1. Introduction
Breast cancer is primarily known for its epithelial origins, yet a rare and diverse group of tumors arises from mesenchymal tissues, representing only 0.5% to 1% of all breast tu- mors. These mesenchymal tumors are characterized by a wide array of histological sub- types, including benign and malignant forms. Among these, malignant mesenchymal tu- mors are particularly concerning due to their aggressive behavior, high rates of local re- currence and overall poor prognosis.
Malignant mesenchymal tumors of the breast, which include variants such as phyllodes tumors and primary breast sarcomas, present unique challenges in both diagnosis and treatment. Their rarity often leads to limited clinical experience, resulting in insufficient data to establish robust management protocols. Most available studies are retrospective and small-scale making it difficult to derive standardized diagnostic criteria or treatment guidelines. Consequently, clinicians are often faced with uncertainty in the clinical man- agement of these tumors leading to heterogeneous practices and outcomes.
The distinct biological behaviors of malignant mesenchymal tumors require a tailored approach to treatment that diverges from conventional breast cancer therapies. This review aims to bridge the gap in knowledge by synthesizing current insights into the diagnosis and management of these aggressive tumors. We will focus on the complex nature of malignant mesenchymal tumors, looking at the diagnostic challenges clinicians encounter and assessing current treatment options, especially the important role of surgery. Additionally, we will explore the evolving roles of chemotherapy and radiotherapy, assessing their impact on patient outcomes and identifying unmet challenges that require further investigation.
By improving our understanding of malignant mesenchymal tumors of the breast, this 76review aims to shed light on new insights and emphasize the urgent need for standardized treatment protocols, ultimately enhancing patient care in this difficult area of oncology.
2. Phyllodes Tumors
Phyllodes tumors (PT) of the breast are neoplasms composed of epithelial and stromal elements [
4].
They were first described in 1938 by Johannes Müller as phyllodes cystosarcomas [
5].
The term "phyllodes" refers to the "leaf-like" appearance these lesions exhibit on histopathological examination due to their stromal component, rich in spindle cells mixed with glandular elements lined by epithelial and myoepithelial cells [
6].
They are characterized by rapid dimensional growth and often present as fibroadenomas on initial radiological examinations (ultrasound and mammography); cytological examination via fine needle aspiration and histological examination via core biopsy may sometimes be inadequate to distinguish a PTs from a fibroadenoma reliably [
7].
Excisional biopsy is recommended for diagnostic purposes in cases of rapid growth of fibroadenoma-like lesions, which exhibit variable biological behavior. Based on histopathological characteristics, PTs are categorized as benign (60–75%), borderline (15–20%), or malignant (10–20%) [
8].
Less aggressive forms behave similarly to fibroadenomas, though they have a greater likelihood of local recurrence (LR). Conversely, malignant tumors can metastasize to distant sites and may occasionally transform histologically into sarcomatous lesions.
According to the 2019 WHO histopathological classification, MPTs are characterized by increased stromal cellularity and atypia, with >10 mitoses per 10 high-power fields (10 HPF), exuberant stromal growth, and infiltrative margins[
9].
National Comprehensive Cancer Network (NCCN) guidelines recommend radical excision of the lesion with ≥1 cm negative margins. Axillary lymph node metastases are rare; therefore, sentinel lymph node biopsy and/or axillary lymphadenectomy are not indicated unless the lymph nodes are suspected at clinical staging [
10].
2.1. EPIDEMIOLOGY AND RISK FACTORS
PTs represent approximately 0.3–0.9% of all breast neoplasms. Among all PTs, malignant ones constitute 10-20% of cases, with an annual incidence of 2.1 cases per million women [
11].
Due to the rarity of these neoplasms, epidemiological data are limited. The median age of onset is 45 years (range 10-82 years) [
12].
High-grade malignant forms are more common in patients older than the median age [
13].
In men, PTs are generally associated with gynecomastia [
9].
No etiological or risk factors have been described for the development of these neoplasms except for Li-Fraumeni syndrome, a rare autosomal dominant condition characterized by the development of multiple tumor types [
14].
2.2. DIAGNOSIS
PTs are a complex type of breast lesions that pose challenges for diagnosis
They often mimic fibroadenomas radiologically [
6,
15,
16,
17].
On mammography, they appear as a well-circumscribed, hyperdense or isodense, round- or oval-shaped mass [
6,
15,
16] with rapid growth and large dimensions.
Ultrasound may show hypoechoic, heterogeneous, or complex cystic and solid echo patterns [
16].
Features such as lobulated shape, heterogeneous internal echo pattern and absence of microcalcification are significant sonographic features used to favor PTs over fibroadenomas [
6,
15].
Specific sonographic features, such as liquefaction and a heterogeneous inner echo, may indicate a higher risk of MPT of the breast [
18] .
On Magnetic Resonance Imaging (MRI), PTs appear as oval, well-circumscribed, isointense on T1-weighted images and heterogeneously hyperintense on T2-weighted images [
15] due to the presence of cysts, necrosis, or septations, which may be related to the tumor’s rapid growth .
Some MRI findings can help determine the risk of malignancy. Non-circumscribed margins, cystic components, irregular cyst walls, peritumoral edema, low signal intensity on T2-weighted images, and low apparent diffusion coefficient (ADC) are correlated to higher histologic grade, presence of stromal hypercellularity, hemorrhagic infarction, and necrosis on histopathology [
16].
2.3. PATHOLOGY ASSESSMENT
PTs are rare biphasic fibroepithelial neoplasms of the breast, accounting for 0.3% to 1% of all primary breast tumors. Their biological behavior can vary significantly, ranging from benign to malignant, with a notable tendency for LR and, in malignant cases, the potential for metastasis. Approximately 10% to 15% of PTs are classified as malignant. The World Health Organization categorizes PTs as benign, borderline, or malignant based on factors such as stromal cellularity, stromal overgrowth, nuclear atypia, mitotic activity, tumor margins, and the presence of malignant heterologous elements [
19].
Benign PTs generally have well-defined borders with slight protrusions into surrounding tissue and a bland, uniform stroma that may resemble an intracanalicular fibroadenoma. Key diagnostic features of benign PTs include a cellular stroma with a subepithelial distribution, a low mitotic count of fewer than 5 mitoses per 10 high-power fields, and a varied stroma that can include hyalinized or myxoid areas.
Borderline tumors, according to the WHO classification, have intermediate characteristics, exhibiting microscopic invasion at the tumor margins along with moderate cellularity and pleomorphism. While borderline tumors may recur locally, they rarely metastasize [
2].
MPTs are characterized by significant nuclear pleomorphism, diffuse stromal cellularity, stromal overgrowth (where the epithelial component is absent in at least one low-power field), the heightened mitotic activity of more than 10 mitoses per 10 HPFs, and infiltrative borders. Malignant heterologous elements indicate malignancy, even if other features are absent.
Differentiating MPTs from pure or metastatic sarcomas can be quite challenging. There is a notable risk of distant metastasis—reportedly up to 16%—especially to the lungs and bones, while involvement of the axillary lymph nodes is rare
Histological factors associated with distant metastases include tumor size over 7 cm, infiltrative borders, marked stromal overgrowth, high stromal cellularity, >5 mitoses per 10 HPFs, and necrosis. MPTs are often resistant to chemotherapy, and the prognosis for metastatic cases is poor [
13,
20] .
The immunohistochemical analysis indicates that benign PTs display low positivity for p53, Ki-67, CD117, EGFR, p16, and VEGF, with an increasing positivity in MPTs. Other Authors have defined stroma-rich PT with a bland appearance as typical CD34 positive, while high-grade MPT usually show loss or lesser grade of CD34 staining [
21].
The majority of MPTs however show some degree of CD34 positivity. Genetic studies have shown that benign and borderline PTs exhibit minimal genetic alterations. In contrast, MPTs frequently display specific genetic changes, such as gains on chromosome 1q, losses on chromosome 13q, and deletions on chromosome 9p21, which are associated with the loss of p16INK4A expression. Additionally, mutations in genes such as TP53, RB1, NF1, PIK3CA, and ERBB4 have been found in MPTs, suggesting potential therapeutic targets. Additionally, mutations in genes such as TP53, RB1, NF1, PIK3CA, and ERBB4 have been found in MPTs, suggesting potential therapeutic targets.
In conclusion, accurate histological assessment of PTs, along with immunohistochemical and genetic analyses, helps differentiate between benign, borderline, and MPT. This comprehensive approach guides appropriate clinical intervention and contributes to improved patient outcomes [
7,
8,
21,
22,
23].
2.4. SURGICAL TREATMENT
Surgical excision is the recommended treatment for operable MPTs [
6,
15,
16,
24,
25,
26].
As for other breast tumors, the choice of surgery should take into account both oncological and cosmetic outcomes [
2,
6,
15,
24].
If an adequate margin and a good cosmetic outcome can be obtained with a wide excision, breast-conserving surgery should be the first choice of treatment.
NCCN guidelines recommend at least a 1 cm excision margins if preoperative assessments suggest the presence of a borderline or MPT, while wide margins are not required for excision of benign PTs [
10,
16,
17,
26,
27] .
However, the evidence of a closer margin at final pathology should be discussed in a multidisciplinary context, as according to NCCN guidelines, mastectomy is not an absolute indication in these cases [
6,
10,
28,
29,
30,
31,
32,
33].
Mastectomy is recommended when the preoperative surgical assessment estimates the inability to obtain adequate margins without causing cosmetic deformities that would be unacceptable to the patient [
10,
13,
15,
16,
26,
34,
35] While tumor size is not associated with LR [
7,
36], most studies agree that patients with evidence of narrow surgical margins at final pathology have higher LR rates than those with negative margins [
6,
21,
31,
32,
37,
38,
39,
40].
These include a systematic review and meta-analyses of 9234 individual cases (18% of these patients having MPT where a positive surgical margin was significantly associated with a higher LR risk (OR 6.85; 95% CI 1.58–29.64) [
24,
27].
However, a meta-analysis of 4 studies including 162 patients demonstrated no differences in LR between ≥ 1 cm and < 1 cm margins [
29,
41].
In current NCCN guidelines, there are no clinical scenarios in which axillary staging is recommended [
10] , so there is no indication to perform axillary surgery[
10,
15,
16,
20,
42,
43,
44].
A palpable lymphadenopathy is noted in 10%-15% of patients presenting with MPT but is usually reactive to tumor necrosis and infected or ulcerated skin lesions, with <1% of lymph nodes being pathologically involved [
45].
2.5. ADJUVANT RADIOTHERAPY
There is no strong consensus on whether RT improves overall survival (OS) or disease-free survival (DFS) in patients with MPT, particularly those with low-grade or completely excised tumors.
Current guidelines from the NCCN and the European Society for Medical Oncology (ESMO) suggest that radiotherapy may be considered for patients with MPT that have high-risk features, such as positive surgical margins, tumor size greater than 5 cm, or high-grade pathology [
10,
46].
Conflicting evidence further complicates the utility of adjuvant RT in MPTs. Retrospective studies have suggested that while RT may reduce LR rates, it does not appear to confer a significant survival benefit [
47].
These findings underscore the role of margin status in the decision to administer RT, suggesting that it may primarily serve as a tool for local control rather than improving long-term survival outcomes.
Pooled analyses and large database studies are essential for accurately assessing the role of therapies in rare diseases, where survival outcomes are challenging to evaluate among a heterogeneous population [
48].
The primary large database analyses regarding the role of radiotherapy in malignant tumors are derived from studies utilizing the Surveillance, Epidemiology, and End Results (SEER) database. The first analysis, published in 2006, examined a total of 821 patients and found that only 76 (9%) received adjuvant radiotherapy. The administration of RT has become more common since the year 2000 [
20].
A recent update of SEER analysis published in 2024, reported results of a larger cohort of 2261 patients, who were diagnosed and treated in the last two decades.
In this larger cohort, at least 20% of patients (20,12%) underwent radiotherapy. Results showed that in terms of OS and breast cancer-specific survival, RT did not have a significant impact [
49].
In conclusion, while adjuvant radiotherapy may be beneficial for certain high-risk patients with MPT—particularly those with positive margins or high-grade histology—the evidence supporting its universal application remains limited. Prospective trials and long-term follow-up studies are needed to refine the guidelines and better delineate which subgroups of patients will benefit most from RT after excision. Until then, treatment decisions should be personalized, carefully considering tumor biology, surgical outcomes, and patient preferences.
2.6. SYSTEMIC TREATMENT
The role of chemotherapy for MPT is controversial and should only be considered after consultation with an expert center.
Many investigators have confirmed the low efficacy of monochemotherapy (Cyclophosphamide, Ifosfamide, Doxorubicin). Currently, treatments typically involve doxorubicin and ifosfamide, and are similar to those for soft tissue sarcoma (STS), as this combination showed the most activity and greatest impact on survival [
44].
Neoadjuvant therapy appears to hold a similar significance, yet some recent case reports indicate that aggressive treatment with alkylating agents should be pursued when feasible[
50] .
Approximately 20% of patients with MPT develop metastases. Generally, research on systemic therapy is limited to small case reports and series that often highlight positive outcomes. However, in practice, patients appear to receive less benefit from chemotherapy, experiencing relatively short durations of response compared to those with more common sarcoma histologies [
51].
Given the rarity of the disease, new therapeutic possibilities are yet to be explored in phase II/III trials. Again, a case report shows the potential of a small molecule targeted to VEGFR2 like apatinib. Specifically for this case, the patient had undergone surgery at the primary site and at lung metastases and subsequent chemoradiotherapy with paclitaxel. At disease progression, apatinib was able to reduce the lung mass and increase the patient’s survival by 8 months, until she was lost to follow-up. Obviously, further cohort and prospective trials are needed to identify a subset of patients suitable for apatinib in the clinical treatment of PTs [
52].
3. Primary Breast Sarcoma
Breast sarcomas comprise a heterogeneous group of non-epithelial neoplasms. They are rare tumors, accounting for <1% of breast neoplasms and <5% of soft tissue sarcomas. The annual incidence is approximately 4.5 cases per million[
53].
They originate from the connective tissue of the breast and can be classified as primary and secondary. Clinically, they mostly present as unilateral lesions with ill-defined margins and fixation to underlying planes, often rapidly growing [
3].
The average size at diagnosis is larger compared to epithelial tumors, with a mean diameter of about 3 cm at the time of pathological analysis, although lesions up to 40 cm in diameter have been described [
54].
The overlying skin and the nipple-areola complex are rarely infiltrated, except for cases of angiosarcoma where thickening, erythema, and/or red-violet discoloration of the skin overlying the lesion may occur [
55].
PBS, like sarcomas in other body sites, are poorly responsive to chemo- and radiotherapeutic treatments, although the role of the latter is still debated; currently, the main treatment remains radical surgical excision [
56,
57].
Due to the rarity of these neoplasms, current knowledge on the subject is based on case reports and retrospective studies from limited samples [
58].
3.1. EPIDEMIOLOGY AND RISK FACTORS
Malignant breast sarcomas predominantly affect women, with a peak incidence in the fifth to sixth decades of life, although they can occur at any age [
59].
The most common subtype is angiosarcoma, which accounts for approximately 30-50% of breast sarcomas [
60,
61].
In some patients, genetic factors (e.g., Li-Fraumeni syndrome or Neurofibromatosis type 1) and environmental factors (exposure to radiation, vinyl chloride, alkylating agents, immunosuppressive agents) may be implicated [
54], while chronic lymphedema after mastectomy and axillary lymph node dissection is a reported risk factor for the onset of lymphangiosarcoma (Stewart-Treves syndrome) [
62].
Unlike carcinomas, breast sarcomas are less influenced by hormonal factors like estrogen, although some subtypes may express hormone receptors.
3.2. PATHOLOGIC ASSESMENT
PBS are histologically heterogeneous: an accurate estimate of subtype distribution is hindered not only by the rarity of these neoplasms but also by the different modalities with which they are classified [
63].
Currently, the classification referred to is that established by the WHO in 2020 [
3].
In case series including all subtypes of PBS, angiosarcoma and undifferentiated pleomorphic sarcoma (previously known as malignant fibrous histiocytoma) constitute the main variants[
64].
Subcategories of undifferentiated/not otherwise specified sarcomas include pleomorphic sarcoma, round cell sarcoma, and spindle cell sarcoma[
65].
In a Canadian national case series of 991 patients with PBS - excluding angiosarcoma - the distribution of histological subtypes was as follows: unspecified sarcomas 26%, spindle cell sarcoma 14%, leiomyosarcoma (LMS) 12%, giant cell sarcoma 10%, stromal sarcoma 6%, malignant fibrous histiocytoma 6%, myxoid fibrosarcoma 4%, dermatofibrosarcoma protuberans 3%, fibrosarcoma 3%, undifferentiated sarcoma 3%, liposarcoma (LPS) 3%, pleomorphic liposarcoma 2%, others 8%[
66].
Grading represents an essential prognostic factor in sarcomas, including those of mammary origin: the elements considered for grading are the extent of tissue differentiation, mitotic count, presence/absence of necrosis, cellularity, and pleomorphism; angiosarcoma is an exception for which the literature results are conflicting [
67].
Primary breast angiosarcoma, although relatively rare, is the most common PBS. It may develop spontaneously but is more usually seen in patients who received radiotherapy to the affected breast. Angiosarcomas are rare aggressive malignant tumors derived from endothelial cells and histologically they show poorly formed irregular vascular channels infiltrating the stroma with pleomorphic hyperchromatic endothelial cells exhibiting high mitotic activity. These tumors often have hematoma-like areas due to extensive vascular proliferation and infiltrative growth without well-defined borders. Additional features include the presence of epithelioid cells in some variants and frequent hemorrhage. Angiosarcomas must be differentiated from benign vascular lesions like hemangiomas which show well-formed vascular channels lined by regular endothelial cells and lack significant atypia and mitotic activity. Tumour cells are positive with a wide range of endothelial markers such as CD31, CD34, D2-40, and ERG [
68].
In postradiation angiosarcoma a strong and diffuse nuclear positivity (14), with a loss or reduced expression of H3K27me3 can be observed [
69,
70].
Pseudosarcomatous lesions such as nodular fasciitis and proliferative myositis can mimic sarcomas histologically but are benign reactive processes. Nodular fasciitis presents with loose myxoid stroma plump spindle cells with abundant cytoplasm often in a tissue culture-like growth pattern and common mitotic figures without atypical forms. Proliferative myositis is characterized by ganglion-like giant cells and a more prominent fascicular growth pattern.
Both entities show rapid growth and high cellularity but lack the atypia and infiltrative behavior of true sarcomas. The differential diagnosis includes low-grade sarcomas like low-grade myxofibrosarcoma which shows more irregular cellular shapes and infiltrative growth[
71,
72,
73].
Metastases to the breast may mimic other high-grade spindle cell breast lesions. Tumours that metastasize to the breast, showing spindle cell morphology, include melanoma, LMS and sarcomatoid renal cell carcinoma. Careful morphology evaluation, the bilaterality of the neoplastic involvement and clinical history together withan appropriate and focused immunohistochemical panel helps to clarify the diagnosis.
Finally, breast spindle cell lesions represent a broad spectrum of morphologically and clinically overlapping entities, ranging from benign/reactive processes to aggressive malignant neoplasias with different management strategies.
The diagnosis of mesenchymal lesions in the breast relies on a combination of histopathological examination, immunohistochemical staining and molecular studies where necessary Immunohistochemical markers are critical in differentiating between lesions for instance CD34 is useful in distinguishing myofibroblastomas and spindle cell lipomas while cytokeratins can help identify metaplastic carcinoma in the differential diagnosis of PTs. Molecular studies such as fluorescence in situ hybridization FISH or polymerase chain reaction PCR can be employed in challenging cases to detect specific genetic alterations like MDM2 amplification in well-differentiated LPS.
Common challenges include histological overlap between benign and malignant lesions heterogeneity within the same tumor especially in PTs and LPSs and ensuring sufficient tissue sampling for accurate diagnosis Understanding these histological features is essential for pathologists and clinicians as accurate histological assessment guides clinical management ranging from conservative follow-up for benign lesions to aggressive surgical and adjuvant therapy for malignant mesenchymal tumors Continued research and advances in histopathological techniques are critical for improving diagnostic accuracy and patient outcomes.
3.2. DIAGNOSIS
The radiological diagnosis of breast sarcomas utilizes the same modalities as those used for breast epithelial tumors.
Although studies concerning imaging findings in breast sarcomas are limited due to their low incidence, it seems that they have some characteristic radiological findings [
54,
74].
Breast sarcomas on mammography often appear as a noncalcified hyperdense mass, with indistinct or circumscribed margins borders [
54,
74,
75], findings that may resemble benign fibroadenomas[
26,
76].
The lack of calcifications and the oval shape of these lesions distinguish them from epithelial tumors; however, these same characteristics are also described in PTs [
54].
Furthermore, mammography may appear normal in the presence of a breast sarcomatous lesion[
76].
Ultrasound sonography is not specific for radiological diagnosis of breast sarcomas.
Usually they appear as irregular and hyperechoic lesions with no shadowing[
74,
75,
76], although hypoechoic lesions with indistinctive borders and a posterior acoustic shadow have been described [
54].
Contrast-enhanced MRI with contrast is the imaging modality of choice because it allows the identification of suspicious malignant changes (irregularly bordered oval masses with T2 hyperintensity and heterogeneous rapid contrast enhancement and washout)[
26,
54,
75,
76].
MRI also allows a proper assessment of the local extension of the disease (showing the extent of involvement of the surrounding skin, fascia, and muscular structures), which are crucial for planning surgery and subsequent radiotherapy[
26].
Core biopsy should be performed with needles 16 G or bigger [
54],and immunohistochemical analysis of specific cytokeratins should be performed[
26,
74].
The use of fine-needle aspiration is not recommended because histologic subtype and grade cannot be accurately determined and also for the association with false-negative results[
26,
74].
All patients with a diagnosis of breast sarcoma should be assessed for distant disease.
Computed tomography (CT) of the chest, abdomen and pelvis, together with a bone scan should be performed. Lungs are the most common site of metastases globally, while angiosarcoma has a propensity for secondary spread to the liver and bone[
26].
The role of positron emission tomography (PET) scans remains unclear in the assessment of breast sarcomas[
26,
54].
3.3. SURGICAL TREATMENT
It is strongly reccomended that breast sarcomas be referred to a sarcoma referral centre to increase OS by investigating clinicopathological features and taking a multidisciplinary approach [
26,
74].
There is widespread acceptance that surgical resection should be the first modality of treatment for primitive breast sarcoma[
54,
56,
75].
However, the best surgical treatment is still debated, and should be tailored on the individual patient.
The goal of surgery is local control , and mastectomy has been considered the gold standard for many years[
26,
54,
76].
The choice between mastectomy and breast conservative surgery is based on the possibility to obtain negative margins at final histology[
26,
54,
76,
77]: in fact, tumor size and excision margin are the two factors that mostly impact survival and recurrence[
32,
57,
59,
78,
79,
80].
In a study by Wang et al, the 5-year DFS and OS rates for size of tumors <5 cm, 5–10 cm and >10 cm, were 75.0% and 88.9%, 70.8% and 75.0%, and 37.5% and 37.5%, respectively [
81].
It is strongly recommended to evaluate the tumor/breast size ratio before surgery to properly perform correct planning of the surgical strategy [
56].
While some studies advocate for mastectomy, showing increased OS over conservative surgery, others have demonstrated no significant advantage [
63,
79,
80,
81,
82,
83,
84,
85].
For smaller localized breast sarcomas most authors recommend wide excision with a margin greater than 1 cm [
75,
76,
78].
In the case of angiosarcomas, it is recommended to obtain at least a 3 cm clear margin during surgery since these lesions often have an infiltrative cutaneous disease that extends well beyond the visible tumor [
75,
76,
78].
In this set of patients, mastectomy with delayed reconstruction remains the most prudent surgical approach[
54,
76].
Because the aesthetic outcome of breast conserving surgery when approaching large resections is often poor, some authors suggest oncoplastic reconstruction with parenchymal rearrangement or reconstruction with local flap as a viable option [
76,
86].
Sentinel lymph node biopsy is not routinely performed in patients with breast sarcomas, as nodal dissemination is very rare[
54,
75,
76,
87].
In a study by Fong et al involving 1772 cases of STS, the rate of nodal metastasis was only 2.6%, while in a study based on the SEER and focused on breast sarcomas, 129 out of 333 patients underwent lymphadenectomy and only six patients (4.7%) were found to have nodal metastases[
88,
89].
Radical dissection of axillary lymph nodes is appropriate in the presence of histologically proven isolated nodal disease or where the purpose of surgery is local control and symptom relief[
54,
76].
3.4. ADJUVANT RADIOTHERAPY
Breast sarcomas are rare malignant tumors for which treatment protocols are not well-established.
Due to its rare incidence (<1% of all breast cancer diagnosis), univocal indication for therapies are still under investigation. Adjuvant RT is recommended after a positive margin resection, in tumors larger than 5 cm and in any high-grade sarcoma, because of the high risk of LR[
90].
PBS are sometimes associated with p53 mutation and Li Fraumeni syndrome, therefore testing for these pathogenic variants should occur before administering adjuvant RT.
3.5. SYSTEMIC TREATMENT
3.5.1. First-Line Treatments
Even though surgery represents the best approach to breast sarcomas, a certain percentage of these tumors appear locally advanced or unresectable at the time of diagnosis, either because of chest wall invasion or, less commonly, the presence of synchronous distant metastases.
However, the rarity of such diseases accounts for the lack of data on the use of chemotherapy and RT in both the neoadjuvant/adjuvant setting and the metastatic one, dictating decisions based on multidisciplinary and case-specific discussions[
91].
PBS have the same clinical history and prognostic factors as sarcomas arising at other sites. Therefore, it is legitimate to use similar treatment strategies[
55].
However, as for STS arising at the extremities, the lack of data makes the role of chemotherapy extremely controversial; therefore, decisions must always be taken on a case-by-case basis and in a multidisciplinary setting[
51].
Moreover, the response to chemotherapy is utterly variable. Thus, treatment is selected based on each histotype sensitivity[
75].
Chemotherapy is especially taken into consideration in treating high-grade lesions or masses with a diameter larger than 5 cm[
92].
For the tumors that present distant metastases at the moment of diagnosis, although no specific prospective phase 3 trial exists, chemotherapy is now the gold standard approach[
93].
Adjuvant chemotherapy, even if associated with a significant reduction of LR, doesn’t improve OS in young patients or in any pathology subgroup. Poor quality of initial surgery is the most important prognostic and predictive factor for the utility of adjuvant chemotherapy in STS. Thus, adjuvant chemotherapy for STS remains an investigational procedure and is not a routine standard of care[
94,
95].
At the state of the art, for the pbs the standard pre- and post-operative therapy combines anthracyclines (mostly epirubicin) and ifosfamide[
96].
Given the high heterogeneity of STS, a tailored therapeutic choice seemed to be best approach. However, in a population of patients with localized high-risk STS, histology-tailored neoadjuvant therapy was not associated with a better DFS or OS, suggesting that the association of anthracyclines and ifosfamide should remain the regimen to choose whenever neoadjuvant chemotherapy is used in such patients[
97].
Specifically, doxorubicin is the core of first-line treatment, and only a few recent studies assess whether it should be administered as a single agent or in combination.
Within the randomized phase III European Organisation for Research and Treatment of Cancer (EORTC) 62012 trial, a total of 455 locally advanced or metastatic, grade 2 or 3 STS patients were randomly assigned to receive either single-agent doxorubicin (75 mg/m2) or doxorubicin (75 mg/m2) with ifosfamide (10 g/m2 over 4 days) with growth factor support. Patients were treated every 3 weeks for a maximum of 6 cycles or until progression. At a median follow-up of 56 months, the OS difference did not achieve statistical significance (14.3 months with the combination and 12.8 months with doxorubicin alone. HR =0.83; P=0.076). Median progression free survival (PFS), however, was 7.4 months with the combination and 4.6 months with doxorubicin alone, for a 26% reduction in risk that was statistically significant (HR =0.74; P=0.003). The objective response rate (ORR) was 26.5% with the combination and 13.6% with doxorubicin. Despite colony-stimulating factor support, the most common grade 3 and 4 adverse events were all more common in the combination than in the doxorubicin alone group: leucopenia (43% vs. 18%), neutropenia (42% vs. 37%), febrile neutropenia (46% vs. 13%), anemia (35% vs. 5%), and thrombocytopenia (33% vs. <1%). The lack of OS advantage but with more toxicities for the combination regimen do not support its routine use in the setting of advanced incurable disease unless there is an immediate need to decrease tumor bulk, improve symptoms or translate into resection[
98].
Combinations are more frequently used in specific sarcoma subgroups. A retrospective analysis showed that the association of doxorubicin and dacarbazine has a favorable activity in both ORR and PFS in patients with advanced LMS [
99].
Furthermore, the LMS-04 randomised phase III trial - which enrolled 150 patients with LMS in a superiority-aimed study for a first-line association of doxorubicin and trabectedin versus doxorubicin alone - showed a significantly longer median PFS in the investigational arm versus the doxorubicin alone group (12,2 months vs 6,2 months). Nevertheless, the drug combination was associated with a higher, yet manageable, grade of toxicity. The most common grade 3-4 adverse events were neutropenia (13% in the doxorubicin alone group vs 80% in the doxorubicin plus trabectedin group), anaemia (5% vs 31%), thrombocytopenia (0% vs 47%), and febrile neutropenia (9% vs 28%). 12% patients in the doxorubicin alone group and 20% patients in the doxorubicin plus trabectedin group has serious adverse events. Considering the significant increase in PFS, doxorubicin plus trabectedin can still be considered as a first-line treatment of metastatic LMS [
100].
3.5.2. New cytotoxic drugs and strategy
Due to doxorubicin’s significant toxicities, several types of anthracyclines have been recently tested in first-line treatment for STS. Aldoxorubicin is a novel albumin-binding prodrug of doxorubicin. It contains a carboxylic hydrazone and covalently binds to albumin in the blood until reaching tumour tissue, where the acidic microenvironment breaks the covalent bond with albumin and releases doxorubicin. This allows for greater doses (3.5–4 times the standard doxorubicin dose) of doxorubicin to be administered while reducing its side effects, especially concerning cardiac function[
101].
A phase IIb clinical trial randomised 140 patients (ratio 2:1) to receive single agent aldoxorubicin (350 mg/m2; dose equivalent to doxorubicin 260 mg/m2) or doxorubicin (75 mg/m2), administered once every 3 weeks for up to 6 cycles. Aldoxorubicin showed superior efficacy in terms of the primary endpoint, as PFS was significantly increased (5.6 vs 2.7 months; P = 0.02) as was the rate of 6-month PFS (46% and 23%; P = 0.02). Overall tumor response rate was also higher in the interventional arm compared to the control group (25% [20 patients, all partial response] vs 0%) and there were no unexpected events, as adverse events were more frequent with aldoxorubicin but mostly manageable. Specifically, grade 3 or 4 neutropenia was more frequent in the interventional arm (29% vs 12%), but not grade 3 or 4 febrile neutropenia (14% vs 18%). Most importantly, no acute cardiotoxicity was observed in either arm, although left ventricular ejection fraction lower than 50% occurred in 3 of 40 patients receiving doxorubicin[
102].
As sarcomas can be therapy-related or recurrent, and considering the impact of a doxorubicin-based treatment on cardiac function, aldoxorubicin is currently a subject of interest in relapsing or progressing tumours. In a pivotal international, multicentre, Phase III trial evaluating PFS as the primary endpoint, 433 patients with relapsed or refractory STS were randomly assigned to receive aldoxorubicin 350 mg/m2 (260 mg/m2 doxorubicin equivalent) every 3 weeks or investigator’s choice of treatment including dacarbazine, doxorubicin, pazopanib, ifosfamide, and gemcitabine/docetaxel with GCSF support. Tumour subtypes included LMS (42.2%), LPS (12.8%), synovial sarcoma (9.6%), and other sarcomas (34.9%).
Approximately two-thirds of the patients received prior doxorubicin in both experimental and control arms. The mean cumulative doxorubicin equivalent dose of aldoxorubicin administered was 1,359.8 mg/m2 (range 260–10,920). Aldoxorubicin demonstrated superior median PFS over investigator’s choice in patients with LMS or LPS, though there was no difference in the overall cohort. Secondary endpoints included ORR, disease control rate (DCR), OS, and safety.
Although the ORR was twice as high in the doxorubicin arm (8.3 vs. 4.2%, P=0.1106), there was no significant difference in median OS (12.88 months vs. 12.16 months, P=0.8555). There was a trend toward significantly improved ORR in the L-sarcomas group, which also achieved statistically significant DCR (41.7 vs 27.0%, P=0.0161). Globally, there were more high-grade adverse events in the doxorubicin group; however, the incidence of cardiotoxicity – defined by a decrease of 20% or more in left ventricular ejection fraction (LVEF) – was lower (3.8 vs. 8.5%).
Thus, aldoxorubicin represents a viable alternative compared to the standard of care in patients with relapsed or refractory metastatic STS[
103].
Within the same molecule class, amrubicin - a third-generation synthetic 9-amino-anthracycline that inhibits topoisomerase II - has been suggested to be less noxious than doxorubicin, particularly regarding cardiac toxicity. Despite phase III trials yet to be conducted on the subject, a phase II multicenter single-arm study was executed to evaluate the efficacy and tolerability of amrubicin in advanced STS. A total of 24 patients received amrubicin (40 mg/m2 for 3 days in 21-day cycles) as a first-line therapy. The best ORR was 13%, the median PFS was 5.8 months, and the median OS was 26 months. Notably, grade 3 to 4 AEs included febrile neutropenia and anemia in 21% of treated patients, but there was no significant cardiac toxicity up to a cumulative dose of 4,800 mg/m2. One patient with metastatic myxoid-liposarcoma (MLPS) with the TLS-CHOP translocation had a durable response, suggesting that further investigation is warranted in this subtype[
104].
On the other hand, newer fosfamides, such as evefosfamide and palifosfamide, have yet to show potential. Both the SARC021 and the PICASSO phase III trials compare the combination of either doxorubicin and evofosfamide or palifosfamide respectively to doxorubicin alone. The combination arm registered higher RR but more toxicity in both cases[
105,
106].
Another frequently used combination to treat metastatic STS is gemcitabine plus docetaxel, considering the superior PFS and OS compared to gemcitabine alone[
107].
The GeDDiS trial was a phase III, randomized, multicenter study that compared the combination of gemcitabine and docetaxel with doxorubicin in patients with previously untreated advanced unresectable or metastatic STS. A total of 257 patients were enrolled to investigate the primary endpoint of 24-week PFS, which was identical between the investigational arm (675 mg/m2 of gemcitabine on days 1 and 8 plus 75 mg/m2 of docetaxel on day 8 every 21 days) and the control arm (75 mg/m2 doxorubicin). However, the investigational arm suffered higher toxicity (83.3% vs. 94.6% dose intensity reductions, 55.5% vs. 45.7% dose delays, and 10.2% vs. 0.8% withdrawals due to unacceptable toxicity in the investigational arm and in the control arm, respectively). Furthermore, no difference in efficacy was highlighted in specific histology subgroups, such as LMS [
108].
Paclitaxel is an interesting molecule, as it has been proven to have sustained activity in angiosarcomas, which represents a rare yet extremely important histotype among breast sarcomas. Most breast angiosarcomas are secondary to previous RT executed on the same area. In the phase II ANGIOTAX study conducted by a French group, 30 patients were administered paclitaxel intravenously as a 60-minute infusion at a dose of 80 mg/m2 on days 1, 8, and 15 of a 4-week cycle. The primary endpoint was the non-progression rate after two cycles. PFS after 2 and 4 months were 74% and 45%, respectively; the median time to progression was 4 months, and the median OS was 8 months. Additionally, naïve patients and pretreated patients has a comparable PFS rate (77% vs 71%). Toxicity was manageable. In conclusion, even considering the limitations of this study, weekly paclitaxel seems to have the potential to be further investigated in future trials[
109].
3.5.3. Second-line treatments
Second-line treatments involve molecules with moderate activities compared to doxorubicin and ifosfamide, one of the oldest ever discovered being dacarbazine. In 1987, Borden et al. conducted a randomized trial comparing single-agent doxorubicin and doxorubicin plus dacarbazine, which showed that dacarbazine improved the overall response frequency of doxorubicin from 16%-18% (depending on the schedule of administration) to 30%. This was particularly highlighted in LMSs. However, the toxicity experimented in the combination group was far greater, thus making the results controversial[
110].
Dacarbazine’s role as a single-agent therapy in a second-line setting is seemingly as old, as in 1991, Buesa et al. demonstrated that a RR of 18% could be achieved in previously treated patients with advanced STS in a phase II study involving 50 patients (44 fully analyzed). 42 of these patients had been exposed to doxorubicin and ifosfamide, and the most represented histologic subtype was LMS (12 patients). However, the duration of response was concise at only 8 weeks. Nonetheless, these data suggest the possible employment of dacarbazine in combination regimens, for instance, in association with gemcitabine[
111].
In fact, this combination has exhibited efficacy in terms of PFS and OS, albeit without robust confirmation from larger clinical trials[
112].
A real-world data analysis conducted as a retrospective study in a single institution involving 95 patients pointed to a benefit in PFS of 3.5 months and an OS of 14.2 months. Patients with translocated histotypes had better PFS, while those with platelet–lymphocyte ratios (PLRs) surpassing a specific threshold or lower albumin levels had poorer OS. The most represented histotype was again LMS (27.4%) [
113].
Among second-line treatments, molecules such as eribulin and trabectedin are mostly targeted at specific subtypes. Eribulin—a microtubule growth inhibitor—is currently approved for use in pretreated patients with recurrent sarcomas. The median OS for eribulin and dacarbazine was 13.5 and 11.5 months, respectively (P=0.017). The OS was significantly improved (15.6 vs. 8.4 months) in LPS cohorts, while no sense was seen in LMS.
Trabectedin is another drug with proven activity for STS that has been investigated as a second-line choice[
114].
A phase 3, randomized, multi-centre study compared trabectedin (administered to 345 patients) and dacarbazine (173 patients) in locally advanced, unresectable, or metastatic LMS and LPS. All patients had to have received prior therapy with an anthracycline and at least one additional systemic regimen. In the trabectedin arm, trabectedin (1.5 mg/m2) was administered on day 1 of every 3 weeks, whereas in the dacarbazine arm, dacarbazine (1 g/m2) was administered by intravenous infusion over 20–120 min on day 1 of every 21-day cycle. OS was the primary endpoint, yet the interim analysis indicated no improvement, and PFS was a secondary endpoint. Fortunately, treatment with trabectedin resulted in a statistically significant improvement in PFS, with a PFS of 4.2 months and 1.5 months for trabectedin and dacarbazine, respectively[
115].
Another phase III randomized, multicenter, open-label study compared trabectedin to best supporting care (BSC) for heavily pretreated patients with STS. It included 103 adult patients, 60.2% of them with L-STS (LPS/ LMS), who were randomised (1:1) to receive trabectedin 1.5 mg/m2 every 3 weeks or BSC. The primary efficacy endpoint was PFS, which was 3.1 months in the trabectedin arm versus 1.5 months in the BSC arm, with benefits observed across almost all analyzed subgroups, but particularly in patients with L-STS (5.1 versus 1.4 months, P 1⁄4 0.0001). Seven patients (13.7%) in the trabectedin arm (all with L-STS) achieved a partial response, while no objective responses were observed in the BSC arm. Toxicity was mostly haematological or hepatic and the analysis of the impact of the treatment on the quality of life showed no impairment[
116].
Temozolomide, being the prodrug of dacarbazine, is an alkylating agent which interferes with the biosynthesis of purines. This drug showed moderate activity in patients with pretreated sarcomas - and especially LMSs - when administered following a prolonged schedule. However, phase II trials were not followed by phase III studies of temozolomide alone [
117,
118].
3.5.4. New treatment in the Target therapy Era
Among targeted treatments, pazopanib is a potent and selective tyrosine kinase inhibitor (TKI) that blocks tumor growth and inhibits angiogenesis. The PALETTE trial included most histological types, excluding LPS, and is the first placebo-controlled phase III trial investigating the activity of a TKI on STS. 369 patients with metastatic or progressive disease (according to RACIST 1.0) during the 6 months (or 12 months after adjuvant therapy) before the screening were enrolled. Every patient had been previously treated with anthracyclines and subjected to a maximum of 4 therapy lines. The median PFS was 4.6 months for pazopanib and 1.6 months for placebo. The delta in OS was not significant as it was calculated that showing a projected difference of 3 months would require more than 750 patients[
119].
LPS was excluded based on the results of a phase II EORTC 62043 study analysing PFR at 12-weeks, as the LPS cohort was the only one not to reach the progression-free at 12 weeks[
120].
Tivozanib – a TKI targeting VEGFR1-3, with activity against PDGFRa/b and cKIT – showed antitumor activity with a promising median PFS and PFS rate at 4 months in a heavily pretreated population of metastatic STS. A Simon two-stage phase II trial was performed using tivozanib given orally at 1.5 mg daily, 3 weeks on 1 week off on a 28-day cycle until disease progression or intolerable toxicity. 58 patients were enrolled and treated with tivozanib. LMS was the most represented STS subgroup (47%) and 27 patients (46%) had received at least 3 lines of therapy prior to study entry. Up to 24 patients (41%) had prior VEGF targeted therapies. Partial response and stable disease were observed in 2 (3.6%) and 30 (54.5%) patients respectively. The 16-week PFS rate was 36.4% and median PFS was 3.5 months. Median OS was 12.2 months. AEs were manageable. Correlative studies demonstrate no correlation between the expression of VEGFR 1, 2 or 3, PDGFRa/b or FGF, and activity of tivozanib[
121].
Anlotinib is another TKI targeting VEGFR, FGFR, PDGFR, C-kit, which has shown sensible efficacy in treating STS previously exposed to anthracyclines-based therapies. In a randomised double- blind, placebo-controlled phase IIb trial involving 233 patients and investigating PFS as a primary endpoint, median PFS was 6.27 months in the anlotinib arm versus 1.47 months in the control arm, the difference was very significant, and the risk of disease progression was reduced by 67%[
122].
This specific agent has been specifically investigated on long-term administration and as a possible maintenance treatment after first-line chemotherapy.
A retrospective analysis was conducted of patients with advanced sarcomas with measurable target, evaluated using RECIST 1.1, with target lesions measures and PFS as the primary endpoint. 22 patients, 14 of which had previously undergone chemotherapy, were assessed as they had taken anlotinib regularly for > 12 months. The primary diseases included alveolar soft part sarcoma, synovial sarcoma, LMS, and others. 9 patients took anlotinib alone, 13 patients underwent combination treatment, 9 of which with chemotherapy and 4 with immunotherapy (anti-PD-1). Complete remission (CR) was achieved in four (18.18%) cases, partial response (PR) in five (22.73%) cases, and stable disease in 13 (59.09%) cases. The mean PFS for the CR, PR, and stable disease groups was 16.50, 14.50, and 29.31 months, respectively (p < 0.05)[
123].
Another study addressed the potential of anlotinib as a maintenance treatment after chemotherapy in STS. It involved 49 patients who achieved partial response or stable disease after first-line anthracycline-based chemotherapy and who all received anlotinib as a maintenance treatment. The primary endpoint of PFS was 9.1 months, while the median OS was not reached, and the 1-year OS rate was 98.0%. The best ORR and DCR were 16% and 94%, respectively. Thus, anlotinib exhibits promising efficacy and tolerable toxicity in patients with advanced STS[
124].
Anlotinib is also being taken into consideration as a first-line treatment for patients not suitable for chemotherapy. A multicenter, open-label, single-arm, phase II clinical trial done in China involved 39 patients who received 12mg anlotinib once daily for 14 days every 3 weeks until disease progression or unacceptable toxicity, with PFS as a primary endpoint. Median follow-up was 13.4 months. At the data cutoff date, the median PFS and median OS were 7.1 months and 24.3 months, respectively. The PFS at 6 months was 60.0%, and the OS at 12 months was 76.6%. However, of the 37 patients eligible for the evaluation of tumor response, only one achieved confirmed partial response; instead, 30 had stable disease and the DCR was 83.8%. The safety profile was manageable as grade 3 events represented 33.3%[
125].
Another interesting small molecule is regorafenib. In a placebo-controlled, randomized phase 2 trial, regorafenib improved PFS for patients with doxorubicin-pretreated advanced nonadipocytic sarcoma. 182 patients were enrolled. In nonadipocytic sarcoma, the PFS was 4.0 months with regorafenib vs 1.0 month with placebo, while the OS was 13.4 months vs 9.0 months. A quality-adjusted time without symptoms of progression or toxicity (Q-TWiST) post hoc exploratory analysis was applied to provide an integrated measure of its clinical benefit and it resulted in a significant improvement[
126].
The ANNOUNCE 2 trial aimed to explore the addition of olaratumab (O) to gemcitabine (G) and docetaxel (D) for advanced STS. In Phase 2, patients were randomized 1:1 from two cohorts (O-naïve and O-pretreated) to 21-day cycles of olaratumab (20 mg/kg Cycle 1 and 15 mg/kg other cycles, Days 1 and 8), gemcitabine (900 mg/m2, Days 1 and 8), and docetaxel (75 mg/m2, Day 8). Since the primary endpoint was not reached, no result can be considered statistically significant, yet the combination of O+G+D showed a possible favorable trend in OS in the O-pretreated cohort and other efficacy outcomes in both cohorts[
127].
At present, at least 20 clinical trials of PD-1/L1 inhibitor plus chemotherapy in the treatment of sarcomas have been registered and are recruiting. The PD-1 inhibitors used in these clinical trials include camrelizumab, durvalumab, nivolumab, pembrolizumab, retifanlimab, sintilimab, and Toripalimab[
128].
Pembrolizumab has meaningful clinical activity in patients with undifferentiated pleomorphic sarcoma or dedifferentiated LPS. One of the first immune checkpoint inhibitors (ICI) trials with positive results was the prospective single-arm phase II trial SARC028, evaluating the anti-PD1 pembrolizumab as a second-line treatment in 80 patients with either STS or bone sarcoma. 80 of 86 patients’ responses were analysed in a median follow-up of 17.8 months. ORR was 18% with a median PFS and OS of 18 and 49 weeks, respectively. Among the subtypes represented in the cohorts, LMS and Ewing’s sarcoma showed no objective response. 9 (11%) patients (5 in the bone sarcoma group and 4 in the STS group) had treatment-emergent serious adverse events, 5 of whom had immune-related serious adverse events, including adrenal insufficiency, pneumonitis and nephritis[
129].
The response of undifferentiated pleomorphic sarcoma (UPS) to pembrolizumab was further confirmed in an expansion cohort of SARC028 with two CRs and 7 PRs in the UPS cohort; however, a response was not seen in the LPS cohort[
130].
A pooled analysis of several clinical trials investigating anti-PD-1/PD-L1 immunotherapy in advanced STS reported alveolar soft part sarcoma (ASPS) and UPS were among the highest responders (48.4% and 15.7%, respectively) and LPS and LMS were among the lowest (7.3% and 6.9%, respectively)[
131].
Furthermore, combination checkpoint inhibition with nivolumab and ipilimumab was evaluated in previously treated patients with advanced STS in the phase II Alliance A091401 trial and compared to single-agent treatment with nivolumab alone. ORR was 16%, median PFS was 4.1 months and OS was 14.3 months. Better responses were seen with combination therapy, with the best responses being in UPS (33%), LMS (14.2), and AS (33%) [
132].
4. Conclusions
Malignant mesenchymal tumors of the breast are rare but extremely aggressive, present- ing significant challenges in both diagnosis and treatment. Due to the lack of standardized guidelines, clinical decision-making remains complex and varies widely. Surgical treat- ment remains the cornerstone of therapy with the primary goal of achieving complete excision to prevent local recurrences and metastases. Surgical options include both breast- conserving surgery and mastectomy, with axillary treatment typically only necessary in cases of biopsy-proven nodal involvement. While the roles of radiotherapy and chemotherapy are still unclear, ongoing research is essential to refine these treatment modalities. The rarity of these tumors complicates the conduction of large-scale studies making it vital for healthcare professionals to adopt a tailored, multidisciplinary approach, especially in high-volume centers specializing in rare malignancies. These settings provide the best opportunity for improving patient outcomes through expertise and collaboration. Ultimately, advancing our understanding of malignant mesenchymal tumors enhancing diagnostic precision and developing comprehensive treatment protocols will be key to improving patient prognoses and managing these challenging malignancies more effec-tively.
Author Contributions
Conceptualization, F.D.L. and A.M.S.; methodology, F.D.L., F.M. and A.M.; software, A.B.; validation, A.M.S., R.M. and G.F.; formal analysis, L.S., B.C and S.D. ; investigation, A.M.S., A.D.L, M.P.F., F.G. and E.J.M.; resources, A.O., F.S. and I.P.; data curation, A.M.S., A.S. and N.D.; writing—original draft preparation, F.D.L, A.M.S., N.B., A.O., A.M., A.F., E.S. and C.V.P.; writing—review and editing, A.M.S. and L.S.; visualization, F.D.L, C.A.,M.P.F and F.M.; supervision, F.D.L, A.M.S., R.M., and G.F.; project administration, F.D.L and A.M.S.; funding acquisition, L.S. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Conflicts of Interest
The authors declare no conflicts of interest.
Abbreviations
The following abbreviations are used in this manuscript:
PTs WHO MPT PBS RT HPF NCCN MRI ADC LR OS DFS ESMO SEER BCSS STS CT PET PFS ORR EORTC LPS LMS LVEF DCR MLPS PLRs BSC TKI CR PR ICI UPS ASPS O G D |
Phyllodes Tumors World Health Organization Malignant Phyllodes Tumors Primary Breast Sarcomas Radiation Therapy High-power fields National Comprehensive Cancer Network Magnetic Resonance Imaging Apparent Diffusion Coefficient Local Recurrence Overall Survival Disease-free survival European Society for Medical Oncology Surveillance, Epidemiology, and End Results Breast Cancer-Specific Survival Soft Tissue Sarcoma Computed tomography Positron Emission Tomography Progression Free Survival Objective Response Rate European Organisation for Research and Treatment of Cancer Liposarcoma Leiomyosarcoma Left Ventricular Ejection Fraction Disease Control Rate Metastatic Myxoid-Liposarcoma Platelet–lymphocyte Ratios Best Supporting Care Tyrosine Kinase Inhibitor Complete Remission Partial Response Immune Checkpoint Inhibitors Undifferentiated Pleomorphic Sarcoma Alveolar Soft Part Sarcoma Olaratumab Gemcitabine Docetaxel |
References
- Tevatia MS, Mishra P, Baranwal AK, Nichat PB, Shelly D, Awasthi S, Sengupta P. Primary Breast Tumors with Mesenchymal Morphology. J Lab Physicians. 2021 Dec;13(4):362–7. [CrossRef]
- Lakhani S, Ellis I, Schnitt S, Tan P, Van de Vijver M. WHO classification of tumors of the breast, 4th ed. France: Lyon: International Agency for Research on Cancer, 2012.
- LINEE GUIDA SARCOMI DEI TESSUTI MOLLI E GIST [Internet]. AIOM. 2024 [cited 2025 Jan 11]. Available from: https://www.aiom.it/linee-guida-aiom-2024-sarcomi-dei-tessuti-molli-e-gist/.
- Telli ML, Horst KC, Guardino AE, Dirbas FM, Carlson RW. Phyllodes tumors of the breast: natural history, diagnosis, and treatment. J Natl Compr Cancer Netw JNCCN. 2007 Mar;5(3):324–30. [CrossRef]
- Müller, Johannes - Über den feinern Bau und die Formen der krankhaften Geschwülste : in zwei Lieferungen; Bd. 1: Lfg. 1: Bogen 1 - 15 und Tafel 1 - 14 [Internet]. [cited 2025 Jan 14]. Available from: https://echo.mpiwg-berlin.mpg.de/ECHOdocuView?url=/permanent/library/84U9MMK0/pageimg&pn=1&mode=imagepath.
- Lissidini G, Mulè A, Santoro A, Papa G, Nicosia L, Cassano E, Ashoor AA, Veronesi P, Pantanowitz L, Hornick JL, Rossi ED. Malignant phyllodes tumor of the breast: a systematic review. Pathologica. 2022 Apr;114(2):111–20. [CrossRef]
- Salvadori B, Cusumano F, Del Bo R, Delledonne V, Grassi M, Rovini D, Saccozzi R, Andreola S, Clemente C. Surgical treatment of phyllodes tumors of the breast. Cancer. 1989 Jun 15;63(12):2532–6.
- Tse GMK, Niu Y, Shi HJ. Phyllodes tumor of the breast: an update. Breast Cancer Tokyo Jpn. 2010;17(1):29–34. [CrossRef]
- Hanby AM, walker C. Tavassoli FA, Devilee P: Pathology and Genetics: Tumours of the Breast and Female Genital Organs. WHO Classification of Tumours series - volume IV. Lyon, France: IARC Press. Breast Cancer Res. 2004 Mar 31;6(3):133.
- Gradishar WJ, Moran MS, Abraham J, Abramson V, Aft R, Agnese D, Allison KH, Anderson B, Bailey J, Burstein HJ, Chen N, Chew H, Dang C, Elias AD, Giordano SH, Goetz MP, Jankowitz RC, Javid SH, Krishnamurthy J, Leitch AM, Lyons J, McCloskey S, McShane M, Mortimer J, Patel SA, Rosenberger LH, Rugo HS, Santa-Maria C, Schneider BP, Smith ML, Soliman H, Stringer-Reasor EM, Telli ML, Wei M, Wisinski KB, Yeung KT, Young JS, Schonfeld R, Kumar R. Breast Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw JNCCN. 2024 Jul;22(5):331–57.
- Bernstein L, Deapen D, Ross RK. The descriptive epidemiology of malignant cystosarcoma phyllodes tumors of the breast. Cancer. 1993 May 15;71(10):3020–4.
- Barrio AV, Clark BD, Goldberg JI, Hoque LW, Bernik SF, Flynn LW, Susnik B, Giri D, Polo K, Patil S, Van Zee KJ. Clinicopathologic features and long-term outcomes of 293 phyllodes tumors of the breast. Ann Surg Oncol. 2007 Oct;14(10):2961–70. [CrossRef]
- Karim RZ, Gerega SK, Yang YH, Spillane A, Carmalt H, Scolyer RA, Lee CS. Phyllodes tumours of the breast: a clinicopathological analysis of 65 cases from a single institution. Breast Edinb Scotl. 2009 Jun;18(3):165–70. [CrossRef]
- Birch JM, Alston RD, McNally RJ, Evans DG, Kelsey AM, Harris M, Eden OB, Varley JM. Relative frequency and morphology of cancers in carriers of germline TP53 mutations. Oncogene. 2001 Aug 2;20(34):4621–8. [CrossRef]
- Papas Y, Asmar AE, Ghandour F, Hajj I. Malignant phyllodes tumors of the breast: A comprehensive literature review. Breast J. 2020;26(2):240–4. [CrossRef]
- Fede ÂB de S, Pereira Souza R, Doi M, De Brot M, Aparecida Bueno de Toledo Osorio C, Rocha Melo Gondim G, Casali-da-Rocha JC, Jbili R, Bitencourt AGV, Alves de Souza J, Caparica Bitton R, Baroni Alves Makdissi F, Moraes Sanches S. Malignant Phyllodes Tumor of the Breast: A Practice Review. Clin Pract. 2021 Apr 6;11(2):205–15.
- Sars C, Sackey H, Frisell J, Dickman PW, Karlsson F, Kindts I, Marta GN, Freitas-Junior R, Tvedskov TF, Kassem L, Ali AS, Ihalainen H, Neron M, Kontos M, Kaidar-Person O, Meattini I, Francken AB, van Duijnhoven F, Moberg IO, Marinko T, Kollar A, Ahmed M, Remoundos D, Banks J, Jagsi R, Dossett LA, Lindqvist EK. Current clinical practice in the management of phyllodes tumors of the breast: an international cross-sectional study among surgeons and oncologists. Breast Cancer Res Treat. 2023 Jun;199(2):293–304. [CrossRef]
- Logistic regression analysis of ultrasound findings in predicting the malignant and benign phyllodes tumor of breast | PLOS ONE [Internet]. [cited 2025 Jan 11]. Available from: https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0265952.
- Tan PH, Ellis I, Allison K, Brogi E, Fox SB, Lakhani S, Lazar AJ, Morris EA, Sahin A, Salgado R, Sapino A, Sasano H, Schnitt S, Sotiriou C, van Diest P, White VA, Lokuhetty D, Cree IA, WHO Classification of Tumours Editorial Board. The 2019 World Health Organization classification of tumours of the breast. Histopathology. 2020 Aug;77(2):181–5.
- Macdonald OK, Lee CM, Tward JD, Chappel CD, Gaffney DK. Malignant phyllodes tumor of the female breast: association of primary therapy with cause-specific survival from the Surveillance, Epidemiology, and End Results (SEER) program. Cancer. 2006 Nov 1;107(9):2127–33.
- Lenhard MS, Kahlert S, Himsl I, Ditsch N, Untch M, Bauerfeind I. Phyllodes tumour of the breast: clinical follow-up of 33 cases of this rare disease. Eur J Obstet Gynecol Reprod Biol. 2008 Jun;138(2):217–21. [CrossRef]
- Moffat CJ, Pinder SE, Dixon AR, Elston CW, Blamey RW, Ellis IO. Phyllodes tumours of the breast: a clinicopathological review of thirty-two cases. Histopathology. 1995 Sep;27(3):205–18. [CrossRef]
- Barth RJ, Wells WA, Mitchell SE, Cole BF. A prospective, multi-institutional study of adjuvant radiotherapy after resection of malignant phyllodes tumors. Ann Surg Oncol. 2009 Aug;16(8):2288–94. [CrossRef]
- Spanheimer PM, Murray MP, Zabor EC, Stempel M, Morrow M, Van Zee KJ, Barrio AV. Long-Term Outcomes After Surgical Treatment of Malignant/Borderline Phyllodes Tumors of the Breast. Ann Surg Oncol. 2019 Jul;26(7):2136–43. [CrossRef]
- Bishr MK, Banks J, Abdelaziz MS, Badawi M, Crane PW, Donigiewicz UJ, Elkorety M, Girgis M, Humphreys A, Isherwood J, Kahan J, Keelan S, Lindqvist EK, Nixon I, Sackey H, Sars C, Soliman H, Touqan N, Remoundos DD, Ahmed M. Multidisciplinary Management of Phyllodes Tumours and Breast Sarcoma: A Cross-sectional Survey of Clinical Practice across the UK and Ireland. Clin Oncol R Coll Radiol G B. 2024 Jan;36(1):e31–9. [CrossRef]
- Esperança-Martins M, Melo-Alvim C, Dâmaso S, Lopes-Brás R, Peniche T, Nogueira-Costa G, Abreu C, Luna Pais H, de Sousa RT, Torres S, Gallego-Paez LM, Martins M, Ribeiro L, Costa L. Breast Sarcomas, Phyllodes Tumors, and Desmoid Tumors: Turning the Magnifying Glass on Rare and Aggressive Entities. Cancers. 2023 Aug 2;15(15):3933. [CrossRef]
- Lu Y, Chen Y, Zhu L, Cartwright P, Song E, Jacobs L, Chen K. Local Recurrence of Benign, Borderline, and Malignant Phyllodes Tumors of the Breast: A Systematic Review and Meta-analysis. Ann Surg Oncol. 2019 May;26(5):1263–75. [CrossRef]
- Liang MI, Ramaswamy B, Patterson CC, McKelvey MT, Gordillo G, Nuovo GJ, Carson WE. Giant breast tumors: surgical management of phyllodes tumors, potential for reconstructive surgery and a review of literature. World J Surg Oncol. 2008 Nov 11;6:117. [CrossRef]
- Risk factors for recurrence and death after primary surgical treatment of malignant phyllodes tumors - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/15525831/.
- Neron M, Sajous C, Thezenas S, Piperno-Neumann S, Reyal F, Laé M, Chakiba C, Michot A, Penel N, Honoré C, Owen C, Bertucci F, Salas S, Saada-Bouzid E, Valentin T, Bompas E, Brahmi M, Ray-Coquard I, Blay JY, Firmin N, French Sarcoma Group (GSF-GETO). Surgical Margins and Adjuvant Therapies in Malignant Phyllodes Tumors of the Breast: A Multicenter Retrospective Study. Ann Surg Oncol. 2020 Jun;27(6):1818–27. [CrossRef]
- Ogunbiyi S, Perry A, Jakate K, Simpson J, George R. Phyllodes tumour of the breast and margins: How much is enough. Can J Surg J Can Chir. 2019 Feb 1;62(1):E19–21. [CrossRef]
- Outcome and predictive factors of local recurrence and distant metastases following primary surgical treatment of high-grade malignant phyllodes tumours of the breast - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/11735168/.
- Singh G, Sharma RK. Immediate breast reconstruction for phyllodes tumors. Breast Edinb Scotl. 2008 Jun;17(3):296–301. [CrossRef]
- Yom CK, Han W, Kim SW, Park SY, Park IA, Noh DY. Reappraisal of conventional risk stratification for local recurrence based on clinical outcomes in 285 resected phyllodes tumors of the breast. Ann Surg Oncol. 2015 Sep;22(9):2912–8. [CrossRef]
- Bhargav PRK, Mishra A, Agarwal G, Agarwal A, Verma AK, Mishra SK. Phyllodes tumour of the breast: clinicopathological analysis of recurrent vs. non-recurrent cases. Asian J Surg. 2009 Oct;32(4):224–8. [CrossRef]
- Cheng SP, Chang YC, Liu TP, Lee JJ, Tzen CY, Liu CL. Phyllodes tumor of the breast: the challenge persists. World J Surg. 2006 Aug;30(8):1414–21. [CrossRef]
- Lim RS, Cordeiro E, Lau J, Lim A, Roberts A, Seely J. Phyllodes Tumors-The Predictors and Detection of Recurrence. Can Assoc Radiol J J Assoc Can Radiol. 2021 May;72(2):251–7. [CrossRef]
- Wen B, Mousadoust D, Warburton R, Pao JS, Dingee C, Chen L, McKevitt E. Phyllodes tumours of the breast: Outcomes and recurrence after excision. Am J Surg. 2020 May;219(5):790–4. [CrossRef]
- Taira N, Takabatake D, Aogi K, Ohsumi S, Takashima S, Nishimura R, Teramoto N. Phyllodes tumor of the breast: stromal overgrowth and histological classification are useful prognosis-predictive factors for local recurrence in patients with a positive surgical margin. Jpn J Clin Oncol. 2007 Oct;37(10):730–6. [CrossRef]
- Shin E, Koo JS. Prognostic factors of breast phyllodes tumors. Histol Histopathol. 2023 Aug;38(8):865–78. [CrossRef]
- Thind A, Patel B, Thind K, Isherwood J, Phillips B, Dhaliwal K, Remoundos DD. Surgical margins for borderline and malignant phyllodes tumours. Ann R Coll Surg Engl. 2020 Mar;102(3):165–73. [CrossRef]
- Chen WH, Cheng SP, Tzen CY, Yang TL, Jeng KS, Liu CL, Liu TP. Surgical treatment of phyllodes tumors of the breast: retrospective review of 172 cases. J Surg Oncol. 2005 Sep 1;91(3):185–94. [CrossRef]
- Zhang Y, Kleer CG. Phyllodes Tumor of the Breast: Histopathologic Features, Differential Diagnosis, and Molecular/Genetic Updates. Arch Pathol Lab Med. 2016 Jul;140(7):665–71. [CrossRef]
- Mitus JW, Blecharz P, Jakubowicz J, Reinfuss M, Walasek T, Wysocki W. Phyllodes tumors of the breast. The treatment results for 340 patients from a single cancer centre. Breast Edinb Scotl. 2019 Feb;43:85–90. [CrossRef]
- Rowell MD, Perry RR, Hsiu JG, Barranco SC. Phyllodes tumors. Am J Surg. 1993 Mar;165(3):376–9.
- Casali PG, Abecassis N, Aro HT, Bauer S, Biagini R, Bielack S, Bonvalot S, Boukovinas I, Bovee JVMG, Brodowicz T, Broto JM, Buonadonna A, De Álava E, Dei Tos AP, Del Muro XG, Dileo P, Eriksson M, Fedenko A, Ferraresi V, Ferrari A, Ferrari S, Frezza AM, Gasperoni S, Gelderblom H, Gil T, Grignani G, Gronchi A, Haas RL, Hassan B, Hohenberger P, Issels R, Joensuu H, Jones RL, Judson I, Jutte P, Kaal S, Kasper B, Kopeckova K, Krákorová DA, Le Cesne A, Lugowska I, Merimsky O, Montemurro M, Pantaleo MA, Piana R, Picci P, Piperno-Neumann S, Pousa AL, Reichardt P, Robinson MH, Rutkowski P, Safwat AA, Schöffski P, Sleijfer S, Stacchiotti S, Sundby Hall K, Unk M, Van Coevorden F, van der Graaf WTA, Whelan J, Wardelmann E, Zaikova O, Blay JY, ESMO Guidelines Committee and EURACAN. Soft tissue and visceral sarcomas: ESMO-EURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol Off J Eur Soc Med Oncol. 2018 Oct 1;29(Suppl 4):iv51–67.
- Chao X, Chen K, Zeng J, Bi Z, Guo M, Chen Y, Yao Y, Wu W, Liang S, Nie Y. Adjuvant radiotherapy and chemotherapy for patients with breast phyllodes tumors: a systematic review and meta-analysis. BMC Cancer. 2019 Apr 23;19(1):372. [CrossRef]
- Marazzi F, Tagliaferri L, Masiello V, Moschella F, Colloca GF, Corvari B, Sanchez AM, Capocchiano ND, Pastorino R, Iacomini C, Lenkowicz J, Masciocchi C, Patarnello S, Franceschini G, Gambacorta MA, Masetti R, Valentini V. GENERATOR Breast DataMart-The Novel Breast Cancer Data Discovery System for Research and Monitoring: Preliminary Results and Future Perspectives. J Pers Med. 2021 Jan 22;11(2):65.
- Yang P, Zhang G, Zhang Y, Zhao W, Tang J, Zeng S, Lv X, Lv L. Effect of adjuvant radiotherapy on overall survival and breast cancer-specific survival of patients with malignant phyllodes tumor of the breast in different age groups: a retrospective observational study based on SEER. Radiat Oncol Lond Engl. 2024 May 21;19(1):59. [CrossRef]
- Moon SH, Jung JH, Lee J, Kim WW, Park HY, Lee JW, Lee SJ. Complete remission of giant malignant phyllodes tumor with lung metastasis: A case report. Medicine (Baltimore). 2019 May;98(22):e15762.
- Li GZ, Raut CP, Hunt KK, Feng M, Chugh R. Breast Sarcomas, Phyllodes Tumors, and Desmoid Tumors: Epidemiology, Diagnosis, Staging, and Histology-Specific Management Considerations. Am Soc Clin Oncol Educ Book Am Soc Clin Oncol Annu Meet. 2021 Mar;41:390–404.
- Apatinib treatment is effective for metastatic malignant phyllodes tumors of the breast: a case report - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/34022875/.
- Orlandi A, Sanchez AM, Calegari MA, D’Archi S, Santoro A, Leone AD, Mulè A, Arena V, Masetti R, Barone C, Franceschini G. Diagnosis and management of breast lymphoma: a single- institution retrospective analysis. Transl Cancer Res [Internet]. 2018 Apr [cited 2024 Mar 29];7(Suppl 3). Available from: https://tcr.amegroups.org/article/view/17996.
- Nizri E, Merimsky O, Lahat G. Optimal management of sarcomas of the breast: an update. Expert Rev Anticancer Ther. 2014 Jun;14(6):705–10. [CrossRef]
- Zelek L, Llombart-Cussac A, Terrier P, Pivot X, Guinebretiere JM, Le Pechoux C, Tursz T, Rochard F, Spielmann M, Le Cesne A. Prognostic factors in primary breast sarcomas: a series of patients with long-term follow-up. J Clin Oncol Off J Am Soc Clin Oncol. 2003 Jul 1;21(13):2583–8. [CrossRef]
- Pasta V, Monti M, Cialini M, Vergine M, Urciuoli P, Iacovelli A, Rea S, D’Orazi V. Primitive sarcoma of the breast: new insight on the proper surgical management. J Exp Clin Cancer Res CR. 2015 Jul 30;34(1):72. [CrossRef]
- Shabahang M, Franceschi D, Sundaram M, Castillo MH, Moffat FL, Frank DS, Rosenberg ER, Bullock KE, Livingstone AS. Surgical management of primary breast sarcoma. Am Surg. 2002 Aug;68(8):673–7; discussion 677. [CrossRef]
- Lahat G, Lev D, Gerstenhaber F, Madewell J, Le-Petross H, Pollock RE. Sarcomas of the breast. Expert Rev Anticancer Ther. 2012 Aug;12(8):1045–51.
- Primary breast sarcoma: clinicopathologic series from the Mayo Clinic and review of the literature - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/15187996/.
- Seinen JM, Styring E, Verstappen V, Vult von Steyern F, Rydholm A, Suurmeijer AJH, Hoekstra HJ. Radiation-associated angiosarcoma after breast cancer: high recurrence rate and poor survival despite surgical treatment with R0 resection. Ann Surg Oncol. 2012 Aug;19(8):2700–6. [CrossRef]
- Vorburger SA, Xing Y, Hunt KK, Lakin GE, Benjamin RS, Feig BW, Pisters PWT, Ballo MT, Chen L, Trent J, Burgess M, Patel S, Pollock RE, Cormier JN. Angiosarcoma of the breast. Cancer. 2005 Dec 15;104(12):2682–8.
- Angiosarcoma (Stewart-Treves syndrome) in postmastectomy patients: report of 10 cases and review of literature - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/26617830/.
- An analysis of 78 breast sarcoma patients without distant metastases at presentation - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/10661345/.
- Pandey M, Mathew A, Abraham EK, Rajan B. Primary sarcoma of the breast. J Surg Oncol. 2004 Sep 1;87(3):121–5. [CrossRef]
- WHO Classification of tumors, 5th Edition. 5th ed. 2020.
- Lee JS, Yoon K, Onyshchenko M. Sarcoma of the Breast: Clinical Characteristics and Outcomes of 991 Patients from the National Cancer Database. Sarcoma. 2021;2021:8828158. [CrossRef]
- Deyrup AT, Weiss SW. Grading of soft tissue sarcomas: the challenge of providing precise information in an imprecise world. Histopathology. 2006 Jan;48(1):42–50. [CrossRef]
- Machado I, Giner F, Lavernia J, Cruz J, Traves V, Requena C, Llombart B, López-Guerrero JA, Llombart-Bosch A. Angiosarcomas: histology, immunohistochemistry and molecular insights with implications for differential diagnosis. Histol Histopathol. 2021 Jan;36(1):3–18. [CrossRef]
- Kuba MG, Xu B, D’Angelo SP, Rosenbaum E, Plitas G, Ross DS, Brogi E, Antonescu CR. The impact of MYC gene amplification on the clinicopathological features and prognosis of radiation-associated angiosarcomas of the breast. Histopathology. 2021 Nov;79(5):836–46. [CrossRef]
- Mentzel T, Kiss K. Reduced H3K27me3 expression in radiation-associated angiosarcoma of the breast. Virchows Arch Int J Pathol. 2018 Mar;472(3):361–8. [CrossRef]
- Dahlstrom J, Buckingham J, Bell S, Jain S. Nodular fasciitis of the breast simulating breast cancer on imaging. Australas Radiol. 2001 Feb;45(1):67–70. [CrossRef]
- Erickson-Johnson MR, Chou MM, Evers BR, Roth CW, Seys AR, Jin L, Ye Y, Lau AW, Wang X, Oliveira AM. Nodular fasciitis: a novel model of transient neoplasia induced by MYH9-USP6 gene fusion. Lab Investig J Tech Methods Pathol. 2011 Oct;91(10):1427–33. [CrossRef]
- Reactive spindle cell nodules of the breast after core biopsy or fine-needle aspiration - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/10664632/.
- Radu I, Scripcariu V, Panuța A, Rusu A, Afrăsânie VA, Cojocaru E, Aniței MG, Alexa-Stratulat T, Terinte C, Șerban CF, Gafton B. Breast Sarcomas-How Different Are They from Breast Carcinomas? Clinical, Pathological, Imaging and Treatment Insights. Diagn Basel Switz. 2023 Apr 7;13(8):1370.
- Al-Benna S, Poggemann K, Steinau HU, Steinstraesser L. Diagnosis and management of primary breast sarcoma. Breast Cancer Res Treat. 2010 Aug;122(3):619–26. [CrossRef]
- Lim SZ, Ong KW, Tan BKT, Selvarajan S, Tan PH. Sarcoma of the breast: an update on a rare entity. J Clin Pathol. 2016 May;69(5):373–81. [CrossRef]
- Ramelli E, Weingertner N, Welsch A, Reix N, Antoni D, Amé S, Molière S, Mathelin C. Innovative approach to lymphadenectomy in breast sarcoma. Bull Cancer (Paris). 2022 Oct;109(10):1017–28. [CrossRef]
- Pencavel TD, Hayes A. Breast sarcoma--a review of diagnosis and management. Int J Surg Lond Engl. 2009 Feb;7(1):20–3. [CrossRef]
- Confavreux C, Lurkin A, Mitton N, Blondet R, Saba C, Ranchère D, Sunyach MP, Thiesse P, Biron P, Blay JY, Ray-Coquard I. Sarcomas and malignant phyllodes tumours of the breast--a retrospective study. Eur J Cancer Oxf Engl 1990. 2006 Nov;42(16):2715–21. [CrossRef]
- Fields RC, Aft RL, Gillanders WE, Eberlein TJ, Margenthaler JA. Treatment and outcomes of patients with primary breast sarcoma. Am J Surg. 2008 Oct;196(4):559–61. [CrossRef]
- Wang F, Jia Y, Tong Z. Comparison of the clinical and prognostic features of primary breast sarcomas and malignant phyllodes tumor. Jpn J Clin Oncol. 2015 Feb;45(2):146–52. [CrossRef]
- Berg JW, Decrosse JJ, Fracchia AA, Farrow J. Stromal sarcomas of the breast. A unified approach to connective tissue sarcomas other than cystosarcoma phyllodes. Cancer. 1962;15:418–24.
- Bousquet G, Confavreux C, Magné N, de Lara CT, Poortmans P, Senkus E, de Lafontan B, Bolla M, Largillier R, Lagneau E, Kadish S, Lemanski C, Ozsahin M, Belkacémi Y. Outcome and prognostic factors in breast sarcoma: a multicenter study from the rare cancer network. Radiother Oncol J Eur Soc Ther Radiol Oncol. 2007 Dec;85(3):355–61. [CrossRef]
- Ciatto S, Bonardi R, Cataliotti L, Cardona G. Sarcomas of the breast: a multicenter series of 70 cases. Neoplasma. 1992;39(6):375–9.
- North JH, McPhee M, Arredondo M, Edge SB. Sarcoma of the breast: implications of the extent of local therapy. Am Surg. 1998 Nov;64(11):1059–61.
- Crosby MA, Chike-Obi CJ, Baumann DP, Sacks JM, Villa MT, Garvey PB, Selber JC, Feig BW. Reconstructive outcomes in patients with sarcoma of the breast. Plast Reconstr Surg. 2010 Dec;126(6):1805–14. [CrossRef]
- Innovative approach to lymphadenectomy in breast sarcoma - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/35717223/.
- Fong Y, Coit DG, Woodruff JM, Brennan MF. Lymph node metastasis from soft tissue sarcoma in adults. Analysis of data from a prospective database of 1772 sarcoma patients. Ann Surg. 1993 Jan;217(1):72–7. [CrossRef]
- Gullett NP, Delman K, Folpe AL, Johnstone PAS. National surgical patterns of care: regional lymphadenectomy of breast sarcomas. Am J Clin Oncol. 2007 Oct;30(5):461–5.
- Kuerer HM. Kuerer’s Breast Surgical Oncology. In: Kuerer’s Breast Surgical Oncology [Internet]. New York, NY: The McGraw-Hill Companies; 2010 [cited 2025 Jan 14]. Available from: accesssurgery.mhmedical.com/content.aspx?aid=1000039191.
- Voutsadakis IA, Zaman K, Leyvraz S. Breast sarcomas: current and future perspectives. Breast Edinb Scotl. 2011 Jun;20(3):199–204. [CrossRef]
- Gutman H, Pollock RE, Ross MI, Benjamin RS, Johnston DA, Janjan NA, Romsdahl MM. Sarcoma of the breast: implications for extent of therapy. The M. D. Anderson experience. Surgery. 1994 Sep;116(3):505–9.
- Kokkali S, Stravodimou A, Duran-Moreno J, Koufopoulos N, Voutsadakis IA, Digklia A. Chemotherapy and targeted treatments of breast sarcoma by histologic subtype. Expert Rev Anticancer Ther. 2021 Jun;21(6):591–604. [CrossRef]
- Bramwell V, Rouesse J, Steward W, Santoro A, Schraffordt-Koops H, Buesa J, Ruka W, Priario J, Wagener T, Burgers M. Adjuvant CYVADIC chemotherapy for adult soft tissue sarcoma--reduced local recurrence but no improvement in survival: a study of the European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group. J Clin Oncol Off J Am Soc Clin Oncol. 1994 Jun;12(6):1137–49. [CrossRef]
- Le Cesne A, Ouali M, Leahy MG, Santoro A, Hoekstra HJ, Hohenberger P, Van Coevorden F, Rutkowski P, Van Hoesel R, Verweij J, Bonvalot S, Steward WP, Gronchi A, Hogendoorn PCW, Litiere S, Marreaud S, Blay JY, Van Der Graaf WTA. Doxorubicin-based adjuvant chemotherapy in soft tissue sarcoma: pooled analysis of two STBSG-EORTC phase III clinical trials. Ann Oncol Off J Eur Soc Med Oncol. 2014 Dec;25(12):2425–32.
- von Mehren M, Randall RL, Benjamin RS, Boles S, Bui MM, Ganjoo KN, George S, Gonzalez RJ, Heslin MJ, Kane JM, Keedy V, Kim E, Koon H, Mayerson J, McCarter M, McGarry SV, Meyer C, Morris ZS, O’Donnell RJ, Pappo AS, Paz IB, Petersen IA, Pfeifer JD, Riedel RF, Ruo B, Schuetze S, Tap WD, Wayne JD, Bergman MA, Scavone JL. Soft Tissue Sarcoma, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw JNCCN. 2018 May;16(5):536–63.
- Gronchi A, Palmerini E, Quagliuolo V, Martin Broto J, Lopez Pousa A, Grignani G, Brunello A, Blay JY, Tendero O, Diaz Beveridge R, Ferraresi V, Lugowska I, Merlo DF, Fontana V, Marchesi E, Braglia L, Donati DM, Palassini E, Bianchi G, Marrari A, Morosi C, Stacchiotti S, Bagué S, Coindre JM, Dei Tos AP, Picci P, Bruzzi P, Casali PG. Neoadjuvant Chemotherapy in High-Risk Soft Tissue Sarcomas: Final Results of a Randomized Trial From Italian (ISG), Spanish (GEIS), French (FSG), and Polish (PSG) Sarcoma Groups. J Clin Oncol Off J Am Soc Clin Oncol. 2020 Jul 1;38(19):2178–86. [CrossRef]
- Judson I, Verweij J, Gelderblom H, Hartmann JT, Schöffski P, Blay JY, Kerst JM, Sufliarsky J, Whelan J, Hohenberger P, Krarup-Hansen A, Alcindor T, Marreaud S, Litière S, Hermans C, Fisher C, Hogendoorn PCW, dei Tos AP, van der Graaf WTA, European Organisation and Treatment of Cancer Soft Tissue and Bone Sarcoma Group. 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. 2014 Apr;15(4):415–23. [CrossRef]
- D’Ambrosio L, Touati N, Blay JY, Grignani G, Flippot R, Czarnecka AM, Piperno-Neumann S, Martin-Broto J, Sanfilippo R, Katz D, Duffaud F, Vincenzi B, Stark DP, Mazzeo F, Tuchscherer A, Chevreau C, Sherriff J, Estival A, Litière S, Sents W, Ray-Coquard I, Tolomeo F, Le Cesne A, Rutkowski P, Stacchiotti S, Kasper B, Gelderblom H, Gronchi A, European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group. 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. 2020 Jun 1;126(11):2637–47.
- Pautier P, Italiano A, Piperno-Neumann S, Chevreau C, Penel N, Firmin N, Boudou-Rouquette P, Bertucci F, Balleyguier C, Lebrun-Ly V, Ray-Coquard I, Kalbacher E, Bardet A, Bompas E, Collard O, Isambert N, Guillemet C, Rios M, Archambaud B, Duffaud F, French Sarcoma Group. Doxorubicin alone versus doxorubicin with trabectedin followed by trabectedin alone as first-line therapy for metastatic or unresectable leiomyosarcoma (LMS-04): a randomised, multicentre, open-label phase 3 trial. Lancet Oncol. 2022 Aug;23(8):1044–54. [CrossRef]
- Liu W, Jiang Q, Zhou Y. Advances of systemic treatment for adult soft-tissue sarcoma. Chin Clin Oncol. 2018 Aug;7(4):42. [CrossRef]
- Chawla SP, Papai Z, Mukhametshina G, Sankhala K, Vasylyev L, Fedenko A, Khamly K, Ganjoo K, Nagarkar R, Wieland S, Levitt DJ. First-Line Aldoxorubicin vs Doxorubicin in Metastatic or Locally Advanced Unresectable Soft-Tissue Sarcoma: A Phase 2b Randomized Clinical Trial. JAMA Oncol. 2015 Dec;1(9):1272–80.
- Chawla S, Ganjoo K, Schuetze S, Papai Z, Tine B, Choy E, Liebner D, Agulnik M, Chawla S, Wieland S, Levitt D. Phase III study of aldoxorubicin vs investigators’ choice as treatment for relapsed/refractory soft tissue sarcomas. J Clin Oncol. 2017 May 20;35:11000–11000. [CrossRef]
- Gupta S, Gouw L, Wright J, Chawla S, Pitt D, Wade M, Boucher K, Sharma S. Phase II study of amrubicin (SM-5887), a synthetic 9-aminoanthracycline, as first line treatment in patients with metastatic or unresectable soft tissue sarcoma: durable response in myxoid liposarcoma with TLS-CHOP translocation. Invest New Drugs. 2016 Apr;34(2):243–52. [CrossRef]
- Tap WD, Papai Z, Van Tine BA, Attia S, Ganjoo KN, Jones RL, Schuetze S, Reed D, Chawla SP, Riedel RF, Krarup-Hansen A, Toulmonde M, Ray-Coquard I, Hohenberger P, Grignani G, Cranmer LD, Okuno S, Agulnik M, Read W, Ryan CW, Alcindor T, Del Muro XFG, Budd GT, Tawbi H, Pearce T, Kroll S, Reinke DK, Schöffski P. Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (TH CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2017 Aug;18(8):1089–103.
- Cianfrocca M, Lee S, Von Roenn J, Tulpule A, Dezube BJ, Aboulafia DM, Ambinder RF, Lee JY, Krown SE, Sparano JA. Randomized trial of paclitaxel versus pegylated liposomal doxorubicin for advanced human immunodeficiency virus-associated Kaposi sarcoma: evidence of symptom palliation from chemotherapy. Cancer. 2010 Aug 15;116(16):3969–77.
- Maki RG, Wathen JK, Patel SR, Priebat DA, Okuno SH, Samuels B, Fanucchi M, Harmon DC, Schuetze SM, Reinke D, Thall PF, Benjamin RS, Baker LH, Hensley ML. Randomized phase II study of gemcitabine and docetaxel compared with gemcitabine alone in patients with metastatic soft tissue sarcomas: results of sarcoma alliance for research through collaboration study 002 [corrected]. J Clin Oncol Off J Am Soc Clin Oncol. 2007 Jul 1;25(19):2755–63.
- Seddon B, Strauss SJ, Whelan J, Leahy M, Woll PJ, Cowie F, Rothermundt C, Wood Z, Benson C, Ali N, Marples M, Veal GJ, Jamieson D, Küver K, Tirabosco R, Forsyth S, Nash S, Dehbi HM, Beare S. 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. 2017 Oct;18(10):1397–410. [CrossRef]
- Penel N, Bui BN, Bay JO, Cupissol D, Ray-Coquard I, Piperno-Neumann S, Kerbrat P, Fournier C, Taieb S, Jimenez M, Isambert N, Peyrade F, Chevreau C, Bompas E, Brain EGC, Blay JY. Phase II trial of weekly paclitaxel for unresectable angiosarcoma: the ANGIOTAX Study. J Clin Oncol Off J Am Soc Clin Oncol. 2008 Nov 10;26(32):5269–74. [CrossRef]
- Borden EC, Amato DA, Rosenbaum C, Enterline HT, Shiraki MJ, Creech RH, Lerner HJ, Carbone PP. Randomized comparison of three adriamycin regimens for metastatic soft tissue sarcomas. J Clin Oncol Off J Am Soc Clin Oncol. 1987 Jun;5(6):840–50. [CrossRef]
- Buesa JM, Mouridsen HT, van Oosterom AT, Verweij J, Wagener T, Steward W, Poveda A, Vestlev PM, Thomas D, Sylvester R. High-dose DTIC in advanced soft-tissue sarcomas in the adult. A phase II study of the E.O.R.T.C. Soft Tissue and Bone Sarcoma Group. Ann Oncol Off J Eur Soc Med Oncol. 1991 Apr;2(4):307–9. [CrossRef]
- García-Del-Muro X, López-Pousa A, Maurel J, Martín J, Martínez-Trufero J, Casado A, Gómez-España A, Fra J, Cruz J, Poveda A, Meana A, Pericay C, Cubedo R, Rubió J, De Juan A, Laínez N, Carrasco JA, de Andrés R, Buesa JM, Spanish Group for Research on Sarcomas. 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 Off J Am Soc Clin Oncol. 2011 Jun 20;29(18):2528–33.
- Gurruchaga Sotés I, Gómez-Mateo MC, Ortega Izquierdo ME, Martínez-Trufero J. Beneficial Use of the Combination of Gemcitabine and Dacarbazine in Advanced Soft Tissue Sarcomas: Real-World Data. Cancers. 2024 Jan 8;16(2):267.
- Delaloge S, Yovine A, Taamma A, Riofrio M, Brain E, Raymond E, Cottu P, Goldwasser F, Jimeno J, Misset JL, Marty M, Cvitkovic E. Ecteinascidin-743: a marine-derived compound in advanced, pretreated sarcoma patients--preliminary evidence of activity. J Clin Oncol Off J Am Soc Clin Oncol. 2001 Mar 1;19(5):1248–55.
- Demetri GD, von Mehren M, Jones RL, Hensley ML, Schuetze SM, Staddon A, Milhem M, Elias A, Ganjoo K, Tawbi H, Van Tine BA, Spira A, Dean A, Khokhar NZ, Park YC, Knoblauch RE, Parekh TV, Maki RG, Patel SR. Efficacy and Safety of Trabectedin or Dacarbazine for Metastatic Liposarcoma or Leiomyosarcoma After Failure of Conventional Chemotherapy: Results of a Phase III Randomized Multicenter Clinical Trial. J Clin Oncol Off J Am Soc Clin Oncol. 2016 Mar 10;34(8):786–93.
- Le Cesne A, Blay JY, Cupissol D, Italiano A, Delcambre C, Penel N, Isambert N, Chevreau C, Bompas E, Bertucci F, Chaigneau L, Piperno-Neumann S, Salas S, Rios M, Guillemet C, Bay JO, Ray-Coquard I, Haddag L, Bonastre J, Kapso R, Fraslin A, Bouvet N, Mir O, Foulon S. A randomized phase III trial comparing trabectedin to best supportive care in patients with pre-treated soft tissue sarcoma: T-SAR, a French Sarcoma Group trial. Ann Oncol Off J Eur Soc Med Oncol. 2021 Aug;32(8):1034–44. [CrossRef]
- Garcia del Muro X, Lopez-Pousa A, Martin J, Buesa JM, Martinez-Trufero J, Casado A, Poveda A, Cruz J, Bover I, Maurel J, Spanish Group for Research on Sarcomas. A phase II trial of temozolomide as a 6-week, continuous, oral schedule in patients with advanced soft tissue sarcoma: a study by the Spanish Group for Research on Sarcomas. Cancer. 2005 Oct 15;104(8):1706–12.
- Talbot SM, Keohan ML, Hesdorffer M, Orrico R, Bagiella E, Troxel AB, Taub RN. A phase II trial of temozolomide in patients with unresectable or metastatic soft tissue sarcoma. Cancer. 2003 Nov 1;98(9):1942–6. [CrossRef]
- van der Graaf WTA, Blay JY, Chawla SP, Kim DW, Bui-Nguyen B, Casali PG, Schöffski P, Aglietta M, Staddon AP, Beppu Y, Le Cesne A, Gelderblom H, Judson IR, Araki N, Ouali M, Marreaud S, Hodge R, Dewji MR, Coens C, Demetri GD, Fletcher CD, Dei Tos AP, Hohenberger P, EORTC Soft Tissue and Bone Sarcoma Group, PALETTE study group. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Lond Engl. 2012 May 19;379(9829):1879–86. [CrossRef]
- Sleijfer S, Ray-Coquard I, Papai Z, Le Cesne A, Scurr M, Schöffski P, Collin F, Pandite L, Marreaud S, De Brauwer A, van Glabbeke M, Verweij J, Blay JY. Pazopanib, a multikinase angiogenesis inhibitor, in patients with relapsed or refractory advanced soft tissue sarcoma: a phase II study from the European organisation for research and treatment of cancer-soft tissue and bone sarcoma group (EORTC study 62043). J Clin Oncol Off J Am Soc Clin Oncol. 2009 Jul 1;27(19):3126–32.
- Agulnik M, Costa RLB, Milhem M, Rademaker AW, Prunder BC, Daniels D, Rhodes BT, Humphreys C, Abbinanti S, Nye L, Cehic R, Polish A, Vintilescu C, McFarland T, Skubitz K, Robinson S, Okuno S, Van Tine BA. A phase II study of tivozanib in patients with metastatic and nonresectable soft-tissue sarcomas. Ann Oncol Off J Eur Soc Med Oncol. 2017 Jan 1;28(1):121–7.
- Chi Y, Yao Y, Wang S, Huang G, Cai Q, Shang G, Wang G, Qu G, Wu Q, Jiang Y, Song J, Chen J, Zhu X, Cai Z, Bai C, Lu Y, Yu Z, Shen J, Cai J. Anlotinib for metastasis soft tissue sarcoma: A randomized, double-blind, placebo-controlled and multi-centered clinical trial. J Clin Oncol. 2018 May 20;36(15_suppl):11503–11503.
- Yao W, Du X, Wang J, Wang X, Zhang P, Niu X. Long-Term Efficacy and Safety of Anlotinib as a Monotherapy and Combined Therapy for Advanced Sarcoma. OncoTargets Ther. 2022;15:669–79. [CrossRef]
- Xu B, Pan Q, Pan H, Li H, Li X, Chen J, Pang D, Zhang B, Weng D, Peng R, Fang M, Zhang X. Anlotinib as a maintenance treatment for advanced soft tissue sarcoma after first-line chemotherapy (ALTER-S006): a multicentre, open-label, single-arm, phase 2 trial. EClinicalMedicine. 2023 Oct;64:102240. [CrossRef]
- Li T, Ye Z, Wei Y, Wang S, Liu Y, Chen J. A phase II study of anlotinib in the first-line treatment of locally advanced or metastatic soft-tissue sarcoma: Updated results. J Clin Oncol. 2022 Jun 1;40(16_suppl):e23559–e23559.
- Berry V, Basson L, Bogart E, Mir O, Blay JY, Italiano A, Bertucci F, Chevreau C, Clisant-Delaine S, Liegl-Antzager B, Tresch-Bruneel E, Wallet J, Taieb S, Decoupigny E, Le Cesne A, Brodowicz T, Penel N. REGOSARC: Regorafenib versus placebo in doxorubicin-refractory soft-tissue sarcoma-A quality-adjusted time without symptoms of progression or toxicity analysis. Cancer. 2017 Jun 15;123(12):2294–302. [CrossRef]
- Attia S, Villalobos V, Hindi N, Wagner AJ, Chmielowski B, Oakley GJ, Peterson PM, Ceccarelli M, Jones RL, Dickson MA. Randomized Phase 2 Clinical Trial of Olaratumab in Combination with Gemcitabine and Docetaxel in Advanced Soft Tissue Sarcomas. Cancers. 2023 Oct 6;15(19):4871. [CrossRef]
- Tian Z, Yao W. PD-1/L1 inhibitor plus chemotherapy in the treatment of sarcomas. Front Immunol. 2022;13:898255.
- Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial - PubMed [Internet]. [cited 2025 Jan 13]. Available from: https://pubmed.ncbi.nlm.nih.gov/28988646/.
- Burgess MA, Bolejack V, Schuetze S, Van Tine BA, Attia S, Riedel RF, Hu JS, Davis LE, Okuno SH, Priebat DA, Movva S, Reed DR, D’Angelo SP, Lazar AJ, Keung EZY, Reinke DK, Baker LH, Maki RG, Patel S, Tawbi HAH. Clinical activity of pembrolizumab (P) in undifferentiated pleomorphic sarcoma (UPS) and dedifferentiated/pleomorphic liposarcoma (LPS): Final results of SARC028 expansion cohorts. J Clin Oncol. 2019 May 20;37(15_suppl):11015–11015.
- Italiano A, Bellera C, D’Angelo S. PD1/PD-L1 targeting in advanced soft-tissue sarcomas: a pooled analysis of phase II trials. J Hematol OncolJ Hematol Oncol. 2020 May 19;13(1):55.
- D’Angelo SP, Mahoney MR, Van Tine BA, Atkins J, Milhem MM, Jahagirdar BN, Antonescu CR, Horvath E, Tap WD, Schwartz GK, Streicher H. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol. 2018 Mar;19(3):416–26.
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).