Fact-checked by Grok 2 weeks ago

Leiomyosarcoma

Leiomyosarcoma is a rare and aggressive form of cancer that originates in the cells lining the walls of hollow organs, such as the intestines, , , and blood vessels, as well as the in women. It belongs to the broader category of sarcomas, accounting for approximately 10-20% of all such tumors, and typically affects adults over the age of 50, with a slight predominance in females for certain sites like the and retroperitoneum. These tumors grow rapidly—potentially doubling in size within a month—and can metastasize through the bloodstream to distant sites like the lungs or liver, making early detection challenging as they often remain asymptomatic until advanced. Symptoms of leiomyosarcoma vary depending on the tumor's location and size but commonly include a noticeable lump or swelling under the skin, persistent pain, unintentional , , , or abnormal in cases involving the or . The exact causes remain unknown, though the disease arises from genetic mutations in cells that lead to uncontrolled growth; it is not typically hereditary but is associated with prior exposure to and rare genetic s such as Li-Fraumeni syndrome (involving TP53 mutations) or hereditary (involving RB1 deletions). Leiomyosarcomas most frequently develop in the (including the retroperitoneum), , or extremities like the thighs, with uterine cases often presenting in perimenopausal women. Treatment primarily involves surgical resection to remove the tumor, which offers the best chance for if the cancer is localized and completely excised, often combined with to prevent recurrence or for larger, higher-, or metastatic tumors. depends on factors such as tumor , , , and at ; five-year rates are approximately 63% for localized , 36% for regional spread, and 14% for distant , with optimal outcomes achieved at specialized high-volume centers. Ongoing research explores targeted therapies and clinical trials to improve management, given the tumor's complex genetic profile, including frequent alterations in genes like RB1 and PTEN.

Definition and Classification

Characteristics

Leiomyosarcoma is a rare and aggressive malignant tumor originating from cells, classified as a that accounts for approximately 10-20% of all such sarcomas. It exhibits smooth muscle differentiation and typically presents as a fast-growing mass with invasive potential. Histologically, leiomyosarcoma is defined by interlacing fascicles of spindle-shaped cells featuring cigar-shaped or blunt-ended nuclei, abundant cytoplasm, and varying degrees of pleomorphism. Key malignant indicators include a high mitotic rate, often exceeding 10 mitoses per 10 high-power fields, along with geographic and hypercellularity. These features distinguish it from benign tumors like leiomyomas. Tumors are graded using the French Federation of Cancer Centers Sarcoma Group (FNCLCC) system, which evaluates three parameters—tumor , mitotic count, and —to classify leiomyosarcomas as low (grade 1), (grade 2), or high (grade 3). Low-grade tumors show better and lower mitotic activity (<10 mitoses/10 HPF) with minimal , while high-grade ones demonstrate poor , elevated mitoses (≥20/10 HPF), and extensive (>50%). Leiomyosarcoma's aggressive biology leads to a substantial risk of local recurrence, reported in up to 40% of cases depending on site and , as well as distant primarily to the lungs, liver, and bones via hematogenous spread. This metastatic potential underscores its poor without comprehensive .

Subtypes

Leiomyosarcoma is classified into subtypes primarily based on anatomical origin, which influences its clinical behavior, , and . Uterine leiomyosarcoma is one of the most common subtypes, particularly in women, followed by those in the retroperitoneum, soft tissues of the , and other sites. Uterine leiomyosarcoma originates from the myometrial and represents the most common gynecological , accounting for 1-2% of all uterine malignancies. Gastrointestinal leiomyosarcoma develops from the of the and is a rare subtype, often presenting with spindle cells arranged in fascicles and variable pleomorphism. Cutaneous leiomyosarcoma arises from dermal or associated vascular structures in , typically manifesting as superficial tumors. Vascular leiomyosarcoma emerges from the walls of blood vessels, such as the , and tends to form large, infiltrative masses. Retroperitoneal leiomyosarcoma occurs in the retroperitoneal soft tissues, frequently growing to significant sizes before detection. Prognostic outcomes vary markedly by subtype, reflecting differences in tumor and detection timing. Cutaneous leiomyosarcomas exhibit better prognosis overall, with a rate of approximately 12% for dermal variants, attributed to their superficial location and lower aggressive potential. In contrast, retroperitoneal leiomyosarcomas are associated with poorer outcomes, including a 5-year overall of 30-40%, largely due to late detection from asymptomatic growth and challenges in achieving complete resection. Rare variants of leiomyosarcoma include the pleomorphic subtype, defined by more than 66% pleomorphic cells with a storiform and highly aggressive , and the myxoid subtype, characterized by over 50% myxoid stroma with fascicular or reticular and similarly aggressive clinical course.

Epidemiology

Incidence and Demographics

Leiomyosarcoma is a rare form of cancer, with an incidence estimated at approximately 0.5 to 1 case per individuals annually in high-income countries. It represents 10% to 20% of all sarcomas, which collectively account for less than 1% of adult malignancies worldwide. Global age-standardized incidence rates for soft tissue sarcomas were 1.16 per in 2021, varying by region and , with higher rates observed in high-income areas. The condition most commonly affects adults in middle to late adulthood, with peak incidence occurring between the ages of 50 and 70 years. Overall, there is a slight female predominance, primarily driven by the substantial proportion of cases originating in the ; in contrast, leiomyosarcomas at non-gynecological sites exhibit a more balanced distribution. Among uterine cases specifically, experience a higher incidence rate—approximately twice that observed in White women. In the United States, around 1,500 to 2,500 new cases are diagnosed each year, according to data from the Surveillance, Epidemiology, and End Results (SEER) program through 2023. Incidence rates have shown stability over the past several decades, with minimal fluctuations in age-adjusted rates reported in recent epidemiological analyses.

Risk Factors

Prior radiation therapy is a well-established environmental risk factor for developing leiomyosarcoma, particularly when administered to the pelvis for conditions such as cervical cancer, with tumors often emerging 10 or more years after exposure. Exposure to alkylating chemotherapy agents, such as those used in treating other malignancies, has also been linked to an elevated risk of secondary leiomyosarcoma, acting additively with radiation in susceptible individuals. Overall, patients with a history of radiation exposure face a 3- to 7-fold higher incidence of leiomyosarcoma compared to the general population. Genetic predispositions significantly contribute to leiomyosarcoma susceptibility, notably through inherited syndromes involving key tumor suppressor genes. Li-Fraumeni syndrome, caused by germline TP53 mutations, is associated with an increased risk of leiomyosarcoma among its broad spectrum of cancers. Similarly, hereditary due to RB1 mutations heightens the likelihood of developing leiomyosarcoma, particularly in survivors of treatment. Neurofibromatosis type 1, resulting from NF1 gene alterations, has been linked to rare cases of leiomyosarcoma arising in soft tissues. Other factors include rare associations with occupational exposure to , a chemical used in plastics manufacturing, which shows a positive correlation with mortality including leiomyosarcoma. In endemic regions, parasitic infections such as mekongi have been infrequently reported in conjunction with small bowel leiomyosarcoma. There are no strong established links between leiomyosarcoma risk and tobacco use or dietary factors.

Pathophysiology

Origin and Mechanism

Leiomyosarcoma arises from the neoplastic transformation of cells or their mesenchymal precursors, primarily in organs such as the , , or blood vessels, and does not originate from . In peripheral sites, many cases emerge from vascular , often within walls, while uterine forms typically derive from myometrial . Gastrointestinal leiomyosarcomas, though rare, originate from the or mural layers of the or colon. The pathogenic mechanism involves uncontrolled proliferation driven by loss of cell cycle regulation, leading to dedifferentiation of normal smooth muscle cells into malignant spindle-shaped cells with nuclear atypia and high mitotic activity. This process is characterized by genomic instability, including chaotic karyotypes and aneuploidy, resulting in intersecting fascicles of atypical cells and tumor necrosis. Dedifferentiation manifests as pleomorphic evolution, where well-differentiated smooth muscle areas transition to high-grade, undifferentiated components, complicating tumor behavior. The contributes to progression by fostering and , which promote local and . induces and alters cellular , while inflammatory cells enhance the aggressive ; hematogenous spread is facilitated through direct vascular by tumor cells. In rare instances, leiomyosarcoma develops via of benign leiomyoma-like lesions, with an estimated incidence of 0.13% to 0.29% among uterine fibroids undergoing surgery. This progression involves secondary genetic alterations in preexisting tumors, leading to aggressive sarcomatous features and potential , such as to the ovaries.

Genetic Alterations

Leiomyosarcomas exhibit a range of genetic alterations that contribute to their tumorigenesis, with frequent inactivation of tumor suppressor genes such as TP53 and RB1. Inactivation of TP53 occurs in approximately 50-60% of cases, often through mutations or deletions, leading to impaired DNA damage response and genomic instability. Similarly, RB1 loss is observed in about 50% of tumors, typically via homozygous deletions on 13q, disrupting regulation. In uterine leiomyosarcomas specifically, MED12 mutations occur in approximately 20% of cases and may drive smooth muscle cell proliferation through Wnt/β-catenin pathway dysregulation. Chromosomal abnormalities in leiomyosarcomas are characterized by complex karyotypes featuring widespread and multiple structural rearrangements, reflecting genomic . Common numerical changes include or partial deletions of 1p, seen in over 50% of cases, which may involve loss of tumor suppressor loci. Deletions in 10q and 13q are also prevalent, occurring in 60-75% and 50-75% of tumors, respectively, often encompassing PTEN on 10q and RB1 on 13q, further promoting oncogenesis. These alterations contribute to aggressive tumor behavior and are detected more frequently in advanced disease stages. Dysregulation of key signaling pathways underscores the molecular heterogeneity of leiomyosarcomas, with the PI3K/AKT/ pathway frequently activated due to PTEN loss or upstream signaling aberrations, enhancing cell survival and proliferation. The MAPK/ERK pathway is similarly implicated, often through feedback activation following PI3K inhibition or direct mutations, promoting tumor growth and . In a subset of cases, overexpression of and CDK4, driven by , inhibits and RB1 functions, respectively, amplifying cell cycle progression and resistance. Gene expression profiling has revealed molecular subtypes of leiomyosarcomas, including those with myogenic signatures reflecting differentiation and inflammatory subtypes marked by immune cell infiltration and expression. These subtypes, such as inflammatory versus myogenic, correlate with , where inflammatory profiles may indicate better responsiveness to but variable overall survival. Such classifications aid in stratifying patients for targeted therapies based on underlying oncogenic programs.

Clinical Features

Signs and Symptoms

Leiomyosarcoma often remains in its early stages, particularly when small, and is frequently discovered incidentally during or procedures performed for unrelated issues. As the tumor enlarges, patients commonly report a firm, painless lump or swelling at the site of origin, which may become tender or painful over time. General systemic symptoms in advanced disease include , unintentional , fever, , and , often resulting from tumor burden or . Site-specific presentations vary depending on the tumor's location and size. For intra-abdominal leiomyosarcomas, , , and gastrointestinal complaints such as or changes in bowel habits are typical, potentially leading to from chronic . Uterine leiomyosarcomas frequently cause abnormal , , or pressure, sometimes mimicking benign fibroids. In gastrointestinal cases, symptoms may include obstructive signs like or due to bowel involvement, while cutaneous leiomyosarcomas can present with ulceration or a growing nodule. Systemic effects beyond local symptoms are uncommon but can include secondary to ongoing bleeding in accessible sites like the or . Paraneoplastic syndromes are rare, with isolated reports of associated with certain leiomyosarcomas due to tumor secretion of insulin-like growth factors.

Sites of Occurrence

Leiomyosarcoma primarily arises from cells in various anatomical locations throughout the body. The is the most common site, accounting for 25-30% of all cases, predominantly in women of reproductive or perimenopausal age. The retroperitoneum represents approximately 20% of cases, often involving the behind the peritoneal lining. and other soft tissues contribute about 15% of occurrences, typically presenting as masses in the limbs or trunk. The accounts for around 10% of cases, with tumors commonly originating in the or . The remaining cases involve the head and neck region or vascular structures, such as large blood vessels like the . Overall, about 50% of leiomyosarcomas develop in the or , reflecting the abundance of in these areas. Primary tumors in the heart or lungs are exceedingly rare, comprising less than 1% of cases due to the limited presence in these organs. Site-specific implications influence and ; retroperitoneal tumors frequently grow large, often exceeding 10 cm at presentation, because the spacious retroperitoneal area allows expansion without early symptoms. In contrast, extremity tumors are more readily detectable early due to visible swelling or palpable masses, facilitating timely intervention. Metastasis in leiomyosarcoma occurs primarily via hematogenous spread, with the lungs being the most frequent site, affected in up to 70% of advanced cases. Other common metastatic locations include the liver and bones, while peritoneal spread is also observed, particularly from abdominal primaries. Multifocal metastases are uncommon, occurring in fewer than 5% of patients at diagnosis.

Diagnosis

Imaging and Laboratory Tests

Imaging for leiomyosarcoma typically begins with for superficial or pelvic masses, which can identify a heterogeneous mass with possible vascular involvement via Doppler assessment. For characterization, (MRI) is the preferred modality, revealing tumors that are isointense to muscle on T1-weighted images, intermediate to hyperintense on T2-weighted images, and often showing areas of or hemorrhage as hyperintense regions on T2 with . Computed (CT) excels in staging by detecting metastases, particularly in the lungs, and demonstrates heterogeneous enhancement with central low-attenuation . -computed (PET-CT) assesses metabolic activity, with standardized uptake values (SUV) often exceeding 3 in malignant lesions, aiding in differentiation from benign counterparts and detection. Laboratory tests for leiomyosarcoma are nonspecific and supportive rather than diagnostic. Basic blood work may reveal due to chronic bleeding or elevated (LDH) as a marker of tumor burden, though these are not unique to the disease. No dedicated tumor markers exist, but in uterine cases, cancer antigen 125 (CA-125) levels can be elevated, correlating with advanced disease or peritoneal involvement. Staging follows the American Joint Committee on Cancer (AJCC) 8th edition system for soft tissue sarcomas, classifying tumors from stage I (low-grade, ≤5 cm, no ) to stage IV (distant ) based on tumor (T1-T4: ≤5 cm to >10 cm), nodal involvement (N0-N1), (M0-M1), and histologic grade (G1-G3 via FNCLCC criteria). This framework guides prognosis and management, with imaging integral to determining T, N, and M descriptors; however, definitive confirmation requires .

Histopathology and Differential Diagnosis

Histopathological diagnosis of leiomyosarcoma typically requires a core needle or excisional biopsy, revealing a malignant mesenchymal neoplasm composed of spindle-shaped cells arranged in intersecting fascicles with elongated, hyperchromatic nuclei and eosinophilic cytoplasm. Microscopic hallmarks include moderate to severe nuclear atypia, coagulative tumor cell necrosis, and a high mitotic rate, often exceeding 5-10 mitoses per 10 high-power fields (HPF). Immunohistochemical staining supports the diagnosis, showing positivity for smooth muscle markers such as smooth muscle actin (SMA), desmin, and h-caldesmon, while typically negative for cytokeratins, which helps exclude epithelial mimics. Diagnostic criteria for emphasize a combination of features: cytologic atypia with nuclear pleomorphism, mitotic activity greater than 5 mitoses per 10 HPF, and geographic , as established in criteria adapted from classifications. An elevated Ki-67 proliferation index, often exceeding 10%, further corroborates high proliferative activity and aids in distinguishing malignant from benign tumors. These criteria are applied judiciously, as tumors lacking all three features may be classified as smooth muscle tumors of uncertain malignant potential (STUMP). Differential diagnosis involves distinguishing leiomyosarcoma from benign and other malignant spindle cell tumors. , a benign counterpart, lacks significant , , and shows low mitotic activity (typically <5 mitoses per 10 HPF). Gastrointestinal stromal tumor (GIST) is differentiated by its positivity for c-KIT (CD117) and DOG1 on immunohistochemistry, contrasting with the desmin positivity and c-KIT negativity in leiomyosarcoma. Fibrosarcoma, now rarely diagnosed as a distinct entity, features collagen-rich stroma and lacks smooth muscle markers, while dedifferentiated liposarcoma can mimic leiomyosarcoma but is identified by MDM2 and CDK4 gene amplification via fluorescence in situ hybridization (FISH). Challenges in diagnosis arise from sampling errors in heterogeneous tumors, where biopsy may miss diagnostic areas, necessitating expert pathologic review and adjunct molecular testing such as MDM2 FISH to rule out liposarcomatous mimics.

Treatment

Surgical Management

Surgery serves as the cornerstone of treatment for localized leiomyosarcoma, with the primary objective being complete tumor resection to achieve negative microscopic margins, known as R0 resection, which is associated with improved local control and survival outcomes. Wide local excision is the standard approach, ideally incorporating margins of greater than 2 cm of normal tissue when anatomically feasible, though margins of 1-2 cm are often sufficient if combined with other modalities to preserve function and reduce recurrence risk. For extremity tumors, limb-sparing procedures are prioritized, focusing on neurovascular preservation while ensuring adequate resection to avoid amputation in most cases. Site-specific considerations guide surgical planning to optimize outcomes. In uterine leiomyosarcoma, the recommended procedure is total hysterectomy with bilateral salpingo-oophorectomy, though ovarian preservation may be considered in premenopausal women, without routine lymphadenectomy due to the low rate of nodal involvement (approximately 5-11%). For retroperitoneal leiomyosarcoma, multivisceral en bloc resection is typically required, involving removal of the tumor along with contiguous organs such as kidney, colon, or pancreas to achieve complete excision and prevent spillage or rupture. In the setting of metastatic disease, surgery may still play a role for select patients with limited oligometastatic burden. Pulmonary metastasectomy, often via wedge resection, is recommended for resectable lung metastases according to guidelines, potentially offering prolonged survival in favorable cases. Palliative debulking procedures can be employed to alleviate symptoms such as pain or obstruction in advanced disease where curative intent is not possible. Potential complications of leiomyosarcoma surgery include intraoperative hemorrhage due to the vascular nature of these tumors and postoperative wound infections, which occur in up to 40% of soft tissue sarcoma resections overall. Success in achieving R0 resection varies by site but reaches 70-80% in localized extremity and uterine cases when performed at specialized centers. Following successful surgery, adjuvant therapies may be considered for high-risk features to further reduce recurrence.

Adjuvant Therapies

Adjuvant chemotherapy for leiomyosarcoma is typically reserved for high-risk cases, such as high-grade tumors larger than 5 cm, following surgical resection. First-line regimens are doxorubicin-based, with combinations like doxorubicin plus demonstrating response rates of 10-30% in advanced settings, though adjuvant use aims to reduce recurrence risk rather than achieve tumor response. For uterine leiomyosarcoma specifically, gemcitabine plus has shown activity, with objective response rates up to 35% in metastatic disease, supporting its consideration in adjuvant contexts for this subtype. Radiation therapy is employed preoperatively or postoperatively to mitigate local recurrence in high-risk leiomyosarcoma, particularly for tumors in the extremities, trunk, or retroperitoneum. Postoperative radiation doses of 50-60 Gy are standard, often delivered over 5-6 weeks, and may improve local control by 10-20% in selected patients. For retroperitoneal leiomyosarcoma, intensity-modulated radiation therapy (IMRT) is preferred to spare surrounding organs and reduce toxicity while targeting the tumor bed effectively. Targeted therapies, including pazopanib and trabectedin, are primarily used for advanced or metastatic leiomyosarcoma but may be considered in adjuvant settings for high-risk disease. Pazopanib, a multi-tyrosine kinase inhibitor, extends progression-free survival to approximately 4.6 months in non-adipocytic sarcomas like leiomyosarcoma, compared to 1.6 months with placebo. Trabectedin, a marine-derived alkylating agent, achieves a median progression-free survival of 4.2 months in uterine leiomyosarcoma, outperforming dacarbazine. In a 2024 phase 3 trial, first-line doxorubicin plus trabectedin improved median overall survival to 32.3 months compared to 25.0 months with doxorubicin alone in patients with advanced leiomyosarcoma. Emerging immunotherapy approaches, such as PD-1 inhibitors like pembrolizumab, show promise in microsatellite instability-high (MSI-high) cases, with case reports indicating durable responses in this rare subset. Neoadjuvant therapies, combining chemotherapy and/or radiation, are utilized to shrink tumors prior to surgery in locally advanced leiomyosarcoma, facilitating resection. Doxorubicin-based neoadjuvant regimens can improve resectability, with modest overall survival benefits of 10-15% observed in high-grade cases, though results from large randomized trials have been mixed, with some meta-analyses indicating modest benefits in high-risk cases. These approaches are integrated with surgery to optimize outcomes in high-risk patients.

Prognosis

Survival Outcomes

Leiomyosarcoma exhibits variable survival outcomes depending on the extent of disease at diagnosis. For localized disease, the 5-year relative survival rate is approximately 60-63%. In contrast, metastatic leiomyosarcoma carries a poorer prognosis, with 5-year survival rates around 13-14%. For the uterine subtype specifically, the overall 5-year survival rate is around 38%. Stage-specific data from large registries highlight these differences. Patients with stage I or II disease achieve 5-year overall survival rates exceeding 70%, often around 76% for stage I and 60% for stage II based on Surveillance, Epidemiology, and End Results (SEER) program analyses. Stage III disease yields 5-year survival of 30-50%, approximately 36% in historical cohorts, while stage IV is associated with rates below 20%, typically 13-18%. These figures, drawn from SEER and European Society for Medical Oncology (ESMO)-aligned studies up to 2025, underscore the impact of early detection. Median survival times further illustrate the trajectory. For advanced disease with systemic therapy, median survival ranges from 12-18 months, though select low-risk metastatic cases may extend to 20 months or more. Recent advancements have yielded modest improvements in survival. A 2024 clinical trial showed that combining trabectedin with doxorubicin improved progression-free survival and overall survival compared to doxorubicin alone in patients with advanced . Targeted agents such as trabectedin and eribulin have demonstrated benefits in median overall survival for advanced cases in clinical trials, with eribulin extending median OS to 13.5 months versus 11.5 months with dacarbazine.

Factors Influencing Prognosis

Several clinical and pathological factors significantly influence the prognosis of leiomyosarcoma, with tumor size being a primary determinant. Tumors larger than 5 cm are associated with worse outcomes, including increased risk of recurrence and reduced disease-specific ; for instance, tumors between 5-10 cm carry a hazard ratio () of approximately 1.8 for disease-specific survival compared to those ≤5 cm, while tumors >10 cm have an of 3.4. Histologic is another critical predictor, with high-grade tumors (typically 2-3, comprising about 90% of cases) linked to poorer survival rates, such as a 5-year survival below 40% in high-grade variants, due to higher metastatic potential. Tumor depth also plays a key role, as deep-seated lesions (e.g., intramuscular or retroperitoneal) confer a worse than superficial ones, independent of size and , owing to challenges in achieving complete resection. Surgical margin status further impacts outcomes, with R1 or resections (microscopic or gross residual disease) increasing the risk of local recurrence approximately 2-fold (HR 2.1) compared to R0 margins. The anatomical location of the tumor modulates , primarily through effects on detection and feasibility. Retroperitoneal and uterine leiomyosarcomas generally have poorer outcomes than those in the , with 5-year -specific survival rates around 67% for abdominal/retroperitoneal sites versus 75-81% for or locations, largely attributable to delayed and more advanced presentation at detection. Presence of at initial substantially worsens survival, roughly halving 5-year rates (e.g., 14% for distant versus 63% for localized), as it indicates advanced stage IV with limited curative options. Patient-related factors, including age and , contribute to prognostic variability. Individuals over 60 years exhibit worse outcomes, with age >60 identified as a significant predictor of reduced survival in multivariate analyses. Poor (e.g., ECOG ≥2) correlates with diminished tolerance to aggressive therapies and shorter overall survival. At the molecular level, mutations such as TP53 are associated with aggressive behavior, including shorter disease-free survival and heightened metastatic risk. Recurrence remains a major challenge, with rates of 40-50% for local recurrence and about 30% for distant occurring within the first 2-3 years post-treatment, particularly in high-risk cases involving large or high-grade tumors.

Uterine Leiomyosarcoma

Specific Features

Uterine leiomyosarcoma (uLMS) represents the most common type of uterine , accounting for approximately 30-40% of all such malignancies, which collectively comprise about 3% of all uterine cancers. Despite its relative prevalence within this rare category, uLMS is frequently misdiagnosed preoperatively as benign uterine (leiomyomas), leading to unintended procedures such as morcellation during minimally invasive surgeries; the incidence of occult uLMS in presumed fibroid cases ranges from 0.12% to 0.57%, highlighting the diagnostic challenges posed by overlapping and clinical features. Clinically, uLMS often presents with symptoms that mimic benign conditions, including postmenopausal in over 50% of cases, rapid uterine enlargement, and pelvic or . Unlike many gynecologic malignancies, uLMS demonstrates an aggressive behavior with early dissemination; approximately 33% of patients have distant metastatic disease at initial diagnosis, commonly involving the lungs, liver, or . Histologically, while conventional spindle-cell uLMS predominates, myxoid and epithelioid variants are more frequently encountered in the compared to extra-uterine sites, comprising up to 10-15% of cases and often requiring specialized immunohistochemical markers like desmin and actin for confirmation. These tumors exhibit variable receptor expression, with and progesterone receptors present in 40-70% of cases, potentially influencing tumor growth and offering avenues for targeted therapies, though their prognostic value remains under investigation. Staging for uLMS follows the International Federation of Gynecology and Obstetrics (FIGO) system adapted for gynecologic sarcomas in 2009, which emphasizes tumor size, cervical involvement, and extrauterine extension to stratify risk; stage I disease (confined to the ) accounts for about 60% of presentations, yet even early-stage cases carry high recurrence risks. Notably, inadvertent morcellation during for presumed benign disease significantly worsens outcomes, with meta-analyses indicating a 62% recurrence rate compared to 39% in non-morcellated cases, alongside increased intra-abdominal spread (39% vs. 9%). This underscores the importance of preoperative risk stratification to avoid such procedures in high-suspicion scenarios.

Comparison with Leiomyomas

Uterine leiomyomas, commonly known as fibroids, are benign tumors that affect up to 70% of women of reproductive age, whereas uterine leiomyosarcomas (LMS) are rare malignant counterparts with an incidence of approximately 1 in 498 women undergoing for presumed fibroids. Leiomyomas are typically hormone-dependent, responding to and progesterone, and exhibit slow, predictable growth patterns confined to the uterine wall without invasion. In contrast, LMS are aggressive, infiltrative tumors independent of hormonal influence, often presenting with rapid enlargement that can distort or breach the uterine architecture. Histologically, the distinctions are critical for . Leiomyomas display low mitotic activity (fewer than 5 mitoses per 10 high-power fields [HPF]), minimal to no cytologic , and absence of coagulative tumor cell , maintaining a uniform fascicular arrangement of cells. LMS, however, are characterized by high mitotic rates (more than 10 mitoses per 10 HPF), moderate to severe nuclear with pleomorphism and hyperchromasia, and coagulative , often accompanied by hypercellularity and infiltrative borders. These features the benign nature of leiomyomas versus the malignant potential of LMS, though atypical variants like cellular or symplastic leiomyomas can mimic LMS and require careful evaluation to avoid misdiagnosis. Clinically, leiomyomas predominantly cause , pelvic pressure, and in premenopausal women due to their gradual expansion, rarely leading to rapid symptoms unless pedunculated or submucosal. LMS more frequently manifests with accelerated tumor growth, postmenopausal bleeding (occurring in about 59% of cases compared to 14% in leiomyomas), and systemic symptoms like pain or , often in perimenopausal or postmenopausal patients. This rapid progression in LMS contrasts sharply with the indolent course of fibroids, which regress postmenopause due to hormonal decline. Diagnostic challenges arise from overlapping imaging features, particularly on MRI, where both leiomyomas and LMS can appear as T2-hyperintense masses, especially with degeneration in fibroids mimicking the heterogeneity seen in sarcomas. This similarity complicates preoperative differentiation, contributing to the risk of inadvertent morcellation during minimally invasive procedures for presumed fibroids, which can disseminate occult LMS cells intra-abdominally. In 2014, the FDA issued a safety communication warning against power morcellation due to this risk, estimating that 1 in 225 to 1 in 580 women with fibroids may harbor an unsuspected uterine sarcoma. Such pitfalls highlight the need for enhanced preoperative , including advanced protocols, to distinguish these entities and prevent iatrogenic spread.