Gastrointestinal Stromal Tumors (GIST)

Gastrointestinal Stromal Tumors (GIST)

Gastrointestinal Stromal Tumors (GIST)

Diagnosis, Treatment, and Mnemonics for Clinicians

Gastrointestinal stromal tumors (GIST) are the most common mesenchymal tumors of the gastrointestinal (GI) tract, originating from the interstitial cells of Cajal, which regulate peristalsis. While GISTs can occur anywhere along the GI tract, the stomach (60%) and small intestine (30%) are the most frequent sites. Though relatively rare, comprising about 1-3% of GI malignancies, recognizing and managing GISTs is crucial due to their potential for malignancy.

Let’s break down the key aspects of GISTs: their pathophysiology, diagnosis, and treatment, with useful mnemonics to aid recall in clinical practice.

Pathophysiology of GISTs

Most GISTs result from activating mutations in the c-KIT proto-oncogene, encoding a tyrosine kinase receptor (CD117). A minority involves mutations in the platelet-derived growth factor receptor alpha (PDGFRA) gene. Both mutations drive uncontrolled cellular proliferation.

Mnemonic: “C-KIT Powers LAFRA ( lafra in hindi) 

– C-KIT mutation

– Platelet-derived Growth Factor Receptor Alpha (PDGFRA) mutation

Clinical Presentation

GISTs can remain asymptomatic for a long time or present with nonspecific symptoms. Typical symptoms depend on the tumor’s location and size and may include:

– Abdominal pain or discomfort

– GI bleeding (occult or overt)

– Fatigue due to anemia

– Early satiety or bloating

– Unintentional weight loss

Mnemonic: “BAG-A Tumor” (Symptoms of GIST)

– Bloating

– Abdominal pain

– GI bleeding

– Anemia

– Tumor mass effect (satiety, discomfort)

Diagnosis of GISTs

The diagnosis of GIST is confirmed through a combination of clinical evaluation, imaging, and histopathology. Key diagnostic tools include:

  1. Endoscopy and Imaging: Endoscopic ultrasound (EUS) or contrast-enhanced CT scans are often used to visualize the tumor and assess its size, location, and metastasis.
  1. Biopsy and Immunohistochemistry: Histopathology from biopsies typically reveals spindle-shaped or epithelioid cells. Immunohistochemistry is crucial, with over 95% of GISTs staining positive for CD117 (C-KIT) and around 60-70% for DOG1 (Discovered on GIST-1).
  1. Mutation Testing: Genetic testing to identify c-KIT and PDGFRA mutations guides targeted therapy decisions.

Must know facts about GIST

Treatment of GISTs

The treatment of GISTs hinges on the size, location, and risk of malignancy, but most tumors require surgical resection. However, the advent of tyrosine kinase inhibitors (TKIs) like imatinib has revolutionized the management of unresectable or metastatic GISTs. Imatinib targets the c-KIT mutation, which drives most GISTs.

  1. Surgical Resection: For localized tumors, surgery is the mainstay of treatment, often leading to curative outcomes if the entire tumor is removed without rupture.
  1. Targeted Therapy Imatinib is the first-line treatment for unresectable or metastatic GISTs, as well as adjuvant therapy in high-risk patients post-surgery. For those resistant to imatinib, second-line therapies include sunitinib and regorafenib.

Prognosis and Follow-Up

The prognosis for GIST depends on factors such as tumor size, mitotic rate, and the presence of metastasis. GISTs under 2 cm with low mitotic rates have an excellent prognosis. Larger or metastatic tumors, or those with a high mitotic index, require long-term follow-up and adjuvant therapy. Regular monitoring via imaging (CT or MRI) is critical to detect recurrence.

Mnemonic: “RISK” for prognosis

– Recurrence monitoring (via imaging)

– Imatinib (adjuvant therapy for high-risk patients)

– Size of the tumor

– KIT mutation status (guides therapy)

Conclusion

GISTs are rare but significant tumors that require a multidisciplinary approach for optimal management. Through a combination of surgery, targeted therapies, and vigilant monitoring, outcomes for patients with GIST have improved significantly.

#INICET #FMG #NEETPG #USMLE

Leave a reply