentiation (e.g., rhabdomyosarcomatous differentiation). Tumors with kit exon 11 mutations are more likely to respond to therapy than those with exon 9 or exon 13 mutations. Those with PDGRA mutations typically do not respond. Additional tyrosine kinase inhibitors are available (e.g., sunitinib malate) for unresponsive lesions, some of which have acquired resistance to imatinib.





Smooth muscle tumors of the gastrointestinal tract


Clinical features


Smooth muscle tumors of the GI tract consist of leiomyoma and leiomyosarcoma. Clinically, smooth muscle neoplasms of the GI tract often present as polypoid lesions extending into the gut lumen. Colonic leiomyomas classically arise in association with the muscularis mucosae and present as distal polyps on screening colonoscopy. GI smooth muscle tumors can also arise as intramural masses associated with the muscularis propria. In general, esophageal leiomyomas often present as intramural masses while leiomyomas and leiomyosarcoma of the colon often have a polypoid configuration. In the esophagus, the vast majority of spindle cell tumors are true leiomyomas—even the AFIP was only able to collect 17 GISTs from the esophagus. Leiomyoma of the esophagus is found in patients with a median age of 35 years with a male (about 70%), and has an indolent course. Leiomyosarcoma of the esophagus is truly rare and it follows an aggressive course. Smooth muscle tumors of small intestine are rare; it is estimated that one smooth muscle tumor occurs for every 36 GISTs. Both leiomyoma and leiomyosarcoma can be encountered throughout the small intestines of adults, presenting as a polypoid or intramural masses. Leiomyoma and leiomyosarcoma in the colon commonly present as polyps. Leiomyoma of the colon has male predominance of 2.4:1, occurs at a median age of 62 years old, and follows a benign course. Leiomyomas can occur extramurally adjoining the colon where they resemble uterine leiomyoma and even express hormone receptors. Leiomyosarcoma of the colon shows a slight male predominance and presents in adults. Leiomyomatosis has been described in small intestine and colon. True leiomyosarcoma is rare in the stomach.




SMOOTH MUSCLE TUMORS OF THE GASTROINTESTINAL TRACT—FACT SHEET


Definition





  • Smooth muscle tumors of the GI tract are spindle cell tumors showing differentiation along smooth muscle lines



Incidence and location





  • These are rare tumors, but leiomyomas are the most common spindle cell tumor of the esophagus



  • Leiomyomas also arise in association with the muscularis mucosae of the colorectum



  • In the remainder of the GI tract, true smooth muscle tumors are far outnumbered by gastrointestinal stromal tumors but tend to be malignant (leiomyosarcomas)



Morbidity and mortality





  • Leiomyomas are benign; approximately 75% of patients with leiomyosarcomas of the GI tract die of their tumors



Gender, race, and age distribution





  • No gender or race preference



  • Occurs in adults (average 60 years)



Clinical features





  • Smooth muscle tumors of the esophagus often present as intramural mass while smooth muscle tumors of the colon usually present as polypoid lesions



  • Smooth muscle tumors of small intestine can present as either intramural mass or polypoid lesions



Prognosis and therapy





  • Poor prognosis for leiomyosarcomas; leiomyomas are benign



  • All lesions except small polyps of colorectum (leiomyomas) are treated surgically; there is no targeted chemotherapy as there is for GISTs and the tumors have no characteristic mutations




Radiologic features


The radiologic features are not specific and are not distinguishable from those of GIST.


Pathologic features


Gross findings


The gross appearance of these tumors is nonspecific. Esophageal leiomyomas form whorled masses similar to uterine leiomyomas. The occasional leiomyomas arising in the muscularis mucosae of the colorectum present as polyps. Leiomyosarcomas of the colorectum have been described as intraluminal bulging polypoid masses.


Microscopic findings


True smooth muscle tumors have the same features of smooth muscle differentiation seen elsewhere in the body, namely perpendicularly oriented fascicles of spindle cells with brightly eosinophilic cytoplasm with longitudinal striations and blunt-ended nuclei with paranuclear vacuoles ( Figs. 7-10 through 7-14 ). Leiomyosarcoma is characterized by mitoses, cytologic atypia, and necrosis.




FIGURE 7-10


This gastric leiomyoma has arisen in association with the muscularis mucosae. It is brightly eosinophilic at low magnification.



FIGURE 7-11


This distal colonic leiomyoma is hypocellular and merges with the residual muscularis mucosae. The nuclei are blunt ended.



FIGURE 7-12


Leiomyosarcoma showing perpendicularly oriented fascicles of spindle cells with striking cytoplasmic eosinophilia (A). Compare the cellularity of this lesion (B) to that seen in Figure 7-11 .



FIGURE 7-13


This leiomyosarcoma is similar to the one depicted in Figure 7-12 A, but has more striking nuclear alterations.



FIGURE 7-14


Leiomyosarcoma. The nuclei have blunt ends and there are scattered paranuclear vacuoles. The cytoplasm has delicate longitudinal striations.




SMOOTH MUSCLE TUMORS OF THE GASTROINTESTINAL TRACT—PATHOLOGIC FEATURES


Gross findings





  • Not specific; esophageal leiomyomas are whorled whitish masses



  • Leiomyosarcomas in other sites are mural and variably necrotic, and usually well marginated



Microscopic findings





  • Spindle cells with perpendicularly oriented fascicles of brightly eosinophilic spindle cells with blunt-ended cigar-shaped nuclei and sometimes paranuclear vacuoles



  • Abundant mitosis, cytologic atypia, and necrosis



Immunohistochemistry





  • Positive for desmin, SMA, calponin, caldesmon



  • Negative for CD117 and S-100 protein



  • Occasional focal keratin common in leiomyosarcoma



  • Usually negative for CD34



Differential diagnosis





  • GIST primarily, which is CD117 positive and desmin negative, and nerve sheath tumors (positive for S-100)




Ancillary studies


Ultrastructural findings


The ultrastructural features are those of smooth muscle differentiation elsewhere (thin filaments, pinocytotic vesicles, attachment plaques, and interrupted external lamina).


Immunohistochemistry


True smooth muscle tumors are actin and desmin reactive and lack CD117/C-kit. Calponin and h-caldesmon are expressed and CD34 is typically absent. S-100 protein is absent. Some lesions display keratin expression, a feature of leiomyosarcomas in general.


Differential diagnosis


The differential diagnosis is primarily with GIST and nerve sheath tumors, and the majority of lesions are readily separable based on immunohistochemistry.


Prognosis and therapy


The prognosis of leiomyomas is excellent in all sites because they are benign. In general, GI tract leiomyosarcomas have a poor prognosis, and about 75% of patients die of their tumors.





Schwannomas


Clinical features


Most schwannomas occur in the stomach, involving the submucosa and muscularis propria. However, as a result of more widespread screening colonoscopy, colonic schwannomas are encountered with increasing frequency in daily practice.




SCHWANNOMAS—FACT SHEET


Definition





  • Gastrointestinal schwannomas are benign nerve sheath tumors that differ from those typically found in the peripheral nervous system by lacking capsules.



Incidence and location





  • These are rare and usually affect the stomach, where they are mural



  • In the colon, schwannomas often present as polypoid lesions



Morbidity and mortality





  • Gastrointestinal schwannomas are benign and typically not syndromic



Gender, race, and age distribution





  • No known gender/race predominance



  • Present in adults



Clinical features





  • Site specific, often incidental



Prognosis and therapy





  • Benign and treated by simple excision




Radiologic and endoscopic features


Gastric tumors are not distinguishable from GISTs. Colonic schwannoma presents as a polypoid lesion ranging from 0.5 to 1.2 cm centered in the mucosa, submucosa, or muscularis propria.


Pathologic features


Gross findings


Schwannomas are grossly similar to GISTs, having a whitish cut surface and well-circumscribed outline without a capsule.


Microscopic findings


GI tract schwannomas are not encapsulated, a feature that distinguishes them from schwannomas in the peripheral nervous system. They have a lymphoid cuff with germinal centers, and intralesional lymphocytes can be seen. They are composed of interlacing bundles of spindle cells that are only loosely palisaded (in contrast to GISTs, which, ironically, often display striking palisading). They appear similar to GISTs, but the lymphoid cuff is a tip-off that these are schwannian ( Figs. 7-15 through 7-18 ). In the colon, schwannomas can be classified into two types: spindle cells and plexiform. Schwannoma with spindle cell features are composed of spindle cells with wavy elongated nuclei with tapering ends, scanty eosinophilic cytoplasm, and indistinct cell borders. Schwannomas with plexiform features form nodules of spindle cells.




FIGURE 7-15


Gastric schwannoma. Note the striking lymphoid cuff at low magnification. This is a helpful diagnostic feature in separating these tumors (always benign) from GISTs (which may behave in a malignant fashion).



FIGURE 7-16


If this gastric schwannoma were mistaken for a GIST, the nuclear variability would result in an interpretation of a GIST of uncertain potential. The lymphoid cuff, as well as scattered intratumoral lymphocytes, are tip-offs that this is instead a benign schwannoma.



FIGURE 7-17


Gastric schwannoma. At intermediate magnification there is a mild inflammatory backdrop within the lesion.



FIGURE 7-18


Gastric schwannoma. At high magnification the tapered ends of the nuclei are apparent.




SCHWANNOMAS—PATHOLOGIC FEATURES


Gross findings





  • Well marginated but not encapsulated, in contrast to most schwannomas



  • Generally mural in the stomach and polypoid in the colon



Microscopic findings





  • Unencapsulated infiltrative spindle cell lesion with admixed plasma cells and scattered lymphocytes



  • Minimal palisading



  • Striking lymphoid cuff at periphery of tumor



  • Spindle and plexiform patterns seen in the colon



Immunohistochemistry





  • Strong diffuse S-100 protein



  • May be positive for CD34



  • Negative muscle markers



  • Negative for CD117



Differential diagnosis





  • Primarily GIST (usually negative for S-100, positive for CD117/C-kit), which lacks the lymphoid cuff



  • Smooth muscle tumors (SMA- and desmin-positive), which have elongated blunt-ends nuclei



  • Neurofibroma, which usually has wiry collagen background and can be associated with NF-1



  • Benign fibroblastic polyp of the colon/perineurioma (negative for S-100, variable EMA), which has bland oval nuclei




Ancillary studies


Immunohistochemistry


These benign tumors are strongly labeled with S-100 protein ( Fig. 7-19 ) and low-affinity nerve growth factor receptor (p75). Schwannomas can show labeling with C34, glial fibrillary acidic protein (GFAP), and collagen type IV with a pericellular pattern. Calretinin is unusually negative in GI tract “schwannomas” (in contrast, schwannomas of the somatic soft tissues typically express calretinin). The tumors lack muscle markers and CD117.


Mar 12, 2019 | Posted by in GASTROENTEROLOGY | Comments Off on Gastrointestinal mesenchymal tumors

Full access? Get Clinical Tree

Get Clinical Tree app for offline access