International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Leiomyosarcoma (LMS) is a rare malignant tumor caused by smooth muscle cells, accounting for ~5-10% of all soft tissue sarcomas (STS) (1,2). They are commonly originating in the uterus, retroperitoneum, gastrointestinal and genitourinary tract (3,4). LMS are known for aggressive behavior and high tendency towards metastasis, with ~30-40% of patients experiencing metastatic progression within the first 5 years after diagnosis (5). The typical metastatic sites for LMS include the lungs, liver and retroperitoneum (3). Cutaneous metastases are exceptionally rare, occurring in 1-2.6% of all cases (6,7), and can often signify advanced disease with poor prognosis.
The presentation of LMS with metastasis to the skin poses unique diagnostic and therapeutic challenges. Unlike primary cutaneous malignancies, skin metastases are secondary manifestations of systemic spread, reflecting the tumor's aggressive nature. These lesions often present as firm, painless nodules or plaques and may mimic benign dermatological conditions, leading to diagnostic delays (8). Early recognition of cutaneous metastases is crucial as it frequently indicates widespread disease and necessitates prompt systemic management.
The present review aims to provide a comprehensive analysis of retroperitoneal LMS (RPLMS) with metastasis to the skin, encompassing its pathophysiology, clinical presentation, diagnostic strategies and treatment options. Additionally, it highlights emerging therapeutic approaches and the prognostic implications of cutaneous involvement. By synthesizing existing knowledge, the present review seeks to enhance the understanding of this rare phenomenon and improve clinical outcomes for affected patients.
STS accounts for <1% of all cancers (9), of which ~40% are located in the abdomen or retroperitoneum (10). The most common abdominal and retroperitoneal STS are gastrointestinal stromal tumors (GIST), LMS and liposarcoma (LS) (11).
LMS is the second most common retroperitoneal sarcoma, with an incidence rate of about 20%, second only to LS (12). In the retroperitoneum, LMS is more commonly found in the vena cava and occasionally in the iliac vein or renal vein (13). In a prospective cohort study on abdominal and retroperitoneal sarcoma in Denmark, 11% of tumors were LMS (9).
RPLMS are also known for aggressive behavior and high tendency towards metastasis, with ~30-40% of patients experiencing metastatic progression within the first 5 years after diagnosis (5). The most common metastatic sites of RPLMS are the lungs, liver and vascular regions. Skin is a less common site of metastasis, with an incidence rate of only 1-2.6% (6,7). This type of metastasis may occur months or years after the diagnosis of the primary tumor and usually means advance stage and a poor prognosis (12). Previously, in the largest review of the reported cases at present, Wang et al (12) reported 65 patients with sarcoma with skin metastases, of which 43% were from LMS (12). LMS commonly affects individuals between the fifth and seventh decades of life, and while LMS overall shows a slight female predominance due to its association with uterine LMS, advanced LMS with skin metastases affects both sexes equally (12).
RPLMS lacks specific clinical manifestations. Most patients are asymptomatic, and the tumor is discovered incidentally. Symptoms such as upper abdominal pain or sore back pain can be more apparent when the tumor becomes big enough to invade surrounding tissues. According to Clark et al (14), the most common finding at diagnosis is a painless, gradually enlarging mass. Some patients primarily present with weight loss and abdominal pain, other with intestinal obstruction and dysphagia.
Cutaneous metastases are also usually asymptomatic. They are usually presented as painless firm, nodular lesions on the trunk, extremities, or scalp corresponding to areas of rich vascularization (8,15). Usually, the presence of cutaneous metastases is frequently accompanied by other systemic symptoms, due to late stage of the disease.
While preoperative diagnosis is very difficult, and most patients are diagnosed by intra-operative exploration and postoperative pathology, imaging plays a critical role in evaluating LMS with skin metastasis. Ultrasound is effective for superficial lesion characterization. On Ultrasound, the mass may appear as hypo echoic nodules with irregular borders. Computed tomography (CT) scan and magnetic resonance imaging (MRI) provide detailed insights into lesion extent and systemic involvement. The National Comprehensive Cancer Network guidelines for intraabdominal and retroperitoneal STS, recommends CT of the chest, abdomen and pelvis with intravenous contrast for diagnosis, occasionally supplemented by MRI of lesions in the pelvis or abdomen. On the CT scan, skin metastases appear as soft tissue nodules within dermis or subcutaneous tissue and may show enhancement after contrast administration (16). MRI is useful for evaluating the extent of skin and soft tissue involvement. Positron emission tomography is invaluable for detecting and monitoring metastatic disease. A multi-modal imaging approach, integrated with clinical and histopathological findings, ensures accurate diagnosis and optimal management of this rare metastatic presentation.
Histopathology is the gold standard for diagnosing LMS and confirming skin metastases. The histopathological examination provides insights into the tumor's morphology, immunohistochemical profile, and differentiation from other malignancies. According to the European Society for Medical Oncology-European Reference Network for rare adult solid cancers report on soft tissue and visceral sarcomas from 2018, all retroperitoneal tumors should be biopsied. The risk of needle track seeding is minimal, if the biopsy is thoroughly planned, and not performed trans-peritoneally (11).
LMS typically shows fascicles of spindle-shaped tumor cells with blunt-ended nuclei and moderate to abundant, brightly eosinophilic fibrillary cytoplasm (17). Useful immunohistochemical markers for retroperitoneal sarcomas include smooth muscle actin (SMA), Desmin, Myogenin, CD34, S100 protein, MDM2, CDK4, STAT6, ALK, CD99, H3K27me3, NKX2.2, TLE1, SOX10, melanocytic markers, cyclin D1, and epithelial markers.
In LMS, at least one myogenic marker (that is, SMA, Desmin, or Caldesmon) is positive, with >70% of cases showing positivity for more than one of these markers. Tumor cells are also positive for cytokeratin and endomysial antibodies in ~40% of cases (18). In a retrospective study of 29 consecutive cases of histologically proven soft tissue LMS by Mestiri et al (19), they found that tumor cells were positive for SMA (79.3%), desmin (68.9%), and h-Caldesmon (58.6%). They were negative for PS100, CD34 and c-Kit in all cases (19).
Mitotic count is also an important histological factor in advance LMS (20). In a recent study by Grimaudo et al (21), which analyzed 121 patients with LMS between June 1999 to May 2022, it was found that mitotic count is an important prognostic factor in early and advanced LMS. In a previous study by Rodríguez-Lomba et al (22), in total of 17 LMS in 12 patients (5 men, 41%; 7 women, 59%), histology found a mean of 11 mitotic figures per 10 high-power field (HPF) and Ki-67 protein expression of 30% in cutaneous tumors; 7.5 mitotic figures per 10 HPF and Ki-67 expression of 60% in subcutaneous forms; and 10.2 mitotic figures per 10 HPF and Ki-67 expression of 48% in metastatic forms. Immunohistochemical staining was positive for Vimentin (>25% of cases), Desmin (>50%), HHF-35 (>75%) and SMA (>75%). S100 staining was focally positive in one cutaneous LMS. CD34 staining was also positive in one cutaneous tumor.
It is vastly agreed that Desmin, SMA and Caldesmon staining are positive in a vast majority of LMS cases. However, a positive CD34 in patients with LMS remains uncommon, and its role in prognosis of these patients remains controversial (23,24). CD34 expression is more commonly associated in patients with LMS originated from vascular areas. In the study of Rodríguez-Lomba et al (22), only one patient was CD34-positive in 17 patients with LMS. Sugiura et al (25) studied the prognostic value of CD34 expression status in 192 patients with myxofibrosarcomas and undifferentiated pleomorphic sarcomas (UPS). Of the 192 patients, 32 patients were positive for CD34. They found that CD34-positive patients (n=32) had a significantly improved overall and distant metastasis-free survival compared with CD34-negative patients (n=160) (P<0.001 and P=0.027, respectively), although local recurrence-free survival was not significantly influenced by their CD34 status.
Histopathology remains to be challenging and may overlap with other tumors, including dermatofibrosarcoma protuberans (DFSP), vascular sarcoma and GIST. The key differential diagnosis between LMS and DFSP includes spindle shaped arrangement of spindle cells, but lack of SMA or histone positivity. In vascular sarcoma, it shows vascular channels arranged by atypical endothelial cells, with CD31 and CD34 positive but SMA negative. Malignant melanoma may mimic LMS, but S-100 and HMB-45 are positive, unlike LMS. In GIST, SMA and CD34 are positive, but CD117 and DOG1 are negative.
Treatment of RPLMS with skin metastasis is often challenging, as the presence of cutaneous lesions indicates advanced, systemic disease (12). The therapeutic approach focuses on systemic control of the primary tumor and its metastases while addressing local skin lesions for symptomatic relief or cosmetic reasons.
Chemotherapy is the first line for advanced LMS with skin metastasis. Doxorubicin is the first-line chemotherapeutic agent for advanced LMS (26). Previously, Judson et al (27), compared the efficacy of monotherapy with doxorubicin (A) and doxorubicin + ifosfamide (AI) regimen in the treatment of patients with advanced STS. The results showed that the objective response rate (ORR) of the AI group was markedly higher than that of monotherapy A group (26% vs. 14%, P<0.006), and the median progression-free survival (mPFS) was also higher than that of monotherapy A group (7.4 months vs. 4.6 months, P=0.003), but the difference in overall survival (OS) between the two groups was not statistically significant (14.3 months vs. 12.8 months, P=0.076). Stratified analysis showed that except for the UPS subgroup which showed significant OS benefits, no other subtypes showed significant OS benefits, and the incidence of adverse reactions was higher with combination therapy.
Similarly, in a retrospective study by D'Ambrosio et al (28), which evaluated the efficacy of first-line treatment with doxorubicin + dacarbazine (AD), doxorubicin + ifosfamide (AI), and doxorubicin (A) for advanced metastatic LMS. In their study, 117 patients (39%) received AD treatment, 71 cases (23%) received AI treatment, and 115 cases (38%) received (A) treatment. Among the 205 patients matched with a 2:1:2 propensity score, the mPFS of the three groups were 9.2 months, 8.2 months, and 4.8 months, respectively, and the ORRs were 30.9, 19.5 and 25.6%, respectively. The mPFS of the AD group was significantly improved compared with that of the A group (HR=0.72, 95% CI 0.52-0.99). The median OS of the three groups were 36.8, 21.9 and 30.3 months, respectively, with no significant difference. In the population with propensity score matching, doxorubicin + dacarbazine showed favorable efficacy in ORR and PFS and is worthy of further evaluation in prospective trials.
In another randomized phase 2 study, the efficacy of gemcitabine + dacarbazine and dacarbazine monotherapy was compared in previously treated patients with STS. The results showed that the median PFS of the gemcitabine + dacarbazine group was 4.2 months, while the dacarbazine monotherapy group was 2 months. The median OS was 16.8 and 8.2 months, respectively, with ORRs of 49 and 25% (29).
Furthermore, in a phase 3 trial called GeDDiS, the efficacy of gemcitabine + docetaxel versus doxorubicin as first-line treatment was compared in advanced unresectable or metastatic STS (including LMS). The results showed that the two regimens were equally effective. For gemcitabine + docetaxel and doxorubicin, the mPFS was 5.5 months and 5.3 months, the OS was 14.5 months and 16.3 months, and the ORR was 20 and 19%, respectively (30).
In a multicenter, open label, and efficacy randomized phase III clinical trial from January 18, 2017 to March 21, 2019, Pautier et al (31), included 150 patients with metastatic or recurrent unresectable LMS, among which were 67 uterine LMS, and 83 soft tissue LMS from 20 centers in the French Sarcoma Group who had not previously received chemotherapy. Randomly allocate (1:1) to receive doxorubicin (75 mg/m2) every 3 weeks for a maximum of 6 cycles; Or doxorubicin (60 mg/m2) + qubetidine (1.1 mg/m2, d1), once every 3 weeks for a maximum of 6 cycles, followed by maintenance treatment with qubetidine alone. There were 76 cases in the doxorubicin group and 74 cases in the doxorubicin + qubetidine group. The results showed that the mPFS of the doxorubicin + qubetidine group was significantly improved compared with that of the doxorubicin group, at 12.2 and 6.2 months, respectively (HR=0.41; 95% CI: 0.29-0.58; P<0.0001). The most common grade 3-4 adverse events were neutropenia, with incidence rates of 12 and 20% in the two groups, respectively.
In terms of targeted therapy, pazopanib is a multi-tyrosine kinase inhibitor that targets vascular endothelial growth factor, platelet-derived growth factor, and c-kit pathways. Pazopanib is approved for non-adipocytic STS, including LMS, and helps stabilize the disease. In a randomized phase III study called PALETTE, the median PFS of the pazopanib group was significantly improved compared with that of the placebo group (4.6 vs. 1.6 months). However, there was no significant difference in OS between the two groups (12.5 vs. 10.7 months), and only 6% of patients experienced objective remission (32).
Anlotinib is another targeted therapy. In a randomized controlled phase IB study (ALTER0203), compared with placebo, anlotinib significantly prolonged PFS and reduced the risk of disease progression (6.27 vs. 1.47 months; HR=0.33; P<0.0001). Subgroup analysis showed that anlotinib significantly prolonged the PFS of patients with various sub-types of LMS (5.83 vs. 1.43 months, P<0.0001) (33). In a retrospective analysis, the ORRs of patients with advanced LMS receiving combination therapy with anlotinib was 19%, the disease control rate (DCR) was 88%, and the median PFS was 8.8 months. The ORR did not show significant improvement, but the DCR and recent survival rate were satisfactory (34).
Regorafenib is another targeted therapy that showed positive results in treatment of advanced sarcomas. In the Phase II REGOSARC clinical study, which included 182 patients with advanced STS, grouped according to pathological types, and observed the efficacy and safety of regorafenib in different pathological types, the results demonstrated that for patients with STS who had previously received anthracycline treatment, regorafenib significantly improved PFS in subgroups of LMS, synovial sarcoma, and other sarcomas (35).
In terms of immunotherapy, immunotherapy with immune checkpoint inhibitors (ICIs) PD-1/PD-L1 antibodies has shown effectiveness in various tumors and has also been attempted in STS. However, currently, the efficacy of immunotherapy alone in LMS is not significant (36-38). In addition, DNA damage repair pathway inhibitors-related studies have found that drugs targeting the DNA damage repair pathway may become potential therapeutic targets.
Combination therapies such as combining chemotherapy, targeted therapy and immunotherapy are promising. In A single arm open phase II clinical trial (NCT04028063) of doxorubicin combined with CTLA-4 inhibitor zalifrelimab and PD-1 inhibitor balstilimab in patients with advanced and metastatic STS, patients with advanced and metastatic sarcoma who had not previously received doxorubicin or ICIs were included. All patients received up to 6 cycles of doxorubicin and received zalifrelimab + balstilimab simultaneously. In the first stage, patients received zalifrelimab + balstilimab treatment for one cycle, and doxorubicin began in the second cycle. In the second stage, the patients used doxorubicin + zalifrelimab + balstilimab simultaneously in the first cycle. A total of 35 patients were enrolled. PFS 6 months was ~46.4%, and mPFS was 24.4 weeks. The ORR was 33.3%, and the DCR was 80.0%. The median duration of remission was 12.8 weeks. Therapeutic effects have been observed in endometrial sarcoma, vascular sarcoma, malignant peripheral nerve sheath tumor, LP, LMS, endometrial stromal sarcoma, UPS and sclerosing epithelioid fibrosarcoma. Compared with patients in the second stage, patients in the first stage showed improved efficacy, including PFS 6 months (56.3 vs. 25.0%), mPFS (31.7 vs. 25.3 weeks), ORR (56 vs. 8.3%) and DCR (94 vs. 0.75%) (39).
Another NitraSarc trial explored the efficacy and safety of Nivolumab and Tribetidine in advanced STS. The study prospectively and non-randomly enrolled patients in Group A (advanced LP, LMS; L-type tumor) and Group B (non-LP, non-LMS; non-L-type tumor). Treatment was divided into late combination cohort (LCC) and early combination cohort (ECC) based on the timing of combination. All patients were first treated with 1.5 mg/m2 of tramadol, followed by 3 cycles of combined treatment with 240 mg of nivolumab, known as LCC. Starting from the second cycle, receiving combination therapy was called ECC. The primary endpoint was PFS at 6 months. A total of 92 patients were included, with 43 in Group A and 49 in Group B. After a median follow-up of 16.6 months, the overall PFS at 6 months of Group A was 47.6% (LCC was 60%, ECC was 36.4%), while the overall PFS at 6 months of Group B was 14.6%. The median PFS of group A was higher than that of group B (5.5 vs. 2.3 months), and the LCC group was longer than the ECC group (9.8 vs. 4.4 months). The median OS of Group A was longer than that of Group B (18.7 vs. 5.6 months), and LCC was longer than ECC (24.6 vs. 13.9 months) (40).
ImmunoSarc2 study, a first-line treatment of advanced LMS with doxorubicin, dacarbazine, and nivolumumab - a phase Ib clinical trial in Spanish sarcoma tissue (GEIS), included adult patients with advanced and metastatic LMS who had an Eastern Cooperative Oncology Group score of 0-1 and had not previously received anthracycline treatment. A total of 20 subjects were included. Among the 16 subjects with assessable efficacy, 9 (56.2%) had partial response (PR), 6 patients (37.5%) had partial response and 2 patients had tumor shrinkage greater than 20%, and 1 (6.3%) had disease progression. The mPFS was 8.67 months, with a median follow-up time of 8 months (41).
In term of surgical treatment, surgical treatment and complete excision with clear margins is the ideal choice in early stages, and surgical removal of isolated skin metastases is only recommended for skin tumor that are causing pain, ulceration, or functional impairment or for pathological reason.
In term of prognosis, advanced RPLMS have a bad prognosis, with frequent metastasis and an overall 5-year survival rate of 30-40% (5). LMS of vascular origin have the worst prognosis with metastasis at the time of diagnosis observed with half of patients (42). The presence of skin metastases itself is a poor prognostic indicator and sign of advanced stage of the disease, with median survival ranging from 14-17 months (43). Age, location of tumor, size, histological grade, clinical stage, surgical margins and mitotic count are crucial prognostic factors in defining outcomes of patients with LMS (19). Guillou et al (44) reported that tumors larger than 10 cm, deep-location and high-grade tumors (G2, G3) were predictive of metastasis in multivariate analysis.
In a study from the National Cancer Database, which included more than 7,000 patients with LMS, age was identified as an independent prognostic factor. The survival statistics improved proportionally to the smaller age of the patient at the time of diagnosis. The authors reported a 3% increase in mortality per additional year of age (45). However, the patient group was homogenous, and there was no subgroup analysis of the effect of age on abdominal LMS specifically.
In a retrospective review of 144 patients with abdominal or retroperitoneal LMS from New York, the 5-year disease free survival of patients was 67%, significantly lower than LMS at other anatomical locations (46). There was a recurrence rate of 51%, which also was higher than for LMS located elsewhere. Distant recurrence was the most common recurrence for LMS at all anatomical sites (53%), but local recurrence was more common amongst patients with intraabdominal or retroperitoneal tumors (30%), than at other anatomical locations (46).
The present comprehensive review has summarized the current knowledge and evidence on advance RPLMS with skin metastases. To the best of our knowledge, this is the first review on advanced LMS with skin metastases. Despite the current biological understanding of advanced LMS has improved over years, management of these patients remains challenging. The current review has revealed a lack of high-quality evidence, and a lack of randomized trials in treatment of advanced LMS. Therefore, there is a great need for more substantial and high-quality research in this area.
RPLMS is an uncommon tumor with aggressive nature, therefore it requires special measures to obtain more evidence. Larger studies, especially international multicenter randomized trials with relevant clinical and patient reported outcomes are highly needed. Treatment should be with multidisciplinary approach, combining systemic therapies for disease control with local interventions for symptom relief.
Not applicable.
Funding: No funding was received.
Not applicable.
QB and XW drafted the manuscript. WL and JX conducted literature review. BZ and ZM revised the manuscript. All authors contributed to the article, read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Poosiripinyo T, Sukpanichyingyong S, Salang K, Sumananont C and Chobpenthai T: Recurrence leiomyosarcoma of the popliteal vein: A rare soft tissue sarcoma. Case Rep Surg. 2023(2788584)2023.PubMed/NCBI View Article : Google Scholar | |
|
Stork T, Hegedüs B, Guder W, Hamacher R, Hardes J, Kaths M, Plönes T, Pöttgen C, Schildhaus HU, Streitbürger A, et al: Prognostic factors for leiomyosarcoma with isolated metastases to the lungs: Impact of metastasectomy. Ann Surg Oncol. 29:4429–4436. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Coindre JM, Terrier P, Guillou L, Le Doussal V, Collin F, Ranchère D, Sastre X, Vilain MO, Bonichon F and N'Guyen Bui B: Predictive value of grade for metastasis development in adult soft tissue sarcomas: A Study of 1240 patients from the French Federation of Cancer Centers Sarcoma Group. Cancer. 91:1914–1926. 2001.PubMed/NCBI View Article : Google Scholar | |
|
Baio R, Intilla O, Molisso G, Di Mauro U, Ferrante D, Di Lorenzo G, Buonerba C, Citarella R, Pagano T and Sanseverino R: Primary leiomyosarcoma of the ureter: A case report. Oncol Lett. 29(258)2025.PubMed/NCBI View Article : Google Scholar | |
|
Filho WMNE, Melo SMC, de Brito Alves R, Junior LAS and da Fonseca Reis Silva D: Retromammary fat, axillary and arm metastases from a retroperitoneal leiomyosarcoma: Report of a case with an indolent behaviour. Ecancermedicalscience. 11(778)2017.PubMed/NCBI View Article : Google Scholar | |
|
Kaur S, Tyagi R, Selhi P, Singh A, Puri H and Sood N: Subcutaneous axillary and scalp metastases from non-gynecological retroperitoneal leiomyosarcoma: An unusual presentation after surgical resection. Rare Tumors. 7(5970)2015.PubMed/NCBI View Article : Google Scholar | |
|
De Giorgi V, Sestini S, Massi D, Papi F, Alfaioli B and Lotti T: Superficial cutaneous leiomyosarcoma: A rare, misleading tumor. Am J Clin Dermatol. 9:185–187. 2008.PubMed/NCBI View Article : Google Scholar | |
|
Swallow CJ, Strauss DC, Bonvalot S, Rutkowski P, Desai A, Gladdy RA, Gonzalez R, Gyorki DE, Fairweather M, van Houdt WJ, et al: Management of primary retroperitoneal sarcoma (RPS) in the Adult: An Updated consensus Approach from the Transatlantic Australasian RPS Working Group. Ann Surg Oncol. 28:7873–7888. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Sogaard AS, Laurberg JM, Sorensen M, Sogaard OS, Wara P, Rasmussen P and Laurberg S: Intraabdominal and retroperitoneal soft-tissue sarcomas-outcome of surgical treatment in primary and recurrent tumors. World J Surg Oncol. 8(81)2010.PubMed/NCBI View Article : Google Scholar | |
|
Brennan MF, Antonescu CR, Moraco N and Singer S: Lessons learned from the Study of 10,000 patients with soft tissue sarcoma. Ann Surg. 260:416–421; discussion 421-422. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Casali PG, Abecassis N, Aro HT, Bauer S, Biagini R, Bielack S, Bonvalot S, Boukovinas I, Bovee JVMG, Brodowicz T, et al: Soft tissue and visceral sarcomas: ESMO-EURACAN clinical Practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 29 (Suppl 4):iv51–iv67. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Wang WL, Bones-Valentin RA, Prieto VG, Pollock RE, Lev DC and Lazar AJ: Sarcoma metastases to the skin: A clinicopathologic study of 65 patients. Cancer. 118:2900–2904. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Øines MN, Smith HG, Preisler L and Penninga L: Leiomyosarcoma of the abdomen and retroperitoneum: A systematic review. Front Surg. 11(1375483)2024.PubMed/NCBI View Article : Google Scholar | |
|
Clark MA, Fisher C, Judson I and Thomas JM: Soft-tissue sarcomas in adults. N Engl J Med. 353:701–711. 2005.PubMed/NCBI View Article : Google Scholar | |
|
Antonescu CR, Blay JY and Bovée MG: WHO Classification of Tumours: Soft Tissue and Bone Tumours. IARC Press, Lyon, 2020. | |
|
Weekes RG, McLeod RA, Reiman HM and Pritchard DJ: CT of soft-tissue neoplasms. AJR Am J Roentgenol. 144:355–360. 1985.PubMed/NCBI View Article : Google Scholar | |
|
Choi JH and Ro JY: Retroperitoneal sarcomas: An Update on the Diagnostic pathology update Approach. Diagnostics (Basel). 10(642)2020.PubMed/NCBI View Article : Google Scholar | |
|
Iwata J and Fletcher CD: Immunohistochemical detection of cytokeratin and epithelial membrane antigen in leiomyosarcoma: A systematic study of 100 cases. Pathol Int. 50:7–14. 2000.PubMed/NCBI View Article : Google Scholar | |
|
Mestiri S, Elghali MA, Bourigua R, Abdessayed N, Nasri S, Amine BA, Missaoui N, Ben Maitig M, Hmissa S, Sriha B and Mokni M: Soft tissue leiomyosarcoma-diagnostics management and prognosis: Data of the registry cancer of the center of Tunisia. Rare Tumors. 11(2036361318820171)2019.PubMed/NCBI View Article : Google Scholar | |
|
Hashimoto H, Daimaru Y, Takeshita S, Tsuneyoshi M and Enjoji M: Prognostic significance of histologic parameters of soft tissue sarcomas. Cancer. 70:2816–2822. 1992.PubMed/NCBI View Article : Google Scholar | |
|
Grimaudo MS, Renne SL, Colombo P, Giordano L, Gennaro N, Laffi A, Cariboni U, Cananzi FCM, Ruspi L, Santoro A and Bertuzzi AF: Prognostic value of mitotic count in leiomyosarcoma: A comprehensive monocentric retrospective study. Hum Pathol. 143:17–23. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Rodríguez-Lomba E, Molina-López I, Parra-Blanco V, Suárez-Fernández R and Pulido-Pérez A: Clinical and histopathologic findings of cutaneous leiomyosarcoma: Correlation with prognosis in 12 patients. Actas Dermosifiliogr (Engl Ed). 109:140–147. 2018.PubMed/NCBI View Article : Google Scholar : (In English, Spanish). | |
|
Zagars GK, Mullen JR and Pollack A: Malignant fibrous histiocytoma: Outcome and prognostic factors following conservation surgery and radiotherapy. Int J Radiat Oncol Biol Phys. 34:983–994. 1996.PubMed/NCBI View Article : Google Scholar | |
|
Yoshimoto M, Yamada Y, Ishihara S, Kohashi K, Toda Y, Ito Y, Yamamoto H, Furue M, Nakashima Y and Oda Y: Comparative study of myxofibrosarcoma with undifferentiated pleomorphic sarcoma: Histopathologic and clinicopathologic review. Am J Surg Pathol. 44:87–97. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Sugiura Y, Machinami R, Matsumoto S, Kanda H, Ae K, Takazawa Y and Takeuchi K: Prognostic value of CD34 expression status in patients with in myxofibrosarcomas and undifferentiated pleomorphic sarcomas. Sci Rep. 11(15494)2021.PubMed/NCBI View Article : Google Scholar | |
|
Scurr M: Histology-driven chemotherapy in soft tissue sarcomas. Curr Treat Options Oncol. 12:32–45. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Judson I, Verweij J, Gelderblom H, Hartmann JT, Schöffski P, Blay JY, Kerst JM, Sufliarsky J, Whelan J, Hohenberger P, et al: Doxorubicin alone versus intensified doxorubicin plus ifosfamide for first-line treatment of advanced or metastatic soft-tissue sarcoma: A randomised controlled phase 3 trial. Lancet Oncol. 15:415–423. 2014.PubMed/NCBI View Article : Google Scholar | |
|
D'Ambrosio L, Touati N, Blay JY, Grignani G, Flippot R, Czarnecka AM, Piperno-Neumann S, Martin-Broto J, Sanfilippo R, Katz D, et al: Doxorubicin plus dacarbazine doxorubicin plus ifosfamide or doxorubicin alone as a first-line treatment for advanced leiomyosarcoma: A propensity score matching analysis from the European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group. Cancer. 126:2637–2647. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Garcia-del-Muro X, Lopez-Pousa A, Maurel J, Martín J, Martínez-Trufero J, Casado A, Gómez-España A, Fra J, Cruz J, Poveda A, et al: Randomized phase II study comparing gemcitabine plus dacarbazine versus dacarbazine alone in patients with previously treated soft tissue sarcoma: A Spanish Group for Research on Sarcomas study. J Clin Oncol. 29:2528–2533. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Seddon B, Strauss SJ, Whelan J, Leahy M, Woll PJ, Cowie F, Rothermundt C, Wood Z, Benson C, Ali N, et al: Gemcitabine and docetaxel versus doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): A randomised controlled phase 3 trial. Lancet Oncol. 18:1397–1410. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Pautier P, Italiano A, Piperno-Neumann S, Chevreau CM, Penel N, Firmin N, Boudou Rouquette P, Bertucci F, Le Brun-Ly V, Ray-Coquard IL, et al: A randomised, multicenter phase-III study comparing doxorubicin (dox) alone versus dox with trabectedin (trab) followed by trab in non-progressive patients (pts) as first-line therapy, in pts with metastatic or unresectable leiomyosarcoma (LMS): Final results of the LMS-04 study (Abstract 1913O). Ann Oncol. 34(S1030)2023. | |
|
van der Graaf WT, Blay JY, Chawla SP, Kim DW, Bui-Nguyen B, Casali PG, Schöffski P, Aglietta M, Staddon AP, Beppu Y, et al: Pazopanib for metastatic soft-tissue sarcoma (PALETTE): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 379:1879–1886. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Chi Y, Yao Y and Wang SS: Anlotinib for metastatic STS: Randomized trial. J Clin Oncol. 36 (Suppl 15)(abstr 11503)2018. | |
|
Zhuang RY, Liu WS and Wang ZM: Anlotinib + chemotherapy in advanced LMS. Zhonghua Zhuanyi Zhongliu Zazhi. 4:106–111. 2021.(In Chinese). | |
|
Tap WD, Wagner AJ, Schoffski P, Martin-Broto J, Krarup-Hansen A, Ganjoo KN, Yen CC, Abdul Razak AR, Spira A, Kawai A, et al: Effect of Doxorubicin plus olaratumab vs doxorubicin plus placebo on survival in patients with advanced soft tissue sarcomas: The ANNOUNCE Randomized Clinical trial. JAMA. 323:1266–1276. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Burgess MA, Bolejack V and Schuetze S: Pembrolizumab in UPS and pleomorphic LPS: SARC028 final results. J Clin Oncol. 37 (Suppl 15)(S11015)2019. | |
|
Saerens M, Brusselaers N, Rotteys Decruyenaere A, Creytens D and Lapeire L: Immune checkpoint inhibitors in treatment of soft-tissue sarcoma: A systematic review and meta-analysis. Eur J Cancer. 152:165–182. 2021.PubMed/NCBI View Article : Google Scholar | |
|
D'Angelo SP, Mahoney MR, Van Tine BA, Atkins J, Milhem MM, Jahagirdar BN, Antonescu CR, Horvath E, Tap WD, Schwartz GK and Streicher H: Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): Two open-label, non-comparative, randomised, phase 2 trial. Lancet Oncol. 19:416–426. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Wilky BA, Maleddu A, Mailhot A, Cartwright C, Gao D, Moreno Tellez C, Powers K, Kemp L, Therrien N, Patel JM, et al: A single-arm, open-label phase 2 trial of doxorubicin plus zalifrelimab, a CTLA-4 inhibitor, with balstilimab, a PD-1 inhibitor, in patients with advanced/metastatic soft tissue sarcomas (Abstract 11501). J Clin Oncol. 41(11501)2023.PubMed/NCBI View Article : Google Scholar | |
|
Reichardt P, Andreou D, Flörcken A, Groß T, Richter S, Kessler T, Kortüm M, Schmidt CA, Kasper B, Wardelmann E, et al: Efficacy and safety of nivolumab and trabectedin in pretreated patients with advanced soft tissue sarcomas (STS): Results of a phase II trial of the German Interdisciplinary Sarcoma Group (GISG-15, NitraSarc) (Abstract 11500). J Clin Oncol. 41(11500)2023. | |
|
Martin-Broto J, Diaz Beveridge R, Moura D, Ramos R, Martinez-Trufero J, Carrasco-Garcia I, Lopez-Pousa A, Gonzalez-Billalabeitia E, Gutierrez A, Cruz Jurado J, et al: ImmunoSarc2: A Spanish Sarcoma Group (GEIS) phase Ib trial of doxorubicin and dacarbazine plus nivolumab in first-line treatment of advanced leiomyosarcoma (Abstract 11502). J Clin Oncol. 41(11502)2023. | |
|
Jensen ML, Jensen OM, Michalski W, Nielsen OS and Keller J: Intradermal and subcutaneous leiomyosarcoma: A clinicopathological and immunohistochemical study of 41 cases. J Cutan Pathol. 23:458–463. 1996.PubMed/NCBI View Article : Google Scholar | |
|
Gronchi A, Miah AB, Dei Tos AP, Abecassis N, Bajpai J, Bauer S, Biagini R, Bielack S, Blay JY, Bolle S, et al: Soft tissue and visceral sarcomas: ESMO-EURACAN-GENTURIS clinical practice guidelines for diagnosis, treatment and follow-up☆. Ann Oncol. 32:1348–1365. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Guillou L, Coindre JM, Bonichon F, Nguyen BB, Terrier P, Collin F, Vilain MO, Mandard AM, Le Doussal V, Leroux A, et al: Comparative study of the National Cancer Institute and French Federation of Cancer Centers Sarcoma Group grading systems in a population of 410 adult patients with soft tissue sarcoma. J Clin Oncol. 15:350–362. 1997.PubMed/NCBI View Article : Google Scholar | |
|
Gootee J, Sioda N, Aurit S, Curtin C and Silberstein P: Important Prognostic factors in leiomyosarcoma survival: A National Cancer Database (NCDB) analysis. Clin Transl Oncol. 22:860–869. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Gladdy RA, Qin LX, Moraco N, Agaram NP, Brennan MF and Singer S: Predictors of Survival and recurrence in primary leiomyosarcoma. Ann Surg Oncol. 20:1851–1857. 2013.PubMed/NCBI View Article : Google Scholar |